1
|
Girase R, Gujarathi NA, Sukhia A, Kota SSN, Patil TS, Aher AA, Agrawal YO, Ojha S, Sharma C, Goyal SN. Targeted nanoliposomes for precision rheumatoid arthritis therapy: a review on mechanisms and in vivo potential. Drug Deliv 2025; 32:2459772. [PMID: 39891600 PMCID: PMC11789225 DOI: 10.1080/10717544.2025.2459772] [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: 10/04/2024] [Revised: 12/26/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory immune-triggered disease that causes synovitis, cartilage degradation, and joint injury. In nanotechnology, conventional liposomes were extensively investigated for RA. However, they frequently undergo rapid clearance, reducing circulation time and therapeutic efficacy. Additionally, their stability in the bloodstream is often compromised, resulting in premature drug release. The current review explores the potential of targeted liposomal-based nanosystems in the treatment of RA. It highlights the pathophysiology of RA, explores selective targeting sites, and elucidates diverse mechanisms of novel liposomal types and their applications. Furthermore, the targeting strategies of pH-sensitive, flexible, surface-modified, PEGylated, acoustic, ROS-mediated, and biofunctionalized liposomes are addressed. Targeted nanoliposomes showed potential in precisely delivering drugs to CD44, SR-A, FR-β, FLS, and toll-like receptors through the high affinity of ligands. In vitro studies interpreted stable release profiles and improved stability. Ex vivo studies on skin demonstrated that ultradeformable and glycerol-conjugated liposomes enhanced drug penetrability. In vivo experiments for liposomal types in the arthritis rat model depicted remarkable efficacy in reducing joint swelling, pro-inflammatory cytokines, and synovial hyperplasia. In conclusion, these targeted liposomes represented a significant leap forward in drug delivery, offering effective therapeutic options for RA. In the future, integrating these advanced liposomes with artificial intelligence, immunotherapy, and precision medicine holds great promise.
Collapse
Affiliation(s)
- Rushikesh Girase
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, India
| | | | - Amey Sukhia
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sri Sai Nikitha Kota
- Department of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, USA
| | | | - Abhijeet A. Aher
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, India
| | | | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Sameer N. Goyal
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, India
| |
Collapse
|
2
|
Li H, Jin X, Chu B, Zhang K, Qin X, Pan S, Zhao Y, Shi H, Zhang J, Wang H, Wen Z, He Y, Sun X. Inflammation Targeting and Responsive Multifunctional Drug-Delivery Nanoplatforms for Combined Therapy of Rheumatoid Arthritis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2500113. [PMID: 40277325 DOI: 10.1002/smll.202500113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/29/2025] [Indexed: 04/26/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by persistent inflammation, joint swelling, pain, and progressive joint destruction. Methotrexate (MTX) is the standard first-line treatment for RA, but its clinical application is hindered by poor water solubility and non-specific delivery. In this work, a multifunctional drug-delivery nanoplatform that targets both macrophages and tumor necrosis factor α (TNFα) is developed to enhance the therapeutic efficacy of MTX in RA. The nanoplatform consists of folic acid (FA, for macrophage targeting) and a TNFα-specific Aptamer (TNFα-Apt), facilitating a dual-targeting strategy that significantly improves the accumulation of MTX at the sites of RA lesions (≈3.5-fold). Moreover, the manganese dioxide (MnO₂) and polydopamine (PDA) coatings on the nanoplatform effectively scavenge reactive oxygen species (ROS), generate oxygen, and promote the polarization of pro-inflammatory M1 macrophages to the anti-inflammatory M2 macrophages. This shift in macrophage polarization restores the expression of key inflammatory cytokines, improving the local inflammatory microenvironment. Ultimately, the nanoplatform significantly ameliorates the inflammation and joint damage in a collagen-induced arthritis (CIA) model, suggesting that this multi-target combination therapy holds considerable potential for the treatment of RA in vivo.
Collapse
Affiliation(s)
- Hongyang Li
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Xiangbowen Jin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Binbin Chu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Kai Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Xuan Qin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Sheng Pan
- Department of Orthopaedics, Second Affiliated Hospital of Soochow University, Osteoporosis Research Institute of Soochow University, Suzhou, 215000, China
| | - Yadan Zhao
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Haoliang Shi
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Jiawei Zhang
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Houyu Wang
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
| | - Zhen Wen
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yao He
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Soochow University, Suzhou, 215123, China
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| | - Xuhui Sun
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Institute of Functional Nano and Soft Materials (FUNSOM) and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| |
Collapse
|
3
|
Abou Hjeily B, Nevaneeth BC, Samborski W, Szekanecz Z, Grygiel-Górniak B. Inflammatory Pathways to Carcinogenesis: Deciphering the Rheumatoid Arthritis-Lung Cancer Connection. Cancers (Basel) 2025; 17:1330. [PMID: 40282506 PMCID: PMC12026397 DOI: 10.3390/cancers17081330] [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: 02/26/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Rheumatoid arthritis (RA) is the most common chronic autoimmune arthropathy. If the disease is aggressive or left untreated, it becomes debilitating, affects a patient's functionality, and reduces the quality of life. Disease-modifying anti-rheumatic drugs (DMARDs), both conventional, targeted, and biological, decrease the disease progression and are key components of effective treatment. Recently, there has been a continuous debate about the possible carcinogenicity of various DMARDs. Lung cancer is a leading cause of cancer death worldwide. The available data show an increased risk of lung cancer in RA patients, but the link between RA and cancer is poorly understood. Carcinogenesis in RA seems to be related to chronic inflammation, familial predisposition, risky behaviors (e.g., smoking), and iatrogenic complications. The main mechanisms of carcinogenic processes in patients with RA are the up-regulation of interleukin-6 (IL-6) cytokine production and wingless/integrated WNT signaling. Up-regulation of WNT5A is an important mechanism that links chronic inflammatory pathways to carcinogenesis observed in RA patients. Concomitant up-regulation of transcription factor STAT3 promotes cell proliferation and inhibits apoptosis. Conversely, suppressed inflammatory processes by DMARDs may decrease the risk of lung cancer. In this article, we discuss the molecular mechanisms of lung cancer in RA and the role of DMARDs in this process. Furthermore, we analyze the molecular effect of drug-induced cancer, which affects transcription factors and thus modulates carcinogenic processes. Finally, we describe risk factors and present preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Boushra Abou Hjeily
- Rheumatology Research Group, Department of Rheumatology, Rehabilitation and Internal Diseases, Poznan University of Medical Science, 61-701 Poznan, Poland
| | - Briana Candace Nevaneeth
- Rheumatology Research Group, Department of Rheumatology, Rehabilitation and Internal Diseases, Poznan University of Medical Science, 61-701 Poznan, Poland
| | - Włodzimierz Samborski
- Department of Rheumatology, Rehabilitation and Internal Diseases, Poznan University of Medical Science, 61-701 Poznan, Poland;
| | - Zoltán Szekanecz
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - Bogna Grygiel-Górniak
- Department of Rheumatology, Rehabilitation and Internal Diseases, Poznan University of Medical Science, 61-701 Poznan, Poland;
| |
Collapse
|
4
|
Huang Z, Wu T, Lu R, Zhou H, Zhang Y, Huang L, Gan Y, He H. Prevalence and clinical significance of anti-neutrophil cytoplasmic antibodies in rheumatoid arthritis-associated interstitial lung disease. BMC Pulm Med 2025; 25:177. [PMID: 40229742 PMCID: PMC11995625 DOI: 10.1186/s12890-025-03644-7] [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: 01/22/2025] [Accepted: 04/02/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Patients with rheumatoid arthritis (RA) or rheumatoid arthritis-associated interstitial lung disease (RA-ILD) are commonly positive for antineutrophil cytoplasmic antibodies (ANCAs). The causal relationship between RA-ILD and ANCAs and the role of ANCAs in RA-ILD remain unclear. The purpose of this study was to estimate the prevalence of ANCAs in RA-ILD patients and to investigate the clinical characteristics and outcomes of ANCA-positive RA-ILD patients. METHODS Data from 104 RA-ILD patients with available ANCA results at our centre from March 2010 to June 2024 were retrospectively reviewed. ANCA positivity was defined as the presence of any one or a combination of perinuclear ANCAs (P-ANCAs), cytoplasmic ANCAs (C-ANCAs), anti-MPO or anti-PR-3. Clinical data from each patient's initial diagnosis were collected and analysed. The clinical characteristics and survival of the ANCA-positive and ANCA-negative RA-ILD groups were compared. RESULTS Thirty-three out of the 104 (31.7%) RA-ILD patients were positive for ANCAs. The percentages of patients positive for P-ANCAs, MPO-ANCAs, C-ANCAs, and PR3-ANCAs were 27.9% (29/104), 1.9% (2/104), 1.9% (2/104), and 1.0% (1/104), respectively. Compared with ANCA-negative RA-ILD patients, a greater proportion of ANCA-positive RA-ILD patients had respiratory symptoms, serum autoantibody (ANA) positivity, poorer baseline pulmonary function, and acute exacerbations of ILD (AE-ILD). The usual interstitial pneumonia (UIP) pattern (57.6%) was the most common chest high-resolution computed tomography (HRCT) pattern observed. ANCA-positive RA-ILD patients were more likely to have traction bronchiectasis (P = 0.029), honeycombing (P < 0.001), and oddly shaped cysts (P = 0.020) than were ANCA-negative RA-ILD patients. Univariate Cox analysis revealed that P-ANCA positivity [hazard ratio (HR) = 2.24, 95% confidence interval (CI): 0.91-5.52; P = 0.046] is a trend of survival association in RA-ILD, but this was not confirmed in the multivariate analysis. Multivariate Cox analyses revealed that a history of smoking (HR = 2.53, 95% CI: 1.10-5.83; P = 0.030) and a systolic pulmonary artery pressure (SPAP) ≥ 37 mmHg (HR = 10.24, 95% CI: 4.07-25.77; P < 0.001) were independently associated with shorter survival in RA-ILD patients. CONCLUSIONS The prevalence of ANCAs in patients with RA-ILD is high, and ANCA testing could be considered in the diagnostic workup for RA-ILD. Oddly shaped cysts with or without a UIP pattern may be a characteristic chest imaging manifestation of ANCA-positive RA-ILD. More attention should be given to RA-ILD patients who have elevated SPAP.
Collapse
Affiliation(s)
- Zhe Huang
- Department of Emergency, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Haishu District, Ningbo, Zhejiang, China
| | - Tingting Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Rongdan Lu
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Haijun Zhou
- Department of Rheumatology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yun Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Li Huang
- Department of Emergency, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Haishu District, Ningbo, Zhejiang, China
| | - Yongxiong Gan
- Department of Emergency, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Haishu District, Ningbo, Zhejiang, China.
| | - Hequn He
- Department of Emergency, The First Affiliated Hospital of Ningbo University, No. 59, Liuting Street, Haishu District, Ningbo, Zhejiang, China.
| |
Collapse
|
5
|
Diomatari C, Martin GP, Jenkins DA, Jani M. Clinical prediction models for medication adverse events in patients with rheumatic and musculoskeletal conditions: A systematic literature review. Semin Arthritis Rheum 2025; 73:152728. [PMID: 40262328 DOI: 10.1016/j.semarthrit.2025.152728] [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: 02/06/2025] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVES This systematic review aims to identify, summarize, and evaluate the methodological quality of existing clinical prediction models (CPMs) that predict adverse events (AEs) associated with medications prescribed for rheumatic and musculoskeletal diseases (RMDs). METHODS We searched PubMed, Embase, and Medline databases up to March 2024. Studies were included if they developed multivariable CPM predicting AEs in adult patients using RMD medications. Data extraction and quality assessment were conducted using the Critical Appraisal and Data Extraction for Systematic Reviews of Prediction Modelling Studies (CHARMS) and Prediction model Risk Of Bias Assessment Tool (PROBAST) checklists to ensure consistent reporting and assess the risk of bias (ROB). RESULTS Of 2406 studies identified, 1734 titles/abstracts were screened, and 38 were reviewed in full. Twelve studies reporting 17 CPMs met eligibility criteria. Most CPMs (76.4 %) focused on rheumatoid arthritis and disease modifying anti-rheumatic drugs (DMARDs) such as methotrexate (69.2 %) and biologic drugs (15.3 %). Cox proportional hazards or logistic regression models were commonly used. Twelve models (70.5 %) had high overall ROB due to inappropriate variable selection methods and sample size. CONCLUSIONS This is the first systematic review summarising CPMs for AEs associated with RMD medications. It highlights that existing CPMs are affected by methodological pitfalls, including inappropriate variable selection and lack of clear sample size justification. Future models could consider a broader range of RMDs and medications. Emerging methods such as machine learning with the ability to model complex interactions, and multi-outcome CPMs to predict several AEs to one class of drug may improve predictions.
Collapse
Affiliation(s)
- Christina Diomatari
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Glen P Martin
- Centre for Health Informatics, Division of Informatics, Imaging and Data Sciences, The University of Manchester, Manchester, UK
| | - David A Jenkins
- Centre for Health Informatics, Division of Informatics, Imaging and Data Sciences, The University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Meghna Jani
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
6
|
Li M, Sun Y, Shan X, Tong Y, Fu Y, Ma X, Sun Z, Xiang Y, Zhu Y, Wang T, Wang X, Zhang J, Niu D. Roles of Immunity and Endogenous Retroelements in the Pathogenesis of Rheumatoid Arthritis and Treatment Strategies. Funct Integr Genomics 2025; 25:85. [PMID: 40205241 DOI: 10.1007/s10142-025-01583-4] [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: 11/18/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic, inflammatory, autoimmune disease that primarily affects the joints. RA usually results in synovial hyperplasia, expansion of "pannus" and destruction of cartilage. The etiology and pathogenesis of RA are not fully understood, but immunity has been shown to play an important role in the development of autoimmune diseases such as RA. In addition, endogenous retroelements, the remnants of ancient retroviruses in the human genome, are involved in cancer and/or immune disorders. As evidenced by increasing evidences that the aberrant expression of retroelements induces innate immunity, despite the fact that the expression of most retroelements has been epigenetically suppressed over a long period of evolution. With the continuous development of disease-modifying anti-rheumatic drugs (DMARDs), RA disease activity has been alleviated and improved. Unfortunately, some patients have a limited response to DMARDs, and the drugs also have the disadvantages of some side effects and high economic costs. This review summarizes the pathogenic mechanisms of RA and endogenous retroelements in autoimmunity, and concludes with a summary of treatments for RA, along with new therapeutic recommendations.
Collapse
Affiliation(s)
- Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Yuhong Tong
- Fourth School of Clinical Medicine, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310053, China
| | - Yite Fu
- Nanjing Outstanding Gene Technology Co, Nanjing, 210018, Jiangsu, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Yun Xiang
- Jinhua Academy of Agricultural Sciences, Jinhua, 321000, Zhejiang, China
| | - Yidan Zhu
- Jinhua Academy of Agricultural Sciences, Jinhua, 321000, Zhejiang, China
| | - Tao Wang
- Nanjing Outstanding Gene Technology Co, Nanjing, 210018, Jiangsu, China
| | - Xin Wang
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
7
|
Khalilzad MA, Mohammadi J, Amirsaadat S, Najafi S, Zare S, Nilforoushzadeh MA, Khalilzad M, Amirkhani MA, Peyrovan A, Khalili SFS, Farahani A, Zare S. Therapeutic potential of apoptotic vesicles in modulating inflammation, immune responses, and tissue regeneration. J Nanobiotechnology 2025; 23:260. [PMID: 40170079 PMCID: PMC11960034 DOI: 10.1186/s12951-025-03278-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
The process of apoptosis plays a crucial role in tissue homeostasis, immune system regulation, and organ formation. Apoptotic vesicles (ApoEVs) are involved in efferocytosis, the process by which phagocytes ingest dead cells. ApoEVs also have potential therapeutic applications in cancer treatment, ischemic diseases, and their anti-inflammatory properties make them incredibly versatile for medical applications. These vesicles can induce apoptosis in cancer cells, provide tumor antigens for cancer vaccines, and even serve as effective drug delivery systems. Moreover, they can target hypoxic cells, inhibit inflammatory cell death pathways, and promote tissue regeneration. Also, their potential in addressing inflammatory disorders such as gastrointestinal ailments, osteoarthritis, and diabetes is promising. Additionally, ApoEVs can polarize anti-inflammatory immune cells and suppress inflammatory immune responses which make them a viable option for addressing the unmet need for novel anti-inflammatory medications. Despite a wealth of reviews examining the applications of ApoEVs, very few have thoroughly investigated the mechanisms underlying their anti-inflammatory effects. This distinctive approach positions the current review as timely and immensely relevant, illuminating the intriguing ways these entities function beyond their established advantages.
Collapse
Affiliation(s)
- Mohammad Amin Khalilzad
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Mohammadi
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran.
| | - Soumayeh Amirsaadat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Laserin Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran.
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran.
| | - Mohammad Ali Nilforoushzadeh
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Skin Repair Research Center, Jordan Dermatology and Hair Transplantation Center, Tehran, Iran.
| | - Mitra Khalilzad
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Aysan Peyrovan
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Atefeh Farahani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Zare
- Laserin Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Kumar H, Gupta N, Dar MO, Dangroo NA, Gupta S. O-Acetyl Bakuchiol Exhibits Analgesic, Anti-Inflammatory and Anti-Arthritic Effects: A Combined In Silico and In Vivo Experimental Study. Chem Biodivers 2025; 22:e202402566. [PMID: 39639544 DOI: 10.1002/cbdv.202402566] [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: 10/08/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 12/07/2024]
Abstract
In the present manuscript, we evaluated the analgesic, anti-inflammatory and anti-arthritic effects of bakuchiol derivative, O-acetyl bakuchiol (BAc), at 5, 10 and 20 mg/kg p.o. doses in adult Sprague-Dawley rats. BAc at 20 mg/kg p.o. exhibited maximum analgesic (47.4%), anti-inflammatory (66.50%) and anti-arthritic effects. Among the different parameters studied for anti-arthritic effects, BAc at 20 mg/kg increased the body weight (2.89%), decreased the paw thickness (72.46%) and decreased the paw inflammation (48.59%) in Freund's complete adjuvant (FCA)-induced arthritic rats. In haematological/serum and biochemical parameters, BAc at 20 mg/kg brought the altered levels of erythrocyte sedimentation rate (ESR), Hb, HCT, RBC, TLC and platelet count, HDL, cholesterol, triglycerides, C-reactive protein (CRP) and rheumatoid arthritis (RA) factor near to the normal. Moreover, the spleen weight was also reduced in BAc-treated rats at 20 mg/kg. Histopathological analysis further revealed that a reduction in paw inflammation was observed in paw ankle joints, and no inflammation was observed in brain, liver, lungs and kidney in BAc-treated rats at 20 mg/kg p.o. Molecular docking studies further revealed that BAc has better binding affinity for tumour necrosis factor alpha (TNF-α) (Protein Data Bank [PDB] ID: 7JRA) -7.19 kcal/mol comparable to standard methotrexate with binding affinity -9.56 kcal/mol. Therefore, the present investigation provided the basis for the development of BAc as a potential candidate for the treatment of arthritis.
Collapse
Affiliation(s)
- Hitesh Kumar
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Nidhi Gupta
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Mohammad Ovais Dar
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Nisar A Dangroo
- Department of Chemistry (Organic Chemistry Division), Islamic University of Science and Technology, Awantipora, J&K, India
| | - Sumeet Gupta
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| |
Collapse
|
9
|
Huang R, He X, Meng Q, Yan G, Dong K, Tian Y. Melittin: A promising therapeutic agent for rheumatoid arthritis treatment. Toxicon 2025; 258:108335. [PMID: 40147796 DOI: 10.1016/j.toxicon.2025.108335] [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: 11/30/2024] [Revised: 02/23/2025] [Accepted: 03/25/2025] [Indexed: 03/29/2025]
Abstract
Rheumatoid Arthritis (RA) is a globally prevalent chronic disease that presents significant challenges in terms of curability. The etiology and onset of RA are not readily identifiable, and as the disease advances, it is characterized by joint pain, swelling, and damage, potentially resulting in paralysis. The adverse effects associated with existing pharmacological treatments are considerable, and prolonged use may pose significant health risks. Melittin, a naturally occurring anti-rheumatic compound, has garnered increasing scholarly interest. Melittin has demonstrated the potential to significantly augment the therapeutic efficacy of certain first-line pharmacological agents while minimizing adverse effects, thereby rendering it appropriate for prolonged use. Melittin's mechanisms of action in treating RA encompass anti-inflammatory effects, immunomodulatory effects, analgesic effects, reduction of cardiovascular disease risk, and organ-protective effects, targeting multiple aspects of RA to alleviate symptoms. Clarifying the biological functions of melittin and its mechanisms in treating RA can provide valuable insights for the application of melittin in the intervention of RA disease progression. However, melittin can cause allergic reactions, hemolysis, and cytotoxicity in the body, which limit its application. Research has shown that strategies including melittin-based nanomodification, immunoconjugation, and structural regulation can improve the specificity of melittin, decrease cytotoxicity, and mitigate the lytic effects on non-target cells of bee venom. These findings suggest that melittin holds promise for clinical applications. This article provides a comprehensive overview of the disease progression associated with RA, examines the biological properties of Mmelittin, and discusses therapeutic strategies for utilizing melittin in the treatment of rheumatoid arthritis. The objective is to mitigate the toxic side effects of melittin while enhancing its targeted anti-inflammatory effects, thereby investigating its potential clinical value in the prevention of rheumatoid arthritis.
Collapse
Affiliation(s)
- Rong Huang
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honeybee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Xiying He
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, 650032, China
| | - Qingxin Meng
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honeybee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Guangyu Yan
- Department of Rheumatology, Zhenxiong County Hospital of Traditional Medicine, Zhaotong, 657299, China
| | - Kun Dong
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honeybee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China.
| | - Yakai Tian
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honeybee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
10
|
Nazakat L, Ali S, Summer M, Nazakat F, Noor S, Riaz A. Pharmacological modes of plant-derived compounds for targeting inflammation in rheumatoid arthritis: A comprehensive review on immunomodulatory perspective. Inflammopharmacology 2025; 33:1537-1581. [PMID: 40074996 DOI: 10.1007/s10787-025-01664-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/28/2024] [Indexed: 03/14/2025]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is one of the most prevalent autoimmune, chronic, inflammatory disease characterized by joint inflammation, synovial swelling, loss of articular structures, swelling, and pain. RA is a major cause of discomfort and disability worldwide, associated with infectious agents, genetic determinants, epigenetic factors, advancing age, obesity, and smoking. Although conventional therapies for RA alleviate the symptoms, but their long-term use is associated with significant side effects. This necessitates the urge to discover complementary and alternative medicine from natural products with minimum side effects. PURPOSE In this review, natural product's potential mechanism of action against RA has been documented in the setting of in-vivo, in-vitro and pre-clinical trials, which provides new treatment opportunities for RA patients. The bioefficacy of these natural product's bioactive compounds must be further studied to discover novel natural medications for RA with high selectivity, improved effectiveness, and economic replacement with minimum side effects. STUDY DESIGN AND METHODS The current review article was designed systematically in chronological order. Plants and their phytochemicals are discussed in an order concerning their mode of action. All the mechanisms of action are depicted in diagrams which are thoroughly generated by the Chembiodraw to maintain the integrity of the work. Moreover, by incorporating the recent data with simple language which is not incorporated previously, we tried to provide a molecular insight to the readers of every level and ethnicity. Moreover, Google Scholar, PubMed, ResearchGate, and Science Direct databases were used to collect the data. SOLUTION Traditionally, various plant extracts and bioactive compounds are effectively used against RA, but their comprehensive pharmacological mechanistic actions are rarely discussed. Therefore, the objective of this study is to systematically review the efficacy and proposed mechanisms of action of different plants and their bioactive compounds including Tripterygium wilfordii Hook F (celastrol and triptolide), Nigella sativa (thymoquinone), Zingiber officinale (shogaols, zingerone), Boswellia serrata (boswellic acids), Curcuma longa (curcumin), and Syzygium aromaticum (eugenol) against rheumatoid arthritis. CONCLUSION These plants have strong anti-inflammatory, anti-oxidant, and anti-arthritic effects in different study designs of rheumatoid arthritis with negligible side effects. Phytomedicines could revolutionize pharmacology as they act through alternative pathways hence seeming biocompatible.
Collapse
Affiliation(s)
- Laiba Nazakat
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan.
| | - Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Fakiha Nazakat
- Department of Chemistry, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Shehzeen Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Anfah Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| |
Collapse
|
11
|
Rangaswamy R, Sneha S, Hemavathy N, Umashankar V, Jeyakanthan J. Computational discovery of AKT serine/threonine kinase 1 inhibitors through shape screening for rheumatoid arthritis intervention. Mol Divers 2025; 29:1287-1303. [PMID: 38970640 DOI: 10.1007/s11030-024-10910-z] [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: 11/29/2023] [Accepted: 06/02/2024] [Indexed: 07/08/2024]
Abstract
Rheumatoid Arthritis (RA) is a chronic, symmetrical inflammatory autoimmune disorder characterized by painful, swollen synovitis and joint erosions, which can cause damage to bone and cartilage and be associated with progressive disability. Despite expanded treatment options, some patients still experience inadequate response or intolerable adverse effects. Consequently, the treatment options for RA remain quite limited. The enzyme AKT1 is crucial in designing drugs for various human diseases, supporting cellular functions like proliferation, survival, metabolism, and angiogenesis in both normal and malignant cells. Therefore, AKT serine/threonine kinase 1 is considered crucial for targeting therapeutic strategies aimed at mitigating RA mechanisms. In this context, directing efforts toward AKT1 represents an innovative approach to developing new anti-arthritis medications. The primary objective of this research is to prioritize AKT1 inhibitors using computational techniques such as molecular modeling and dynamics simulation (MDS) and shape-based virtual screening (SBVS). A combined SBVS approach was employed to predict potent inhibitors against AKT1 by screening a pool of compounds sourced from the ChemDiv and IMPPAT databases. From the SBVS results, only the top three compounds, ChemDiv_7266, ChemDiv_2796, and ChemDiv_9468, were subjected to stability analysis based on their high binding affinity and favorable ADME/Tox properties. The SBVS findings have revealed that critical residues, including Glu17, Gly37, Glu85, and Arg273, significantly contribute to the successful binding of the highest-ranked lead compounds at the active site of AKT1. This insight helps to understand the specific binding mechanism of these leads in inhibiting RA, facilitating the rational design of more effective therapeutic agents.
Collapse
Affiliation(s)
- Raghu Rangaswamy
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Tamil Nadu, Karaikudi, 630 003, India
| | - Subramaniyan Sneha
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Tamil Nadu, Karaikudi, 630 003, India
| | - Nagarajan Hemavathy
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Tamil Nadu, Karaikudi, 630 003, India
| | - Vetrivel Umashankar
- Virology & Biotechnology/Bioinformatics Division, ICMR-National Institute for Research in Tuberculosis, Chennai, Tamil Nadu, 600 031, India
| | - Jeyaraman Jeyakanthan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Tamil Nadu, Karaikudi, 630 003, India.
| |
Collapse
|
12
|
Viswanathan A, Brahma N, Vimal S. The future of autoimmune disease care: merging genomic insights and imaging for enhanced prognosis in rheumatoid arthritis. Clin Rheumatol 2025; 44:1863-1866. [PMID: 39985657 DOI: 10.1007/s10067-024-07234-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/22/2024] [Accepted: 11/06/2024] [Indexed: 02/24/2025]
Affiliation(s)
- Akshaya Viswanathan
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to Be University), Chennai, 600 077, Tamil Nadu, India
| | - Neha Brahma
- Department of Biochemistry, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, 602105, Tamil Nadu, India
| | - S Vimal
- Department of Biochemistry, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, 602105, Tamil Nadu, India.
| |
Collapse
|
13
|
Li Y, Duan Q, Huang J, Zhao P, Cai K. Advances in injectable drug delivery systems for the treatment of rheumatoid arthritis. BIOMATERIALS TRANSLATIONAL 2025; 6:40-54. [PMID: 40313572 PMCID: PMC12041806 DOI: 10.12336/biomatertransl.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/04/2024] [Accepted: 11/01/2024] [Indexed: 05/03/2025]
Abstract
Rheumatoid arthritis is a chronic autoimmune disease characterised by inflammation and progressive joint damage, necessitating innovative therapeutic strategies. Conventional rheumatoid arthritis treatments, including disease-modifying antirheumatic drugs, nonsteroidal anti-inflammatory drugs, glucocorticoids, and biologics, often administered through systemic or intra-articular ways. These drugs often have low accumulation and/or retention in articular cartilage, causing dose-limiting toxicities and reduced efficacy. This review summarises recent advances in injectable drug delivery systems, specifically hydrogels, microspheres, and nanoparticles, highlighting their potential to enhance rheumatoid arthritis therapy. The outstanding potential of these systems was demonstrated; however, substantial research remains to be conducted to optimise their performance and safety.
Collapse
Affiliation(s)
- Ying Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Qiaojian Duan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Jinjin Huang
- Morgridge College of Education, University of Denver, Denver, CO, USA
| | - Peng Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
14
|
Sami MM, Jamai MJ, Alkareem TAA, Ayram NB. Low progesterone levels and their role in the co-existence of polycystic ovary syndrome and rheumatoid arthritis: A comprehensive analysis among Iraqi patient. J Steroid Biochem Mol Biol 2025; 247:106680. [PMID: 39870327 DOI: 10.1016/j.jsbmb.2025.106680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Polycystic ovarian syndrome (PCOS) is a low-grade and chronic inflammation defined by irregular hormonal status that primarily triggers females in their reproductive age. Multi cysts are a primary manifestation of PCOS; a high level of androgen production characterizes the condition via ovaries. Rheumatoid arthritis (RA) is a chronic, systemic, and symmetrical inflammatory autoimmune disease that affects 1-2 % of adults. Females are more likely to generate RA. During the inflammatory activity, immune cells attack the synovium and the synovial space. This invasion is essential in releasing many cytokines in the synovial and joint spaces, leading to joint damage and pain, stiffens, heat, and tenderness in the joint. To evaluate the strength of the link between PCOS and RA, the cross-sectional study examined hormonal, metabolic, and autoantibodies in PCOS, RA as a positive control and the study groups. Statistical analysis Shapiro-Wilk test, student t-test, one-way ANOVA, and multi-linear regression analysis were used to evaluate the results. The data highlights significant values for the BMI, WHR, and hirsutism of PCOS and RA groups in comparison to the negative control. The ANOVA results of these parameters also showed a significant p < 0.05 among the groups. According to the negative control, the levels of insulin, HOMA-IR, testosterone, LH, estradiol, and CRP showed a substantial increase in the PCOS group. Also, the RA group showed a significant p < 0.05 rise in CRP, RF, and Ani-CCP, and the ANOVA results showed significant value among the groups under investigation. Progesterone D as a model showed a correlation with Anti-CCP B, RF C, Anti-CCP C, CRP D, RF D, and Anti-CCP D with the highest level of f2 between other models. In addition, statistical tests show that progesterone D with R2= 0.565 and RMSE equal to 0.996 have heteroscedasticity, which means that low levels of progesterone are associated inversely with high levels of RF and Anit-CCP. There is a relative association between the progesterone D model and corresponding predictions. Regardless of solid f2, only 56 % of the sample shows an association between the model and predictors; this relation may differ if we consider the study's limitations.
Collapse
Affiliation(s)
- Mohammed Mahdi Sami
- Department of Remote Sensing, College of Remote Sensing and geophysics-Al-Karkh University of Science, Baghdad, Iraq.
| | - Mataz J Jamai
- Department of Remote Sensing, College of Remote Sensing and geophysics-Al-Karkh University of Science, Baghdad, Iraq.
| | - Tamara Ahmed Abd Alkareem
- Department of Remote Sensing, College of Remote Sensing and geophysics-Al-Karkh University of Science, Baghdad, Iraq.
| | - Nabeel Bunyan Ayram
- Department of chemistry-College of Science, Mustansiriyah University, Baghdad, Iraq.
| |
Collapse
|
15
|
Rafati A, Ramezani R, Esmaeili Gouvarchin Ghaleh H, Bahrami S, Alvanegh AG, Masoudi MR. Calcitriol Treated Mesenchymal Stem Cells Modulated Immune Response in Collagen-Induced Rheumatoid Arthritis in BALB/c Mice. Transplant Proc 2025; 57:355-363. [PMID: 39837674 DOI: 10.1016/j.transproceed.2024.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND AND AIM Rheumatoid arthritis (RA) is a chronic inflammatory disease that primarily involves synovial joints. During the past decade, disease-modifying antirheumatic drugs and biologic agents have been introduced for the treatment of RA. However, they have limitations, including incomplete treatment response, adverse effects requiring drug withdrawal, fall off in efficacy over time, high cost of biologic agents, and refractory cases. Consequently, there is a need to establish safe and effective advanced therapeutic modalities for RA to overcome the shortcomings of current treatments. METHODS MSCs after isolation were exposed to 200 nM calcitriol. Rheumatoid arthritis was induced in BALB/c mice using collagen and Freund's complete adjuvant. One week after immunization, the mice were divided into 3 groups including without treatment, groups treated with untreated and treated MSCs. One week after the last injection, mice sacrificed and samples were taken and the desired evaluations were done. RESULTS Our results revealed that the respiratory burst capacity, neutrophil phagocytosis, and nitric oxide production in the population of splenocytes were higher in the positive control group compared to the treatment groups. Also, the level of production of IL-4, IL-10 and TGF-β cytokines and INF-γ and IL-17 cytokines showed a significant increase and decrease, respectively, compared to the positive control group. CONCLUSION Treatment of MSCs with calcitriol leads to an improvement in regulatory function and inhibitory effects on inflammatory mediators of innate immune cells, particularly splenocytes, in a rheumatoid arthritis model compared to untreated mesenchymal stem cells.
Collapse
Affiliation(s)
- Alireza Rafati
- Department of Medical Genetics, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Reihaneh Ramezani
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hadi Esmaeili Gouvarchin Ghaleh
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shabnam Bahrami
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Mahmood Reza Masoudi
- School of Medical Sciences, Emam Reza Hospital Sirjan Faculty of Medical Sciences, Sirjan, Iran
| |
Collapse
|
16
|
Liu L, Ambe K, Onishi M, Yoshii Y, Makino T, Tohkin M. Comparison of the Efficacy and Safety of Disease-Modifying Antirheumatic Drugs Combination Therapies: A Systematic Review and Network Meta-Analysis. Clin Transl Sci 2025; 18:e70156. [PMID: 40033744 PMCID: PMC11876292 DOI: 10.1111/cts.70156] [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: 11/21/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025] Open
Abstract
There are several disease-modifying antirheumatic drugs currently available to treat rheumatoid arthritis (RA). However, the optimal combination therapy with methotrexate for treating RA remains unclear. We aimed to identify combination therapies with high-efficacy and safety by employing the Bayesian method in a network meta-analysis. We systematically searched PubMed, Embase, CENTRAL, Ichushi web, and PMDA review reports and application materials through October 2020, and found 86 randomized controlled trials. The primary efficacy outcome was the 50% improvement rate according to the American College of Rheumatology criteria (ACR50), and the primary safety outcome was the incidence of serious adverse events. We calculated odds ratios (ORs) and its 95% credible intervals (CrIs) between each treatment, and the surface under the cumulative ranking curve (SUCRA) score for each treatment to rank disease-modifying antirheumatic drug combinations. Individually, most disease-modifying antirheumatic drugs combined with methotrexate are more likely to achieve ACR50 than methotrexate monotherapy, with significant differences (p < 0.05), whereas the incidence of serious adverse events was not significantly different compared with methotrexate monotherapy (p > 0.05). Infliximab combined with methotrexate had the highest efficacy ranking (OR = 10.53, 95% CrI: [3.20, 42.87], SUCRA score: 0.884), and etanercept combined with methotrexate had the highest safety ranking (OR = 0.29, 95% CrI: [0.03, 2.04], SUCRA score: 0.893). Comprehensive cluster analysis revealed that the combination of etanercept, an Fc-fusion protein targeting tumor necrosis factor α, with methotrexate demonstrated higher efficacy and safety. These findings could support the selection of combination therapies for the treatment of RA.
Collapse
Affiliation(s)
- Linfeng Liu
- Department of Regulatory Science, Graduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
- Department of Pharmacognosy, Graduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| | - Kaori Ambe
- Department of Regulatory Science, Graduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| | - Mayu Onishi
- Department of Regulatory Science, Graduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| | - Yuka Yoshii
- Department of Regulatory Science, Graduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| | - Toshiaki Makino
- Department of Pharmacognosy, Graduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| | - Masahiro Tohkin
- Department of Regulatory Science, Graduate School of Pharmaceutical SciencesNagoya City UniversityNagoyaJapan
| |
Collapse
|
17
|
Sharma S, Ghosh R, Marianesan AB, Hussain S, Pandey JD, Kumar M. Nanostructured lipid carriers in Rheumatoid Arthritis: treatment, advancements and applications. Inflammopharmacology 2025; 33:941-958. [PMID: 40025299 DOI: 10.1007/s10787-025-01669-2] [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: 01/02/2025] [Accepted: 01/21/2025] [Indexed: 03/04/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects the joints and causes pain, swelling, and deformity. Current treatments, including nonsteroidal anti-inflammatory drugs (NSAIDs), corticosteroids, and disease-modifying antirheumatic drugs, often have limited efficacy and adverse side effects. Nanostructured lipid carriers (NLCs) are promising drug delivery agents for treating RA. NLCs are comprised of solid and liquid lipids, forming a nanostructured matrix that enhances drug solubility, stability, and controlled release. They offer advantages over traditional carriers such as improved skin penetration, increased bioavailability, and reduced systemic side effects. Topical NLC formulations show improved stability and skin absorption, targeting drugs specifically to the affected joints, thus reducing the required dose and systemic exposure. Studies on NLCs for delivering anti-inflammatory and antirheumatic drugs, such as methotrexate, indomethacin, and curcumin, in RA animal models indicate the potential for improved therapeutic efficacy and safety. NLCs represent a promising approach for targeted RA drug delivery, offering better efficacy, fewer side effects, and higher patient compliance. However, further research is needed to optimize NLC formulations and evaluate their clinical efficacy and safety in RA patients. The development of NLC-based drug delivery systems for RA treatment may lead to more effective and well-tolerated therapies, thereby improving the quality of life of patients with this debilitating disease.
Collapse
Affiliation(s)
- Swarnika Sharma
- Hari College of Pharmacy, Malhipur Road Jandheri, Saharanpur, Uttar Pradesh, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India
| | | | - Sumaya Hussain
- College of Pharmacy,, Stephens Group of Institutions, Jammu, 181102, Jammu and Kashmir, India
| | - Jai Deo Pandey
- Rajarshi Rananjay Sinh College of Pharmacy, Maharaja Bhawan Baksh Singh Nagar, Amethi, Uttar Pradesh, 227405, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India.
| |
Collapse
|
18
|
Cong S, Wang N, Pei H, Li Z, Meng Y, Maimaitituersun S, Zhao X, Wan R, Wan Q, Luo L, Bian Y, Wen W, Cui H. Syringin inhibits the crosstalk between macrophages and fibroblast-like synoviocytes to treat rheumatoid arthritis via PDE4. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156401. [PMID: 39842374 DOI: 10.1016/j.phymed.2025.156401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/28/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Syringin (SRG) is well-known for its anti-inflammatory effects. However, its pharmacological mechanisms against rheumatoid arthritis (RA) are not fully understood. MATERIALS AND METHODS We assessed the anti-RA effects of SRG using a collagen-induced arthritis (CIA) rat model. And, we employed single-cell RNA sequencing (scRNA-seq) to analyze the changes in cell types and gene expression in the synovial tissues. Building on these observations, we investigated the effects of SRG on M1 macrophage polarization and RA-fibroblast-like synoviocytes (FLS) proliferation. RESULTS Our findings highlighted the anti-RA effects of SRG on CIA rat. scRNA-seq of rat synovial tissues revealed significant changes in M1 and RA-FLS. Specifically, SRG decreased the levels of inflammatory factors in the supernatants of LPS and IFN-γ induced THP-1 cells and downregulated M1-polarized markers in these cells. Further analysis indicated that SRG's regulation of phosphodiesterase 4 (PDE4) and its associated factors was crucial for its anti-M1 polarization effects. Besides, we found that SRG inhibited the activation of FLS in vivo but showed no direct effects on RA-FLS in vitro. However, in RA-FLS, co-cultured with supernatant from SRG-treated M1-polarized THP-1 cells exhibited lower ability of cell proliferation and activation as compared to co-cultured with supernatant from M1-polarized THP-1 cells. CONCLUSION By integrating scRNA-seq analysis with in vivo and in vitro validations, our study revealed that SRG achieved its anti-RA effects by blocking the interaction between macrophages and RA-FLS, with PDE4 playing a central role. This study may provide a novel research paradigm in studying the multi-cell regulatory mechanisms of natural compounds.
Collapse
Affiliation(s)
- Shan Cong
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Ning Wang
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Huan Pei
- School of Basic Medical, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Zixuan Li
- Medical School, Tibet University, Lhasa 850000, China
| | - Yan Meng
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Saimire Maimaitituersun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Xue Zhao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Rong Wan
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China
| | - Qianqian Wan
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Li Luo
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xinjiang Medical University, Urumchi 830054, China.
| | - Yuhong Bian
- School of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Weibo Wen
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Huantian Cui
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
19
|
Wang X, Yue J, Guo S, Rahmatulla A, Li S, Liu Y, Chen Y. Dissolving microneedles: A transdermal drug delivery system for the treatment of rheumatoid arthritis. Int J Pharm 2025; 671:125206. [PMID: 39799999 DOI: 10.1016/j.ijpharm.2025.125206] [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: 11/16/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder that impacts around 1% of the global population. Up to 20% of people become disabled within a year, which has a severely negative impact on their health and quality of life. RA has a complicated pathogenic mechanism, which initially affects small joints and progresses to larger ones over time. It can damage the skin, eyes, heart, kidney, and lung. Oral medications, intra-articular injections, and other treatments are being used; nevertheless, they have drawbacks, including low bioavailability, numerous adverse effects, and poor patient compliance. Dissolving microneedles (DMNs) are a safe and painless method for transdermal drug delivery, achieved through their ability to physically penetrate the epidermal barrier. They enable targeted drug delivery, significantly enhancing the bioavailability of medications and improving patient compliance. DMNs are particularly effective in delivering both lipophilic and high molecular weight biomolecules. The superior bioavailability of DMNs is demonstrated by the fact that low-dose DMN administration can achieve up to 25.8 times higher bioavailability compared to oral administration. This paper provides a comprehensive review of recent advancements in the use of DMNs for RA treatment, encompassing various materials (such as hyaluronic acid, chitosan, etc.), fabrication techniques (such as the two-step casting method, photopolymerization), and performance evaluations (including morphology, mechanical properties, skin penetration capability, solubility, and pharmacodynamics). Additionally, a thorough safety assessment has been conducted, revealing that DMNs cause minimal skin irritation and exhibit low cytotoxicity, ensuring their safety for clinical application. DMNs provide a highly effective and promising alternative to oral and injectable drug delivery systems, offering a novel therapeutic approach for RA patients that significantly improves treatment outcomes and enhances their quality of life.
Collapse
Affiliation(s)
- Xueni Wang
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China
| | - Jiang Yue
- Department of Endocrinology and Metabolism Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai China
| | - Shijie Guo
- Shengzhou Silk Protein Biotechnology Application Research Institute Zhejiang China
| | - Aysha Rahmatulla
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China
| | - Shuangshuang Li
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China
| | - Yang Liu
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China.
| | - Yuzhou Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin China.
| |
Collapse
|
20
|
Mohaddes AA, Saatchi MA, Afshari Chamanabadi M, Saatchi S, Rostami S, Askari VR. Quantum Health Accelerator ® Ameliorates CFA-Induced Animal Model of Rheumatoid Arthritis: Investigating the Role of Immunomodulatory and Anti-Oxidative Effects. Brain Sci 2025; 15:232. [PMID: 40149754 PMCID: PMC11940038 DOI: 10.3390/brainsci15030232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Introduction: Rheumatoid arthritis (RA) is a systemic inflammatory and autoimmune disease characterized by joint swelling, pain, damage to the cartilage, and disability. In the present study, we aimed to evaluate the anti-oxidant, anti-inflammatory, and immune-modulatory properties of Quantum Health Accelerator® as water enriched with vital bio-quantum information/energy (EW) following complete Freund's adjuvant (CFA)-induced RA in rats. Methods: Forty adult male Wistar rats (180-220 g) were divided into five groups. Arthritis was induced on day one using a single subcutaneous injection of CFA into the left hind footpad of the rat. Rats were assigned to receive methotrexate (MTX, 2 mg/kg/week, intraperitoneally), EW (orally, instead of normal water ad libitum), or their combination for 29 days. The anti-RA activities were determined by paw edema, joint diameter, arthritis score, and several nociceptive behavioral tests (thermal hyperalgesia, cold allodynia, and tactile allodynia). The levels of inflammatory (TNF-α, CRP, RF, and anti-CCP), anti-inflammatory (IL-10), and oxidative stress (NO, MDA, and GSH) markers were measured in serum. In addition, the levels of IFN-γ, IL-4, IL-17, and TGF-β were assessed in the spleen-isolated lymphocytes. Results: We found that treatment with MTX, EW, and their combination remarkably ameliorated thermal hyperalgesia, cold allodynia, and tactile allodynia results following CFA-induced RA in rats. In addition, EW also notably attenuated arthritis score, joint diameter, inflammatory cytokines, and oxidative markers while propagating anti-inflammatory and anti-oxidative mediators. Conclusions: We reveal that EW possesses anti-arthritic effects, possibly through anti-oxidative, anti-inflammatory, and immunomodulatory properties. Collectively, EW may be a promising therapeutic agent for treating RA.
Collapse
Affiliation(s)
- Ali Akbar Mohaddes
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Mohammad Ali Saatchi
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Marziyeh Afshari Chamanabadi
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Saeed Saatchi
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Sadra Rostami
- International Group of Ali Akbar Mohaddes Company, License NO 1090645, Dubai 35360-97797, United Arab Emirates; (A.A.M.); (M.A.S.); (M.A.C.); (S.S.); (S.R.)
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
| |
Collapse
|
21
|
Shakeel L, Shaukat A, Khaliq N, Kashif A, Mujeeb A, Adnan Z, Taj J, Akilimali A. Rheumatoid arthritis: a comprehensive overview of genetic markers, emerging therapies, and personalized medicine. Ann Med Surg (Lond) 2025; 87:696-710. [PMID: 40110258 PMCID: PMC11918739 DOI: 10.1097/ms9.0000000000002890] [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: 09/13/2024] [Accepted: 12/09/2024] [Indexed: 03/22/2025] Open
Abstract
Rheumatoid arthritis (RA) is a prevalent autoimmune disorder marked by chronic inflammatory arthritis and systemic effects. The etiology of RA is complex, involving genetic factors like HLA-DR4 and HLA-DR1, as well as environmental influences, particularly smoking, which heightens disease risk. Affecting approximately 1% of the global population, RA is associated with considerable morbidity and mortality, with its prevalence expected to increase due to demographic shifts, especially in certain regions. RA symptoms commonly manifest between ages 35 and 60 but can also affect children under 16 in cases of juvenile RA. Symptoms include prolonged joint stiffness, pain, fatigue, and, in advanced cases, joint deformities. Current treatment approaches involve disease-modifying antirheumatic drugs, biologics, and glucocorticoids to manage symptoms and slow disease progression, though these treatments often present limitations due to adverse effects and varied patient response. The identification of genetic markers, such as HLA-DRB1 and PTPN22, supports the growing emphasis on personalized treatment strategies that account for genetic and lifestyle factors. Non-pharmacological approaches - diet adjustments, physical activity, and stress management - are increasingly valued for their complementary role in RA management. Lifestyle interventions, including whole-food, plant-based diets and physical therapy, show promise in reducing inflammation and improving joint function. Technological advancements, like telemedicine, mobile health applications, and artificial intelligence, are enhancing RA diagnosis and treatment, making care more precise and accessible. Despite these advancements, RA remains incurable, necessitating continued research into novel therapeutic targets and approaches. A comprehensive, patient-centered approach that integrates lifestyle modifications, preventive strategies, and innovative treatments is essential for improving RA management and patient outcomes.
Collapse
Affiliation(s)
- Laiba Shakeel
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Ayesha Shaukat
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Nawal Khaliq
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Aayat Kashif
- Department of Internal Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Azka Mujeeb
- Department of Internal Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Zahabia Adnan
- Department of Internal Medicine, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Javeria Taj
- Department of Internal Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Aymar Akilimali
- Department of research, Medical Research circle, Goma, Democratic Republic of the Congo
| |
Collapse
|
22
|
Martinez de la Torre A, Clausen AB, Burden AM, Weiler S. Fracture-Related Safety Reporting of JAK Inhibitors: An Analysis from the WHO Global VigiBase. Drug Saf 2025; 48:191-201. [PMID: 39604587 PMCID: PMC11785614 DOI: 10.1007/s40264-024-01490-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION The Janus kinase (JAK) inhibitors are treatment options for autoimmune diseases. Numerous safety concerns have been raised. The European Medicines Agency updated the product information of tofacitinib to include the risk of fractures-but not for other JAK inhibitors. We conducted a global pharmacovigilance analysis of previously investigated JAK inhibitors to investigate a potential class effect. METHODS Individual case safety reports (ICSRs) for all licensed JAK inhibitors were identified from the global WHO pharmacovigilance database. The primary outcome of interest was a bone fracture. Disproportionality analyses using reporting odds ratios (RORs) were conducted. RESULTS We identified 122,037 ICSRs for tofacitinib, 27,786 ICSRs for upadacitinib, 14,616 ICSRs for baricitinib, 830 for filgotinib, and 350 for abrocitinib. Among the ICSRs, we identified 2198 (1.8%), 634 (2.3%), and 144 (1.0%) reports, where a bone fracture was reported for tofacitinib, upadacitinib, and baricitinib, respectively. Few reports were available for the newest drugs filgotinib (10) and abrocitinib (1). JAK inhibitors were associated with increased reporting for fracture: tofacitinib (ROR 3.34, 95% confidence interval [CI] 3.20-3.48), upadacitinib (ROR 4.23, 95% CI 3.80-4.48), baricitinib (ROR 1.80, 95% CI 1.52-2.11) and filgotinib (ROR 2.24, 95% CI 1.11-3.94). Patients with bone fractures were more often female, older and had a higher number of co-reported medications. They were more likely to use glucocorticoids, opioids, and bisphosphonates. CONCLUSION The results from this pharmacovigilance analysis, based on a spontaneous reporting database associated with inherent limitations, suggest a potential risk of fractures with JAK inhibitors, indicating that a class effect cannot be ruled out.
Collapse
Affiliation(s)
- Adrian Martinez de la Torre
- Pharmacoepidemiology Group, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich ETH Zürich, HCI H 407 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Andreas Bech Clausen
- Pharmacoepidemiology Group, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich ETH Zürich, HCI H 407 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea M Burden
- Pharmacoepidemiology Group, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich ETH Zürich, HCI H 407 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Stefan Weiler
- Pharmacoepidemiology Group, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology Zurich ETH Zürich, HCI H 407 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland.
- Institute of Primary Care, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Ferrara F, Zovi A, Langella R, Trama U, Nava E, Comentale F, Primiano F, Russo G, Capuozzo M. Analysis of prescriptive monitoring regarding the current therapeutic landscape of rheumatoid arthritis: the experience of an Italian local health authority. Expert Rev Pharmacoecon Outcomes Res 2025; 25:227-233. [PMID: 39340396 DOI: 10.1080/14737167.2024.2411431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024]
Abstract
INTRODUCTION Rheumatoid Arthritis (RA) is among the most prevalent chronic inflammatory diseases affecting millions of people with a significant expenditure of resources by National Healthcare Systems. This study aimed to analyze all available treatments exclusively for RA to highlight the costs of each treatment type and raise awareness of the use of biosimilar drugs. METHODS In an Italian healthcare authority, all prescriptions made for the diagnosis of RA were extracted to verify consumption expressed in Defined Daily Dose (DDD) and the expenditure incurred expressed in euros. Consequently, a grouping into three major drug categories has been performed: anti - tumor necrosis factor alpha agents (TNFα), other injectable formulations, and novel oral formulations. RESULTS Prescriptions for the second half-year 2022 and 2023 have been analyzed, with a total cost of almost 7 million euros in the sample considered. All pharmaceutical categories showed an increase in consumption, but only anti- TNFα recorded a decrease in costs from 25% in 2022 to 22% in 2023, thanks to the lower cost of the biosimilar drug. CONCLUSION The costs of RA may represent a significant spending commitment for central governments. As a result, actions are needed to encourage the preferential use of biosimilar drugs.
Collapse
Affiliation(s)
| | | | - Roberto Langella
- Italian Society of Hospital Pharmacy (SIFO), SIFO Secretariat of the Lombardy Region, Milan, Italy
| | - Ugo Trama
- General Direction for Health Protection and Coordination of the Campania Regional Health System, Campania Region, Naples, Italy
| | | | | | | | | | | |
Collapse
|
24
|
Zhao H, Wang Y, Ren J. Helicobacter pylori and rheumatoid arthritis: Investigation of relation from traditional Chinese medicine. Microb Pathog 2025; 199:107239. [PMID: 39708982 DOI: 10.1016/j.micpath.2024.107239] [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: 08/28/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune condition that predominantly affects synovial joints, manifesting with joint swelling, pain, and stiffness. In advanced stages, unchecked inflammation can inflict damage on bone and cartilage, resulting in disabilities and deformities of the joints. Additionally, systemic and extra-articular complications may arise due to the consequences of uncontrolled inflammation. Helicobacter pylori (H. pylori) is one of the most prevalent chronic bacterial infections in humans. This microorganism is a spiral-shaped, flagellated, microaerophilic gram-negative bacterium. Prolonged exposure leads to the activation of the immune system, with infected gastric mucosa epithelial cells continuously producing cytokines. This production, in turn, triggers the generation of antibodies as well as T Helper 1 and T Helper 2 effector T cells. The persistent antigenic stimulation resulting from H. pylori infection could lead to the progression of autoimmune diseases. Numerous clinical and pharmacological trials have illustrated the efficacy of traditional Chinese medicine against H. pylori. This review aims to delve into the connection between H. pylori and rheumatoid arthritis so as understand the pathogenesis. The concluding section of this review explores the interplay of Chinese medicine and Helicobacter pylori concerning rheumatoid arthritis.
Collapse
Affiliation(s)
- Hua Zhao
- Department of Rheumatism and Immunology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), No.4, Renmin Road, Shibei District, Qingdao, 266033, China
| | - Yige Wang
- Shandong University of Traditional Chinese Medicine, No.16369, Jingshi Road, Lixia District, Jinan, 250013, China
| | - Jiahui Ren
- Department of Rheumatism and Immunology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), No.4, Renmin Road, Shibei District, Qingdao, 266033, China
| |
Collapse
|
25
|
Tanaka Y, Atsumi T, Aletaha D, Schulze-Koops H, Fukada H, Watson C, Takeuchi T. The Uncoupling of Disease Activity from Joint Structural Progression in Patients with Rheumatoid Arthritis Treated with Filgotinib. Rheumatol Ther 2025; 12:53-66. [PMID: 39592547 PMCID: PMC11751281 DOI: 10.1007/s40744-024-00725-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
INTRODUCTION While modern treatments can prevent progressive bone destruction in patients with rheumatoid arthritis (RA) achieving clinical remission, it is unclear whether residual clinical activity may cause or be associated with progressive joint damage. This post hoc analysis evaluated the association between clinical disease activity and structural progression in patients with RA treated with filgotinib (FIL) in FINCH 1 (NCT02889796). METHODS Patients with RA and inadequate response to methotrexate (MTX) use were randomized 3:3:2:3 to FIL 200 mg (FIL200) or FIL 100 mg (FIL100) once daily, adalimumab 40 mg biweekly, or placebo, all with background MTX. We evaluated the change from baseline (CFB) in modified total Sharp score (mTSS), erosion score, and joint space narrowing score among patients achieving Clinical Disease Activity Index (CDAI) remission (CDAI ≤ 2.8), low disease activity (LDA; 2.8 < CDAI ≤ 10), medium disease activity (MDA; 10 < CDAI ≤ 22), and high disease activity (HDA; CDAI > 22) at 24 weeks. RESULTS At week 24, the least squares (LS) mean CFB in mTSS was similarly low across treatments among patients who achieved CDAI remission (range 0.00-0.11) or LDA (n = 285 and 575, respectively). In patients with MDA and HDA (n = 471 and 157, respectively), smaller LS mean CFB in mTSS was seen in the FIL200 group vs. the placebo group (P < 0.05 for both). CONCLUSIONS RA clinical remission and LDA achievement were associated with suppressed progression of joint destruction over 24 weeks in all treatment groups. Only FIL200 significantly inhibited joint damage compared with placebo in patients with MDA or HDA, indicating an uncoupling of clinical disease activity and structural progression in patients receiving FIL200. TRIAL REGISTRATION NCT02889796.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan.
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - Hendrik Schulze-Koops
- Division of Rheumatology and Clinical Immunology, Department of Medicine IV, Ludwig Maximilian University, Munich, Germany
| | | | | | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Saitama Medical University, Saitama, Japan
| |
Collapse
|
26
|
Qamar T, Ansari MS, Masihuddin, Mukherjee S. MicroRNAs as Biomarker in Rheumatoid Arthritis: Pathogenesis to Clinical Relevance. J Cell Biochem 2025; 126:e30690. [PMID: 39710998 DOI: 10.1002/jcb.30690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/19/2024] [Accepted: 11/29/2024] [Indexed: 12/24/2024]
Abstract
MicroRNAs (miRNAs) have emerged as intricate players in rheumatoid arthritis (RA), holding promise as discerning biomarkers for diagnostic and prognostic purposes. The lack of sensitivity and specificity in current diagnostic techniques, such as rheumatoid factor (RF) and anti-citrullinated protein antibodies (ACPA), causes diagnosis delays in RA. The miR-146a and miR-155 act in inflammatory cascades and reduce joint deterioration, and miR-223 is paradoxical, acting differently in different illness scenarios. The microenvironment of RA is shaped by the complex modulation of gene expression and cytokine dynamics by miR-126 and miR-24. miRNAs serve as a promising candidate for precision medicine in the management of RA. There are obstacles encountered in validation, delivery optimization, and off-target effect mitigation before miRNA-based biomarkers may be applied in clinical settings. Machine learning (ML) and artificial intelligence (AI) have been used to integrate miRNA expression patterns with clinical data to greatly advance the treatment of RA. Because of the disease's inherent complexity and variability, these state-of-the-art models provide accurate predictions regarding the onset, development, and response to treatment of RA. By using clinical information and miRNA expression data, ML algorithms are revolutionizing the treatment of RA by predicting the onset and course of the disease with remarkably high accuracy. The development of therapeutic modalities and miRNA profiling has great potential to transform the diagnosis, prognosis, and treatment of RA, providing fresh hope for better patient outcomes.
Collapse
Affiliation(s)
- Tooba Qamar
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Md Samsuddin Ansari
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Masihuddin
- Department of Science and Mathematics, Indian Institute of Information Technology, Guwahati, India
| | - Sayali Mukherjee
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, Uttar Pradesh, India
| |
Collapse
|
27
|
Hijjawi N, Tout FS, Azaizeh B, Aljaafreh B. The role of vitamins D, B12, C, and K in modulating inflammation and disease management in rheumatoid arthritis: a comprehensive review. Clin Rheumatol 2025; 44:591-600. [PMID: 39722107 DOI: 10.1007/s10067-024-07285-9] [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: 11/07/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder marked by joint inflammation and destruction. Recent studies emphasize the importance of vitamins D, B12, C, and K in managing RA and enhancing patient health. Vitamin D deficiency is common in RA patients and correlates with increased disease severity, indicating its potential to modulate immune responses and reduce inflammation. Supplementation has shown promise in improving disease activity scores and lowering inflammatory markers. Vitamin B12 is vital for energy and neurological function; its deficiency can worsen fatigue in RA sufferers. Vitamin C, with its antioxidant properties, aids collagen synthesis and may reduce joint inflammation. Vitamin K, particularly through Matrix Gla-Protein (MGP), is essential for bone health and may help prevent joint calcification and osteoporosis. Collectively, these vitamins play critical roles in immune modulation, inflammation reduction, and bone health in RA management, warranting further research on optimal dosages and combinations for effective treatment strategies.
Collapse
Affiliation(s)
- Nawal Hijjawi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Faten S Tout
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan.
| | - Baraah Azaizeh
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Baraah Aljaafreh
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
28
|
Liu J, Jiang Y, Zhang S, Liu S, Su J, Lin C, He X, Wu R, Yang L, Liu H, Duan X, Xu S, Luo H, Liu J, Xie Q, Mi C, Chen L, Zhang N, Gong H, Zhu J, Li Y, Wei H, Qian L, Wang J, Shi X, Jin H, Jiang Z, Xie X, Zhan F, Geng X, Zheng Z, Du Z, Dong G, Sun Y, Zeng X. Ivarmacitinib, a selective Janus kinase 1 inhibitor, in patients with moderate-to-severe active rheumatoid arthritis and inadequate response to conventional synthetic DMARDs: results from a phase III randomised clinical trial. Ann Rheum Dis 2025; 84:188-200. [PMID: 39919893 DOI: 10.1136/ard-2024-226385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
OBJECTIVE To assess the efficacy/safety of ivarmacitinib, a selective Janus kinase (JAK) 1 inhibitor, in patients with moderate-to-severe active rheumatoid arthritis (RA) who had an inadequate response to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs). METHODS Patients were randomised (1:1:1) to receive either placebo (n=188), ivarmacitinib 4 mg (n=189) or ivarmacitinib 8 mg (n=189) once daily, with background csDMARDs allowed. After 24 weeks, patients on placebo switched to ivarmacitinib 4 mg for an additional 28 weeks, while those on ivarmacitinib continued their initial dosage. The primary endpoint was the proportion of patients achieving a 20% improvement in the American College of Rheumatology response criteria (ACR20) at week 24. RESULTS At week 24, ACR20 response rates were significantly higher in the ivarmacitinib 4 mg (70.4%) and 8 mg (75.1%) groups compared with the placebo group (40.4%; both p<0.0001). Both ivarmacitinib doses achieved numerically higher Disease Activity Score 28-joint count C reactive protein of <2.6/≤3.2 response rates compared with placebo. Improvements in efficacy and patient-reported outcomes were sustained through 52 weeks and were noted in patients who switched from placebo after week 24. During the placebo-controlled period, treatment-emergent adverse events (TEAEs) occurred in 81.5% and 90.5% of patients in the ivarmacitinib 4 mg and 8 mg groups, versus 79.3% in the placebo group. Infection-related TEAEs were slightly higher in the ivarmacitinib groups. CONCLUSIONS Ivarmacitinib may offer a potential therapeutic option for patients with RA who have an inadequate response to csDMARDs, with a safety profile that was generally manageable over 1 year of treatment and similar to other JAK inhibitors. TRIAL REGISTRATION NUMBER NCT04333771.
Collapse
Affiliation(s)
- Jinjing Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China
| | - Ying Jiang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China
| | - Shangzhu Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China
| | - Shengyun Liu
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingbo Su
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Changsong Lin
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong He
- Department of Rheumatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Rui Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lei Yang
- Department of Rheumatology and Immunology, Zhengzhou Central Hospital, Zhengzhou, China
| | - Huaxiang Liu
- Rheumatology Department, Qilu Hospital of Shandong University, Jinan, China
| | - Xinwang Duan
- Rheumatology and Immunology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shengqian Xu
- Rheumatology and Immunology Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Luo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Liu
- Department of Rheumatology and Immunology Research Institute, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital of Sichuan University, Chengdu, China
| | - Cundong Mi
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lin Chen
- Department of Rheumatic Immunity, Jilin Province People's Hospital, Changchun, China
| | - Ning Zhang
- Rheumatology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Huiping Gong
- Department of Rheumatology and Clinical Immunology, Peace Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Jing Zhu
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Yasong Li
- Department of Rheumatology and Clinical Immunology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Hua Wei
- Rheumatology and Immunology Department, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Long Qian
- Department of Rheumatology and Clinical Immunology, The Second Hospital of Anhui Medical University, Anhui, China
| | - Jian Wang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Bengbu Medical College, Anhui, China
| | - Xiaofei Shi
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Henan University of Science & Technology, Luoyang, China
| | - Hongtao Jin
- Department of Rheumatology and Immunology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhenyu Jiang
- Department of Rheumatic Immunity, The First Hospital of Jilin University, Changchun, China
| | - Xi Xie
- Department of Clinical Immunology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Feng Zhan
- Department of Rheumatology and Immunology, Hainan General Hospital, Haikou, China
| | - Xiuqin Geng
- Department of Rheumatology and Immunology, Xinxiang Central Hospital, Xinxiang, China
| | - Zhaohui Zheng
- Department of Clinical Immunology, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Zhengfu Du
- Department of Rheumatology and Immunology, Yuncheng Central Hospital, Yuncheng, China
| | - Guangchao Dong
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Yuqi Sun
- Biometric Department, Jiangsu Hengrui Pharmaceuticals, Shanghai, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
29
|
Arcilla CK, Kaeley GS, Thway M. Long-Standing Untreated or Inadequately Treated Seropositive Rheumatoid Arthritis in the Golden Age of Disease-Modifying Antirheumatic Drugs. Cureus 2025; 17:e78474. [PMID: 40051928 PMCID: PMC11883720 DOI: 10.7759/cureus.78474] [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] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease presenting with articular and extra-articular manifestations that could be related to genetic and environmental factors. Seropositive RA is marked by the presence of autoantibodies, which are associated with worse prognoses and increased risk of extra-articular manifestations, including cardiovascular diseases, lung disorders, and malignancies. Untreated RA can lead to substantial morbidity, reduced life expectancy, and poor quality of life. The development of disease-modifying antirheumatic drugs has paved the way for improving disease outcomes and reducing morbidity and complications related to disease. However, adherence to therapy remains a challenge, often influenced by drug intolerance, misconceptions, and negative beliefs regarding potential side effects and drug toxicity. Here, we report three cases presenting with devastating complications due to long-standing untreated or inadequately treated seropositive RA. This emphasizes the huge disease burden and poor quality of life as part of the natural course and progression of untreated RA. Comprehensive patient education on the disease course, treatment benefits, and potential side effects is essential to enhance adherence and facilitate shared decision-making.
Collapse
Affiliation(s)
- Cristine K Arcilla
- Department of Rheumatology, University of Florida College of Medicine - Jacksonville, Jacksonville, USA
| | - Gurjit S Kaeley
- Department of Rheumatology, University of Florida College of Medicine - Jacksonville, Jacksonville, USA
| | - Myint Thway
- Department of Rheumatology, Baptist Health, Jacksonville, USA
| |
Collapse
|
30
|
Tamburini B, Di Liberto D, Pratelli G, Rizzo C, Barbera LL, Lauricella M, Carlisi D, Maggio A, Palumbo Piccionello A, D’Anneo A, Caccamo N, Guggino G. Extra Virgin Olive Oil Polyphenol-Enriched Extracts Exert Antioxidant and Anti-Inflammatory Effects on Peripheral Blood Mononuclear Cells from Rheumatoid Arthritis Patients. Antioxidants (Basel) 2025; 14:171. [PMID: 40002358 PMCID: PMC11851824 DOI: 10.3390/antiox14020171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Rheumatoid arthritis (RA) is a long-term systemic autoimmune disorder that causes joint inflammation, swelling, pain, bone erosion, and deformities. Recent findings emphasize the anti-inflammatory and antioxidant properties of bioactive natural compounds, such as polyphenols extracted from plants and fruits, and their possible synergistic effect when used in combination with current therapies to improve the prognosis and symptoms of inflammatory rheumatic diseases. Here, we report that Sicilian extra virgin olive oil polyphenol-enriched extracts (PE-EVOOs) reduce intracellular reactive oxygen species (ROS) and pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin-1 β (IL-1β), in peripheral mononuclear cells (PBMCs) obtained from both RA patients and healthy subjects (HSs) treated with lipopolysaccharides (LPS) as a control. HPLC-ESI-MS analysis highlighted that PE-EVOOs are rich in different polyphenolic compounds responsible for many of the observed biological effects. At molecular levels, Western blotting analyses revealed that PE-EVOO treatment is associated with the downregulation of the phosphorylated and active form of the inflammatory transcription factor NF-κB and the pro-inflammatory enzyme cyclooxygenase 2 (COX2). In addition, PE-EVOOs upregulated the transcription factor Nrf2 and its target antioxidant enzyme catalase and manganese superoxide dismutase (MnSOD). Collectively, these results suggest a possible use of PE-EVOOs as potential adjuvants for the treatment of RA.
Collapse
Affiliation(s)
- Bartolo Tamburini
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Section of Immunology, University of Palermo, 90127 Palermo, Italy; (B.T.); (N.C.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (C.R.); (L.L.B.); (G.G.)
| | - Diana Di Liberto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Section of Biochemistry, University of Palermo, 90127 Palermo, Italy; (D.D.L.); (G.P.); (D.C.)
| | - Giovanni Pratelli
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Section of Biochemistry, University of Palermo, 90127 Palermo, Italy; (D.D.L.); (G.P.); (D.C.)
| | - Chiara Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (C.R.); (L.L.B.); (G.G.)
| | - Lidia La Barbera
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (C.R.); (L.L.B.); (G.G.)
| | - Marianna Lauricella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Section of Biochemistry, University of Palermo, 90127 Palermo, Italy; (D.D.L.); (G.P.); (D.C.)
| | - Daniela Carlisi
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Section of Biochemistry, University of Palermo, 90127 Palermo, Italy; (D.D.L.); (G.P.); (D.C.)
| | - Antonella Maggio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (A.P.P.)
| | - Antonio Palumbo Piccionello
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, 90128 Palermo, Italy; (A.M.); (A.P.P.)
| | - Antonella D’Anneo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), Laboratory of Biochemistry, University of Palermo, 90127 Palermo, Italy;
| | - Nadia Caccamo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Section of Immunology, University of Palermo, 90127 Palermo, Italy; (B.T.); (N.C.)
| | - Giuliana Guggino
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University Hospital “P. Giaccone”, 90127 Palermo, Italy; (C.R.); (L.L.B.); (G.G.)
| |
Collapse
|
31
|
Westhovens R, Winthrop KL, Kavanaugh A, Greenwald M, Dagna L, Cseuz R, Besuyen R, de Vries D, Modgill V, Le LH, Genovese MC, Emery P, Verschueren P, Alten R. Safety and efficacy of filgotinib in patients with rheumatoid arthritis: final results of the DARWIN 3 long-term extension study. RMD Open 2025; 11:e004857. [PMID: 39884731 PMCID: PMC11784233 DOI: 10.1136/rmdopen-2024-004857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/27/2024] [Indexed: 02/01/2025] Open
Abstract
OBJECTIVES DARWIN 3 (ClinicalTrials.gov: NCT02065700) assessed the safety and efficacy of filgotinib in a long-term extension (LTE) of two phase II randomised controlled rheumatoid arthritis (RA) trials. METHODS Eligible patients completing the 24-week DARWIN 1 (filgotinib plus methotrexate) and DARWIN 2 (filgotinib monotherapy) trials could enrol. Patients received filgotinib 200 mg/day, except 15 men who received filgotinib 100 mg/day. The primary endpoints were safety and tolerability, which were assessed by the incidence of treatment-emergent adverse events (TEAEs). Safety and efficacy analyses included all enrolled patients who received ≥1 dose of filgotinib in DARWIN 3. RESULTS 739 patients entered the LTE. The total patient-years of exposure (PYE) to filgotinib was 3706.3 years; the mean exposure duration was 259.8 weeks. 497 patients (67.3%) discontinued prematurely (including 266 TEAEs and 172 withdrawals due to the patient's decision or 'sponsor request'). Overall exposure-adjusted incidence rate (EAIR) was 67 (95% CI 62 to 72.2)/100 PYE for TEAEs and 3.8 (95% CI 3.2 to 4.5)/100 PYE for serious TEAEs. EAIR of infections was 23.3 (95% CI 21.2 to 25.6)/100 PYE, 1.3 (95% CI 0.9 to 1.7)/100 PYE for serious infections and 1.3 (95% CI 0.9 to 1.7)/100 PYE for herpes zoster. EAIRs of major adverse cardiovascular events (0.19 (95% CI 0.8 to 0.39)/100 PYE) and malignancies (0.6 (95% CI 0.4 to 0.9)/100 PYE) were low. Disease response assessed using non-responder imputation plateaued at LTE week 12 before slowly declining over time, with overall American College of Rheumatology (ACR)20/50/70 response rates of 26.9%/20.2%/14.7% at week 396. CONCLUSION Filgotinib was well tolerated in patients with RA for up to 8 years. Safety and efficacy profiles were maintained in patients previously receiving either filgotinib plus methotrexate or filgotinib monotherapy.
Collapse
Affiliation(s)
- Rene Westhovens
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Kevin L Winthrop
- School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Arthur Kavanaugh
- Division of Rheumatology, Autoimmunity and Inflammation, University of California San Diego, La Jolla, California, USA
| | - Maria Greenwald
- Department of Rheumatology, Desert Medical Advances, Rancho Mirage, California, USA
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), Department of Internal Medicine, IRCCS San Raffaele Hospital, Milan, Italy
- Department of Internal Medicine, Medical School, Vita-Salute San Raffaele University, Milan, Italy
| | - Regina Cseuz
- Department of Rheumatology, Revita Reumatológiai Kft, Budapest, Hungary
| | - Robin Besuyen
- Clinical Development, Galapagos BV, Leiden, Netherlands
| | - Dick de Vries
- Clinical Development, Galapagos BV, Leiden, Netherlands
| | | | - Ly Huong Le
- Biostatistics, Galapagos NV, Mechelen, Belgium
| | - Mark C Genovese
- Clinical Development, Gilead Sciences Inc, Foster City, California, USA
| | - Paul Emery
- University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, NIHR Leeds Biomedical Research Centre, Leeds, UK
| | - Patrick Verschueren
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Rieke Alten
- Department of Internal Medicine and Rheumatology, Schlosspark Klinik, University Medicine Berlin, Berlin, Germany
| |
Collapse
|
32
|
Moradmand H, Ren L. Multistage deep learning methods for automating radiographic sharp score prediction in rheumatoid arthritis. Sci Rep 2025; 15:3391. [PMID: 39870749 PMCID: PMC11772782 DOI: 10.1038/s41598-025-86073-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/08/2025] [Indexed: 01/29/2025] Open
Abstract
The Sharp-van der Heijde score (SvH) is crucial for assessing joint damage in rheumatoid arthritis (RA) through radiographic images. However, manual scoring is time-consuming and subject to variability. This study proposes a multistage deep learning model to predict the Overall Sharp Score (OSS) from hand X-ray images. The framework involves four stages: image preprocessing, hand segmentation with UNet, joint identification via YOLOv7, and OSS prediction utilizing a custom Vision Transformer (ViT). Evaluation metrics included Intersection over Union (IoU), Mean Absolute Error (MAE), Root Mean Squared Error (RMSE), Huber loss, and Intraclass Correlation Coefficient (ICC). The model was trained using stratified group 3-fold cross-validation on a dataset of 679 patients and tested externally on 291 subjects. The joint identification model achieved 99% accuracy. The ViT model achieved the best OSS prediction for patients with Sharp scores < 50. It achieved a Huber loss of 4.9, an RMSE of 9.73, and an MAE of 5.35, demonstrating a strong correlation with expert scores (ICC = 0.702, P < 0.001). This study is the first to apply a ViT for OSS prediction in RA. It presents an efficient and automated alternative for overall damage assessment. This approach may reduce reliance on manual scoring.
Collapse
Affiliation(s)
- Hajar Moradmand
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Lei Ren
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
33
|
Arshad S, Cameron B, Joglekar AV. Immunopeptidomics for autoimmunity: unlocking the chamber of immune secrets. NPJ Syst Biol Appl 2025; 11:10. [PMID: 39833247 PMCID: PMC11747513 DOI: 10.1038/s41540-024-00482-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025] Open
Abstract
T cells mediate pathogenesis of several autoimmune disorders by recognizing self-epitopes presented on Major Histocompatibility Complex (MHC) or Human Leukocyte Antigen (HLA) complex. The majority of autoantigens presented to T cells in various autoimmune disorders are not known, which has impeded autoantigen identification. Recent advances in immunopeptidomics have started to unravel the repertoire of antigenic epitopes presented on MHC. In several autoimmune diseases, immunopeptidomics has led to the identification of novel autoantigens and has enhanced our understanding of the mechanisms behind autoimmunity. Especially, immunopeptidomics has provided key evidence to explain the genetic risk posed by HLA alleles. In this review, we shed light on how immunopeptidomics can be leveraged to discover potential autoantigens. We highlight the application of immunopeptidomics in Type 1 Diabetes (T1D), Systemic Lupus Erythematosus (SLE), and Rheumatoid Arthritis (RA). Finally, we highlight the practical considerations of implementing immunopeptidomics successfully and the technical challenges that need to be addressed. Overall, this review will provide an important context for using immunopeptidomics for understanding autoimmunity.
Collapse
Affiliation(s)
- Sanya Arshad
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Benjamin Cameron
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alok V Joglekar
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
34
|
Kurtulus Ozturk E, Ozturk S, Kelesoglu Dincer AB. The reliability and validity of superb microvascular imaging as a potential disease activity marker in rheumatoid arthritis. ULTRASCHALL IN DER MEDIZIN (STUTTGART, GERMANY : 1980) 2025. [PMID: 39510536 DOI: 10.1055/a-2463-8297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease of unknown etiology that causes progressive and destructive inflammation in the joints. Superb microvascular imaging (SMI) is a new ultrasound technique that allows visualization of slow blood flow in synovitis. This study aimed to report on the clinical value and utility of the SMI technique and its grading for monitoring RA by determining the correlation with clinical disease activity scores (DAS 28) and power Doppler ultrasound (PDUS).All RA patients with clinically apparent synovitis were assessed using DAS 28. Synovitis was investigated with PDUS and SMI and each joint was graded semi-quantitatively. All assessments were carried out at baseline and repeated at least at the 4-month follow-up. Correlations between scores were investigated using Spearman's correlation.60 RA patients with 552 affected joints were recruited. Clinical and sonographic scores were significantly improved at follow-up (p<0.001). SMI showed significantly more joint count and flow signal scores than clinical examination and PDUS. Moderate correlations were found between the SMI score and clinical scores (p<0.001, 0.586 for SMI score vs. DAS 28-CRP, p=0.001, 0.432 for SMI vs. DAS 28-ESR). There were also stronger correlations between the SMI score and PDUS score at both baseline and follow-up (p<0.001, r = 0.817, 0.842 respectively).SMI provides greater utility and ability to detect synovial vascularity and to monitor disease activity than PDUS. A new activity scoring system based on SMI and clinical objective findings is required to improve reliability and validity.
Collapse
Affiliation(s)
| | - Saffet Ozturk
- Radiology, Ministry of Health Ankara Etlik City Hospital, Ankara, Turkey
| | | |
Collapse
|
35
|
Sari MHM, Cervi VF, Custódio VN, Prado VC, da Motta KP, Luchese C, Wilhelm EA, Ferreira LM, Cruz L. Blended ƙ-carrageenan and xanthan gum hydrogel containing ketoprofen-loaded nanoemulsions: Design, characterization, and evaluation in an animal model of rheumatoid arthritis. Drug Deliv Transl Res 2025:10.1007/s13346-024-01786-5. [PMID: 39821868 DOI: 10.1007/s13346-024-01786-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2024] [Indexed: 01/19/2025]
Abstract
This study reports the preparation of hydrogels (HG) made with xanthan gum (XG) and ƙ-carrageenan (KC) polysaccharides containing ketoprofen (KET)-loaded nanoemulsions (NK) and their evaluation in a rheumatoid arthritis (RA) model. The nano-based HGs exhibited nanometric-sized droplets (~ 100 nm), an acidic pH (5.10-6.83), drug content above 85%, a suitable spreadability factor, and pseudoplastic flow behavior. The most promising blend (HGCX 2:1) demonstrated sustained KET release, reaching 81.44 ± 6.11% after 5 h, and superior drug concentration in the skin layers (237.91 ± 41.0 µg/g). The formulation was selected due to its enhanced bioadhesiveness, with the HG-NK formulation showing the highest bioadhesion force and occlusion factor. RA was induced by complete Freund's adjuvant (CFA) intraplantar injection into the left hind paw of male and female Swiss mice. Treatments with HGs were applied to the animals' dorsal region for 7 days. Notably, HG-NK demonstrated remarkable efficacy, reversing mechanical sensitivity in male mice and significantly reducing thermal sensitivity in both genders. Moreover, HG-NK provided a significant reduction in paw edema (52-fold in males, 27-fold in females) and inflammatory markers, such as myeloperoxidase activity (32-fold in males, 14-fold in females) and lipid peroxidation (2.5-fold in males, twofold in females). The formulation also promoted greater permeation of KET across the skin. These findings underscore the significant reduction in inflammatory markers by the HG-NK formulation, highlighting its potent anti-inflammatory effects and potential as a promising therapeutic strategy for managing RA.
Collapse
Affiliation(s)
- Marcel Henrique Marcondes Sari
- Departamento de Farmácia Industrial, Laboratório de Tecnologia Farmacêutica, Programa de Pós-Graduação Em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, RS, CEP 97105-900, Brazil.
- Departamento de Análises Clínicas, Universidade Federal Do Paraná, Curitiba, 80210-170, Brazil.
| | - Verônica Ferrari Cervi
- Departamento de Farmácia Industrial, Laboratório de Tecnologia Farmacêutica, Programa de Pós-Graduação Em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, RS, CEP 97105-900, Brazil
| | - Vanessa Neuenschwander Custódio
- Departamento de Farmácia Industrial, Laboratório de Tecnologia Farmacêutica, Programa de Pós-Graduação Em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, RS, CEP 97105-900, Brazil
| | - Vinicius Costa Prado
- Departamento de Farmácia Industrial, Laboratório de Tecnologia Farmacêutica, Programa de Pós-Graduação Em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, RS, CEP 97105-900, Brazil
| | - Ketlyn Pereira da Motta
- Postgraduate Program in Biochemistry and Bioprospecting, Research Laboratory in Biochemical Pharmacology (LaFarBio), Federal University of Pelotas (UFPel), Pelotas, Brazil
| | - Cristiane Luchese
- Postgraduate Program in Biochemistry and Bioprospecting, Research Laboratory in Biochemical Pharmacology (LaFarBio), Federal University of Pelotas (UFPel), Pelotas, Brazil
| | - Ethel Antunes Wilhelm
- Postgraduate Program in Biochemistry and Bioprospecting, Research Laboratory in Biochemical Pharmacology (LaFarBio), Federal University of Pelotas (UFPel), Pelotas, Brazil
| | - Luana Mota Ferreira
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Paraná, Curitiba, Brazil
| | - Letícia Cruz
- Departamento de Farmácia Industrial, Laboratório de Tecnologia Farmacêutica, Programa de Pós-Graduação Em Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria, RS, CEP 97105-900, Brazil.
| |
Collapse
|
36
|
Wang X, Pan L, Niu D, Zhou J, Shen M, Zeng Z, Gong W, Yang E, Tang Y, Cheng G, Sun C. Jingfang Granules alleviates the lipid peroxidation induced ferroptosis in rheumatoid arthritis rats by regulating gut microbiota and metabolism of short chain fatty acids. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119160. [PMID: 39608616 DOI: 10.1016/j.jep.2024.119160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/21/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation, bone and cartilage damage, musculoskeletal pain, swelling, and stiffness. Inflammation is one of the key factors that induce RA. Jingfang Granule (JFG) is a traditional Chinese medicine (TCM) with significant anti-inflammatory effects. Clinical studies have confirmed that JFG can be used to treat RA, but the mechanism is still vague. PURPOSE This study was designed to evaluate the protective function and the mechanism of JFG on rats with RA. STUDY DESIGN AND METHODS Complete Freud's Adjuvant (CFA) was used to establish a rat RA model, and JFG or Diclofenac Sodium (Dic) was orally administered. Foot swelling and hematoxylin eosin (H&E) staining were used to test the therapeutic effect of JFG on RA treatment, while ELISA kits were used to detect serum cytokines. Malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), catalase (CAT), and reactive oxygen species (ROS) were used to evaluate oxidative stress levels. The integration of label-free proteomics, fecal short chain fatty acid (SCFA) targeted metabolomics, peripheral blood SCFA, medium and long chain fatty acid targeted metabolomics, and 16S rDNA sequencing of gut microbiota were used to screen the mechanism. Western blot technology was used to validate the results of multiple omics studies. Serum D-Lactic acid, lipopolysaccharide specific IgA antibody (LPS IgA), diamine oxidase (DAO), and colon Claudin 5 and ZO-1 were used to evaluate the intestinal barrier. RESULTS The results confirmed that JFG effectively protected rats from RA injury, which was confirmed by improved foot swelling and synovial pathology. At the same time, JFG reduced the levels of TNF-α, IL-1β, and IL-6 in serum by inhibiting the NLRP3 inflammasome signaling pathway and TLR4/NF-κB signaling pathway in synovial tissue. Multiple omics studies indicated that JFG increased the abundance of gut microbiota and regulated the number of gut bacteria, thereby increased the levels of Acetic acid, Propionic acid, and Butyric acid in the gut and serum of RA rats, which activated AMPK to regulate fatty acid metabolism and fatty acid biosynthesis, thereby inhibited lipid oxidative stress induced ferroptosis to improve tissue damage caused by RA. Meanwhile, JFG improved the intestinal barrier by upregulating the expresses of Claudin 5 and ZO-1, which was confirmed by low concentrations of D-Lactic acid, LPS-SIgA and DAO in serum. CONCLUSIONS This study confirmed that JFG improved the disturbance of fatty acid metabolism by modulating gut microbiota and the production of fecal SCFAs to activate AMPK, and then inhibited ferroptosis caused by lipid oxidative stress in synovium tissue and prevented AR injury. This study proposes for the first time to investigate the mechanism of JFG treatment for RA from the perspective of the "Gut-joint" axis, and provides a promising approach for the treatment of RA.
Collapse
Affiliation(s)
- Xiuwen Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China.
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Dejun Niu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Jidong Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Mengmeng Shen
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Zhen Zeng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Wenqiao Gong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Enhua Yang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Yunfeng Tang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Guoliang Cheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Chenghong Sun
- College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, China.
| |
Collapse
|
37
|
Verma A, Patel P, Almalki WH, Sahebkar A, Kurmi BD, Kesharwani P. Recent Advances in Drug Delivery Approaches for Rheumatoid Arthritis. Curr Med Chem 2025; 32:396-415. [PMID: 37581524 DOI: 10.2174/0929867331666230815112818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/25/2023] [Accepted: 05/23/2023] [Indexed: 08/16/2023]
Abstract
Morbidity, disability, and healthcare expenses associated with rheumatoid arthritis (RA) impose a considerable health and economical burden on both patients and healthcare systems. This review aimed to examine the pathophysiological aspects of RA that may help design different types of drugs and drug delivery systems. These include monoclonal antibodies, immunoglobulins, tiny chemicals, and transgenes for gene therapy. These novel nanocarrier-based therapies target the underlying biological processes involved in RA while minimizing the systemic adverse effects of drugs.
Collapse
Affiliation(s)
- Abhishek Verma
- Department of Quality Assurance, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 24381, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Chennai, India
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
38
|
Rani L, Mathur P, Verma R, Kumar V, Mishra AK, Sahoo PK. Translation Research in Therapeutic Approaches from Conventional to Novel Nano-therapeutics for Rheumatoid Arthritis Treatment. Curr Rheumatol Rev 2025; 21:37-53. [PMID: 38629371 DOI: 10.2174/0115733971288433240408062359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/05/2025]
Abstract
Rheumatoid arthritis is a systemic autoimmune disorder related to joint inflammation, bone erosion, and deformity. Numerous studies indicate that the causes and consequences of RA are still being debated, and therapeutic strategies are in the translation stage. Non-steroidal anti-inflammatory drugs continue to be often used to relieve pain. Still, due to their poor efficacy, failure to halt the spread of the disease, and undesirable adverse effects, they are no longer regarded as first-line treatments. The development of biologic DMRDs designed to reduce the inflammatory response led to substantial changes to the strategy for managing this disease. Although biologic DMRDs have made significant strides in the management of Rheumatoid arthritis, certain patients' lack of response to biological approaches and therapy cessation due to systemic toxicity are unresolved problems. Therefore, to improve the in vivo effect and reduce systemic adverse effects, new approaches are needed to proactively target and transport therapeutic molecules to target sites. The intriguing method of nanotechnology enables the encapsulation of drugs to prevent their deterioration and systemic adverse effects. The next generation of Rheumatoid arthritis therapies might be based on advances in nanomaterial-based drug delivery, Trojan horse, and antibody targeting approaches. This article presents an overview of the advancements in Rheumatoid arthritis therapy, ranging from traditional methods to recent cutting-edge, ongoing pre-clinical and clinical approaches.
Collapse
Affiliation(s)
- Laxmi Rani
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Science and Research, DPSR University, Sector-3, MB Road Pushp Vihar, New Delhi, 110017, India
| | - Pooja Mathur
- Department of Pharmacy, School of Medical and Allied Sciences, G D Goenka University, Sohna, Gurugram, Haryana, 122103, India
| | - Ravinder Verma
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University, Bhiwani, 127021, India
| | - Vivek Kumar
- Institute of Pharmacy, Shri Ram College of Pharmacy, Karnal, India
| | - Ashwini Kumar Mishra
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Science and Research, DPSR University, Sector-3, MB Road Pushp Vihar, New Delhi, 110017, India
| | - Pravat Kumar Sahoo
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Science and Research, DPSR University, Sector-3, MB Road Pushp Vihar, New Delhi, 110017, India
| |
Collapse
|
39
|
Ben-Shabat N, Fisher L, Maixner N, Arow M, Ozeri DJ, Patt YS, Watad A, Amital H, Gendelman O. Arthroplasty rates and risk in familial Mediterranean fever patients: a large population-based study. Rheumatology (Oxford) 2025; 64:352-357. [PMID: 38317059 DOI: 10.1093/rheumatology/keae086] [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: 10/28/2023] [Revised: 12/22/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND FMF is a genetic disorder characterized by recurrent episodes of fever and inflammation in various organs, including the joints. Traditionally, the arthritis of FMF has been considered relatively harmless. However, anecdotal evidence has suggested that it may contribute to long-term joint damage, which may necessitate surgical joint replacement. This study aimed to investigate the rates of arthroplasty among FMF patients and compare it with those of the general population. METHODS The study used the electronic database of the largest health-care organization in Israel to identify 9769 FMF patients diagnosed between 2000 and 2016. A similar number of age-, gender-, and residency-matched controls were also identified. The rates of arthroplasty were compared between the two groups. A logistic regression model predicting the need for arthroplasty within the FMF group was formed to identify potential risk factors. RESULTS Of the 9769 FMF patients, 114 (1.2%) underwent arthroplasty, compared with 64 (0.7%) of the control group [unadjusted odds ratio (OR) = 1.79, 95% CI 1.32-2.43; partially adjusted OR = 1.97, 95% CI 1.40-2.77; fully adjusted OR = 1.92, 95% CI 1.35-2.72]. Within the FMF cohort, those of North African origin had a significantly higher risk of arthroplasty (OR = 6.89, 95% CI 5.09-9.33; P < 0.001). CONCLUSION FMF patients can experience long-term joint damage that may require arthroplasty. Although this complication is relatively uncommon in FMF patients, it occurs almost twice as frequently as compared with the general population. FMF patients of North African origin are at an even higher risk.
Collapse
Affiliation(s)
- Niv Ben-Shabat
- Department of Internal Medicine B, Sheba Medical Center, Tel-Hashomer, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Lior Fisher
- Department of Internal Medicine B, Sheba Medical Center, Tel-Hashomer, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Nitzan Maixner
- Department of Internal Medicine B, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Institute of Oncology, Sheba Medical Center, Tel-Hashomer, Israel
- Cancer Research Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Mohamad Arow
- Department of Internal Medicine B, Sheba Medical Center, Tel-Hashomer, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - David J Ozeri
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yonatan Shneor Patt
- Department of Internal Medicine B, Sheba Medical Center, Tel-Hashomer, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Abdulla Watad
- Department of Internal Medicine B, Sheba Medical Center, Tel-Hashomer, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Institute of Molecular Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
| | - Howard Amital
- Department of Internal Medicine B, Sheba Medical Center, Tel-Hashomer, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Omer Gendelman
- Department of Internal Medicine B, Sheba Medical Center, Tel-Hashomer, Israel
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
40
|
Saini K, Chauhan S, Dar MO, Gupta S, Singh IP, Rawal RK, Gupta N. In Silico and In Vivo Evaluation of Anti-Arthritic Effects of Bakuchiol from Psoralea corylifolia Seeds in Experimental Rat Model. Chem Biodivers 2025; 22:e202401606. [PMID: 39262119 DOI: 10.1002/cbdv.202401606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/13/2024]
Abstract
Rheumatoid arthritis is an autoimmune disease mainly affecting the joints categorized by inflammation, swelling of the synovium and decrease in the joint movement. Bakuchiol, a meroterpene class of natural product present in Psoralea corylifolia known to possess anti-inflammatory effects by a variety of mechanisms. However, its effects in rheumatoid arthritis still remain unclear. In the present investigation, we studied the anti-arthritic effects of bakuchiol via in silico and in vivo experiments. It also showed antioxidant effects measured using DPPH assay where it showed free radical scavenging activity with IC50 value 468.26 μg/ml. Molecular Docking studies carried out on COX-1 (PDB ID: 3 N8Z), COX-2 (PDB ID: 4PH9) and TNF-α (PDB ID: 7JRA), proteins involved in inflammation in arthritis. Bakuchiol showed the maximum binding affinity for TNF-α with binding affinity score is -7.29 kcal/moland less affinity was observed for COX-1 and 2. In vivo antiarthritic effects were studied in arthritic female wistar rats model prepared by intradermal injection of freund's complete adjuvant. Bakuchiol was administered orally at dose of 10,20 and 40 mg/kg for 21 days. Our treatment showed that bakuchiol at 20 and 40 mg/kg exhibited significant anti-inflammatory effects (p<0.001) showed by significant decrease in paw volume, paw diameter, spleen and thymus weight and increase in pain threshold and body weight in arthritic rat model. A significant decrease in hematological parameters such as total leukocyte count (TLC), platelet count, CRP and rheumatoid arthritis factor (RF) and increase in red blood cells count, ESR and hemoglobin further demonstrated that bakuchiol treatment suppresses the progression of adjuvant induced arthritis (AIA) in arthritic rat model. Histological analysis further revealed that bakuchiol ameliorates the pathological manifestations of AIA and reverse the abnormality induced by AIA in rats shown by protection against bone necrosis involved with low influx of inflammatory cells. Therefore, in silico and in vivo results revealed that bakuchiol has the potential to be developed as potent antiarthritic agent.
Collapse
Affiliation(s)
- Kamal Saini
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India, 133207
| | - Samrat Chauhan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India, 140401
| | - Mohammad Ovais Dar
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India, 133207
| | - Sumeet Gupta
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India, 133207
| | - Inder Pal Singh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India, 160062
| | - Ravindra K Rawal
- Natural Product Chemistry Group, Chemical Sciences and Technology Division, CSIR-North East Institute of Science and Techno logy, Jorhat, Assam, 785006, India
| | - Nidhi Gupta
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India, 133207
| |
Collapse
|
41
|
Pimple P, Shah J, Singh P. Emerging Phytochemical Formulations for Management of Rheumatoid Arthritis: A Review. Curr Drug Deliv 2025; 22:15-40. [PMID: 38299275 DOI: 10.2174/0115672018270434240105110330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/02/2023] [Accepted: 12/17/2023] [Indexed: 02/02/2024]
Abstract
Rheumatoid arthritis (RA) is a T-cell-mediated chronic inflammatory disorder affecting 0.5-1% of the global population. The disease with unknown etiology causes slow destruction of joints, advancing to significant deterioration of an individual's quality of life. The present treatment strategy comprises the use of disease-modifying anti-rheumatic drugs (DMARDs) coupled with or without nonsteroidal anti-inflammatory drugs or glucocorticoids. Additionally, involves co-therapy of injectable biological DMARDs in case of persistent or recurrent arthritis. The availability of biological DMARDs and the implementation of the treat-to-target approach have significantly improved the outcomes for patients suffering from RA. Nevertheless, RA requires continuous attention due to inadequate response of patients, development of tolerance and severe side effects associated with long-term use of available treatment regimens. An estimated 60-90% of patients use alternative methods of treatment, such as herbal therapies, for the management of RA symptoms. Over the past few decades, researchers have exploring natural phytochemicals to alleviate RA and associated symptoms. Enormous plant-origin phytochemicals such as alkaloids, flavonoids, steroids, terpenoids and polyphenols have shown anti-inflammatory and immunomodulatory activity against RA. However, phytochemicals have certain limitations, such as high molecular weight, poor water solubility, poor permeability, poor stability and extensive first-pass metabolism, limiting absorption and bioavailability. The use of nanotechnology has aided to extensively improve the pharmacokinetic profile and stability of encapsulated drugs. The current review provides detailed information on the therapeutic potential of phytochemicals. Furthermore, the review focuses on developed phytochemical formulations for RA, with emphasis on clinical trials, regulatory aspects, present challenges, and future prospects.
Collapse
Affiliation(s)
- Prachi Pimple
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Mumbai 400056, India
| | - Jenny Shah
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Mumbai 400056, India
| | - Prabha Singh
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle, Mumbai 400056, India
| |
Collapse
|
42
|
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
|
43
|
Makhija M, Manchanda D, Sharma M. Nano-based Therapeutics for Rheumatoid Arthritis: Recent Patents and Development. RECENT PATENTS ON NANOTECHNOLOGY 2025; 19:56-75. [PMID: 37691226 DOI: 10.2174/1872210518666230905155459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease marked by inflammation of synovium and generation of autoantibodies. Bone and cartilage are frequently damaged along with weakening of tendons and ligaments resulting in disability. An effective RA treatment needs a multi-disciplinary approach which relies upon pathophysiology that is still partially understood. In RA patients, inflammation was induced by pro-inflammatory cytokines including IL-1, IL-6 & IL-10. The conventional dosage regimens for treating RA have drawbacks such as ineffectiveness, greater doses, frequent dosing, relatively expensive and serious adverse effects. To formulate an effective treatment plan for RA, research teams have recently focused on producing several nanoformulations containing anti-inflammatory APIs with an aim to target the inflamed area. Nanomedicines have recently gained popularity in the treatment of RA. Interestingly, unbelievable improvements have been observed in current years in diagnosis and management of RA utilizing nanotechnology. Various patents and clinical trial data have been reported in relevance to RA treatment.
Collapse
Affiliation(s)
- Manish Makhija
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, India
| | - Deeksha Manchanda
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, India
| | - Manu Sharma
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
| |
Collapse
|
44
|
Rajalekshmi R, Agrawal DK. Therapeutic Efficacy of Medicinal Plants with Allopathic Medicine in Musculoskeletal Diseases. INTERNATIONAL JOURNAL OF PLANT, ANIMAL AND ENVIRONMENTAL SCIENCES 2024; 14:104-129. [PMID: 39866300 PMCID: PMC11765655 DOI: 10.26502/ijpaes.4490170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Musculoskeletal diseases encompass a diverse array of disorders affecting the muscles, bones, joints, and connective tissues, leading to significant impairments in mobility, function, and quality of life. Affecting over 1.3 billion individuals globally, musculoskeletal diseases represent a major source of disability and economic burden. Conventional treatment modalities, including pharmacological interventions and surgical procedures, are frequently limited by adverse side effects, prolonged recovery periods, and patient dissatisfaction, particularly when focused solely on symptom management. In response, complementary and alternative medicine, particularly the use of medicinal plants, has garnered increasing interest to enhance the management of musculoskeletal diseases. Medicinal plants possess a wide spectrum of pharmacologically active compounds with anti-inflammatory, analgesic, and antioxidant properties, making them promising adjuncts to conventional therapies. This review critically evaluates the potential synergy between medicinal plants and allopathic medicine for the management of musculoskeletal diseases, with an emphasis on integrated therapy that combines both modalities. Specifically, a critical discussion is presented on how medicinal plants with scientifically supported pharmacological properties can augment the therapeutic efficacy of conventional medications, reduce their doses, and mitigate adverse effects. Furthermore, the challenges associated with incorporating herbal medicine into established healthcare systems are discussed, including the need for rigorous clinical validation, standardization, and regulatory frameworks. Overall, the article underscores the potential of integrated therapeutic approaches to improve clinical outcomes, enhance patient well-being, and establish a more sustainable model for the treatment of musculoskeletal diseases.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
45
|
Maharaj N, Uppada DR, Reddy N, Reddy P, Batalov A, Lvanova D, Staykova N, Baranauskaite A, Hassan LA. Comparing immunogenicity and safety following transition from reference rituximab to biosimilar rituximab (DRL_RI) in patients with rheumatoid arthritis: a randomized, double-blind, phase 3 study. Arthritis Res Ther 2024; 26:225. [PMID: 39709462 DOI: 10.1186/s13075-024-03456-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024] Open
Abstract
OBJECTIVES To assess immunogenicity and safety in patients with active rheumatoid arthritis (RA) transitioning from rituximab [US-licensed rituximab: Reference Product (RP); EU-approved rituximab: Reference Medicinal Product (RMP)] to DRL_RI (proposed rituximab biosimilar), in comparison to those continuing on RP/RMP. METHODS This double-blind, randomized, Phase 3 study included 140 RA patients having prior exposure to RP/RMP; transitioned to DRL_RI (n = 70) or continued with RP/RMP (n = 70) for two 1000 mg infusions on Days 1 and 15. Assessments included Time-matched Rituximab Concentration (TMRC), anti-drug antibodies (ADAs), neutralizing antibodies (NAbs) and ADA titre over 12 weeks, and safety follow-up till 26 weeks. RESULTS The mean age of subjects was 59.8 years (range: 24, 86) and the mean BMI was 27.76 kg/m2 (range: 17.5, 52.0). Incidence of ADA after dosing was low in both groups: 1.4% in DRL_RI group on Day 15, Week 8, and Week 12; and 2.9% in RP/RMP group at Week 12. Only 1 patient in DRL_RI group was positive for NAbs at Week 8. ADA titre values did not significantly differ between the two groups. The time-matched rituximab concentration was comparable between groups, indicating no interference for immunogenicity assessment. Treatment-emergent adverse events (TEAEs) were reported by 34.3% and 38.6% patients, respectively, in DRL_RI and RP/RMP groups. Incidences of TEAEs that were drug-related, leading to treatment discontinuation, grade ≥ 3, or serious, were also comparable. CONCLUSION Immunogenicity was low and comparable in RA patients transitioning to DRL_RI or continuing on RP/RMP. The overall safety profile in patients transitioning to DRL_RI did not appear to differ in frequency, severity, or quality from patients continuing on RP/RMP and was in line with the known safety profile of rituximab. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT0426877 EudraCT:2019-002810-37 US IND 112766.
Collapse
Affiliation(s)
- Narendra Maharaj
- Clinical Development - Biologics, Dr. Reddy's Laboratories Ltd., Bachupally, Hyderabad, 500090, India.
| | - Dharma Rao Uppada
- Clinical Development - Biologics, Dr. Reddy's Laboratories Ltd., Bachupally, Hyderabad, 500090, India
| | - Naveen Reddy
- Clinical Development - Biologics, Dr. Reddy's Laboratories Ltd., Bachupally, Hyderabad, 500090, India
| | - Pramod Reddy
- Clinical Development - Biologics, Dr. Reddy's Laboratories Ltd., Bachupally, Hyderabad, 500090, India
| | - Anastas Batalov
- Medical Faculty, Medical University of Plovdiv, University Hospital "Kaspela", Clinic of Rheumatology, Plovdiv, 4000, Bulgaria
| | - Delina Lvanova
- Diagnostic and Consulting Center Aleksandrovska EOOD, Sofia, 1431, Bulgaria
| | - Nedyalka Staykova
- Outpatient Clinic for Specialized Medical Help - Medical Center Kuchuk Paris OOD, Plovdiv, 4004, Bulgaria
| | - Asta Baranauskaite
- Lithuanian University of Health Sciences Kaunas, Kaunas, LT-50161, Lithuania
| | | |
Collapse
|
46
|
Pareek A, Mehlawat K, Tripathi K, Pareek A, Chaudhary S, Ratan Y, Apostolopoulos V, Chuturgoon A. Melittin as a therapeutic agent for rheumatoid arthritis: mechanistic insights, advanced delivery systems, and future perspectives. Front Immunol 2024; 15:1510693. [PMID: 39759520 PMCID: PMC11695321 DOI: 10.3389/fimmu.2024.1510693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Rheumatoid arthritis (RA), a condition characterized by joint deterioration through the action of matrix metalloproteinases (MMPs), is prevalent worldwide. Bee venom (BV) has traditionally been used in Chinese medicine for pain, arthritis, rheumatism, skin diseases, etc. BV is enriched with active substances, notably melittin and phospholipase A2 (PLA2), offering significant therapeutic potential. Hence, the review summarizes current insights into BV's composition, antiarthritic mechanism and pharmacological benefits, focusing on melittin. Constituting 50-60% of BV, melittin notably downregulates nuclear factor Kappa B (NF-κB) activity, inhibits MMP-1 and MMP-8, and diminishes tumor necrosis factor (TNF-α), all of which contribute to the mitigation of type 2 collagen degradation. Despite its potential, melittin exhibits hemolytic activity and can significantly affect cell membranes, limiting its application, which poses a challenge to its therapeutic use. To overcome these challenges, delivery techniques utilizing nanocarriers and modifications in amino acid sequencing have been developed. Recent advancements in delivery systems, including nanocarriers, transdermal patches, and nanoemulsions, aim to minimize toxicity, expanding its therapeutic utility for RA. This article explores these novel strategies, underlining the evolving role of melittin in RA management.
Collapse
Affiliation(s)
- Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | | | | | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | | | - Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Vasso Apostolopoulos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Anil Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
47
|
Hong X, Jiang F. Association Between Dietary Niacin Intake and Rheumatoid Arthritis in American Women: A Study Based on National Health and Nutrition Examination Survey Database. Int J Womens Health 2024; 16:2209-2219. [PMID: 39717391 PMCID: PMC11665156 DOI: 10.2147/ijwh.s482294] [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: 06/11/2024] [Accepted: 11/30/2024] [Indexed: 12/25/2024] Open
Abstract
Objective This study aimed to explore the association between dietary niacin intake and rheumatoid arthritis (RA) in American women through the National Health and Nutrition Examination Survey (NHANES) database. Methods A retrospective analysis was conducted based on NHANES 2003-2016 data. Dietary niacin intake was stratified using weighted quartiles and association of dietary niacin intake with RA was explored using weighted logistic regression models and restricted cubic splines (RCS). Subgroup analysis was conducted, adjusting for all confounding factors, and a likelihood ratio test was utilized to determine significant covariates for the interaction term. Stratified analysis was conducted on significant covariates to determine their impact on the association of dietary niacin intake with RA. Results Fourteen thousand five hundred and thirty-nine American women were selected according to inclusion and exclusion criteria, among whom 845 (4.4%) had RA. Compared with American women without RA, American women with RA had significantly lower dietary niacin intake (18.90 vs 21.22, P<0.001). Logistic regression models and RCS analysis reported a significant linear negative correlation between dietary niacin intake and prevalence of RA (Odds Ratio (OR) < 1, P < 0.05, P-non-linear >0.05). The interaction-term P-values showed that this association was significantly influenced by poverty income ratio (PIR), education level, Body Mass Index (BMI), and smoking (P for interaction < 0.05). Stratified analysis unveiled that this association was particularly significant in individuals aged ≥ 40 years (OR: 0.98, 95% Confidence Interval (CI): 0.97-0.99, P < 0.05), PIR > 3.5 (OR: 0.96, 95% CI: 0.93-0.99, P < 0.05), with a college education or higher (OR: 0.97, 95% CI: 0.94-0.99, P < 0.01), BMI ≥ 30kg/m² (OR: 0.98, 95% CI: 0.96-0.99, P < 0.05), non-smokers (OR: 0.97, 95% CI: 0.95-0.99, P < 0.01), or former smokers (OR: 0.95, 95% CI: 0.95-0.99, P < 0.05). Conclusion Increased dietary niacin intake was associated with a reduced prevalence of RA, especially in women aged ≥40, PIR > 3.5, with at least a college education, BMI ≥ 30kg/m², and currently non-smokers.
Collapse
Affiliation(s)
- Xuelian Hong
- Department of Rheumatology and Immunology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua City, Zhejiang Province, 321000, People’s Republic of China
| | - Fengfeng Jiang
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua City, Zhejiang Province, 321000, People’s Republic of China
| |
Collapse
|
48
|
Dixit T, Vaidya A, Ravindran S. Therapeutic potential of antibody-drug conjugates possessing bifunctional anti-inflammatory action in the pathogenies of rheumatoid arthritis. Arthritis Res Ther 2024; 26:216. [PMID: 39695738 PMCID: PMC11656801 DOI: 10.1186/s13075-024-03452-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
In an age where there is a remarkable upsurge in developing precision medicines, antibody-drug conjugates (ADCs) have emerged as a progressive therapeutic strategy. ADCs typically consist of monoclonal antibodies (mAb) conjugated to the cytotoxic payloads by utilizing a linker, combining the benefits of definitive target specificity of mAbs and potent killing impact of payload to achieve precise and efficient elimination of target cells. In addition to their well-established role in oncology, ADCs are currently demonstrating encouraging potential in addressing the unmet requirements in the treatment of autoimmune conditions such as rheumatoid arthritis (RA). Prevalent long-term autoimmune disease RA costs billions of dollars annually but still, there is a lack of precision-targeted therapeutics with minimal side effects. This review provides an overview of the RA pathogenesis, pre-existing therapies, and their limitations, the introduction of ADCs in RA treatment, the mechanism of ADCs, and a summary of ADCs in preclinical and clinical trials. Based on the literature we also propose a strategy in ADC synthesis, which may increase the efficiency in targeting multifactorial diseases like RA. We propose to utilize DMARDs (Disease-modifying anti-rheumatic drugs), the first-line treatment for RA, as a payload for ADC synthesis. DMARDs are the only class of medication that limits the disease progression, but their efficacy is limited due to off-target toxicities. Hence, utilizing them as payload will help to deliver them directly at the targeted site, reducing their off-target toxicity, which in turn will increase their efficiency in targeting disease. Also, as mAbs are not sufficient to achieve remission, they are given in combinations with DMARDs. Hence, synthesizing ADCs may reduce the multiple and higher dosages given to patients, which in turn may enhance patient compliance.
Collapse
Affiliation(s)
- Tanu Dixit
- Symbiosis School of Biological Sciences (SSBS), Faculty of Medical & Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, 412115, India
| | - Anuradha Vaidya
- Symbiosis School of Biological Sciences (SSBS), Faculty of Medical & Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, 412115, India
| | - Selvan Ravindran
- Symbiosis School of Biological Sciences (SSBS), Faculty of Medical & Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, 412115, India.
| |
Collapse
|
49
|
Mathew S, Ashraf S, Shorter S, Tozzi G, Koutsikou S, Ovsepian SV. Neurobiological Correlates of Rheumatoid Arthritis and Osteoarthritis: Remodelling and Plasticity of Nociceptive and Autonomic Innervations in Synovial Joints. Neuroscientist 2024:10738584241293049. [PMID: 39668598 DOI: 10.1177/10738584241293049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Swelling, stiffness, and pain in synovial joints are primary hallmarks of osteoarthritis and rheumatoid arthritis. Hyperactivity of nociceptors and excessive release of inflammatory factors and pain mediators play a crucial role, with emerging data suggesting extensive remodelling and plasticity of joint innervations. Herein, we review structural, functional, and molecular alterations in sensory and autonomic axons wiring arthritic joints and revisit mechanisms implicated in the sensitization of nociceptors, leading to chronic pain. Sprouting and reorganization of sensory and autonomic fibers with the invasion of ectopic branches into surrounding inflamed tissues are associated with the upregulation of pain markers. These changes are frequently complemented by a phenotypic switch of sensory and autonomic profiles and activation of silent axons, inferring homeostatic adjustments and reprogramming of innervations. Identifying critical molecular players and neurobiological mechanisms underpinning the rewiring and sensitization of joints is likely to elucidate causatives of neuroinflammation and chronic pain, assisting in finding new therapeutic targets and opportunities for interventions.
Collapse
Affiliation(s)
- Sharon Mathew
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
| | - Sadaf Ashraf
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| | - Susan Shorter
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
| | - Gianluca Tozzi
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
| | - Stella Koutsikou
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| | - Saak V Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
- Faculty of Medicine, Tbilisi State University, Tbilisi, Republic of Georgia
| |
Collapse
|
50
|
Karnam S, Jindal AB, Paul AT. Quality by design-based optimization of teriflunomide and quercetin combinational topical transferosomes for the treatment of rheumatoid arthritis. Int J Pharm 2024; 666:124829. [PMID: 39406305 DOI: 10.1016/j.ijpharm.2024.124829] [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: 06/24/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
Rheumatoid arthritis (RA) is an immune-mediated inflammatory disease. Combination therapy is anticipated to surpass monotherapy by targeting multiple pathways involved in RA progression. The present aim is to develop a combination of Teriflunomide (TFD) and Quercetin (QCN) loaded transferosomal gel to enhance permeability and achieve localized delivery to joint tissues. TFD or QCN transferosomes were optimized employing a 3-level, 3-factorial design Box-Behnken design (BBD). The transferosomes exhibited sustained in-vitro drug release. The topical combination gel underwent thorough evaluation of rheology, and also ex-vivo studies showed enhanced permeability through rat skin. The synergistic combination of TFD and QCN effectively suppressed NO, TNF-α and IL-6 levels in in-vitro RAW 264.7 cells. The cytotoxicity in HaCaT cell lines indicates non-toxicity of the gel, further confirmed by skin irritation study conducted in rats. The in-vivo anti-arthritic activity was evaluated in complete freund's adjuvant induced rat paw edema model illustrates the effectiveness of the combination transferosomal gel compared to other treatment groups. In conclusion, the topical delivery of TFD and QCN combination transferosomal gel demonstrated anti-arthritic activity through localized delivery whichallows for dose reduction, thereby may reduce the systemic drug exposure and mitigate the side effects associated with oral administration of TFD.
Collapse
Affiliation(s)
- Sriravali Karnam
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Pilani Campus, Rajasthan 333031, India
| | - Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Pilani Campus, Rajasthan 333031, India
| | - Atish T Paul
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|