1
|
Nayak D, Shetty MM, Halagali P, Rathnanand M, Gopinathan A, John J, Krishna Tippavajhala V. Formulation, optimization and evaluation of ibuprofen loaded menthosomes for transdermal delivery. Int J Pharm 2024; 665:124671. [PMID: 39245088 DOI: 10.1016/j.ijpharm.2024.124671] [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: 06/10/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
The study aimed to improve the transdermal permeation of IBU utilizing menthosomes as a vesicular carrier. IBU-loaded menthosomes were formulated by thin film hydration & optimized using 23 factorial designs (Design Expert® version 13 software). In vitro & ex vivo skin permeation analysis of IBU-encapsulated menthosomes was studied across the rat skin sample. In vivo pharmacodynamic activity was studied in an arthritis rat model. The optimized IBU-loaded menthosomes exhibited an optimum vesicle size of 214.2 ± 2.96 nm, Zeta potential of -21.1 ± 2.72 mV, (PDI) Polydispersity Index of 0.267 ± 0.018 with Entrapment efficiency (EE%) of 78.7 ± 2.73 %. The in vitro & ex vivo skin penetration study displayed enhanced release of drug of 77.02 ± 1.0 % and 40.91 ± 0.81 % respectively, compared to conventional liposomes. In vivo pharmacodynamic study on carrageenan-induced paw edema in Wistar albino rats demonstrated superior anti-inflammatory activity of the optimized IBU-encapsulated menthosomes (**p < 0.01) and effective inhibition of paw edema (34.04 ± 0.155 %). The formalin test indicated a significant analgesic effect of optimized formulation during the chronic phase of analgesia (*p < 0.05) compared to the control group. Thus, the developed and optimized drug-loaded menthosomes could serve as a suitable vesicular delivery carrier in enhancing the transdermal delivery of other NSAID drugs.
Collapse
Affiliation(s)
- Devika Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manisha M Shetty
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Praveen Halagali
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mahalaxmi Rathnanand
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Adarsh Gopinathan
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Jeena John
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Vamshi Krishna Tippavajhala
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
2
|
Xu M, Fu T, Zhang C, An Z, Yan J, Lu Z, Wu H, Liu J, Qiu L, Shi L, Lin J, Cao Y, Pei R. Prolonged, staged, and self-regulated methotrexate release coupled with ROS scavenging in an injectable hydrogel for rheumatoid arthritis therapy. J Control Release 2024; 375:60-73. [PMID: 39216600 DOI: 10.1016/j.jconrel.2024.08.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Rheumatoid arthritis (RA) remains a formidable healthcare challenge due to its chronic nature and potential for irreversible joint damage. Methotrexate (MTX) is a cornerstone treatment for RA but carries significant risks of adverse effects with repeated administration, necessitating the exploration of alternative delivery methods. Injectable hydrogels loaded with MTX for intra-articular injection present a promising solution, allowing sustained drug release directly into affected joints. However, current hydrogel systems often lack extended therapeutic periods and the ability to self-regulate drug release according to disease state. Furthermore, RA is associated with excessive production of reactive oxygen species (ROS), which exacerbates inflammation and joint damage. Herein, we developed an advanced injectable hydrogel (MPDANPs/MTX HA-PEG gel) based on "bio-orthogonal chemistry", combining hyaluronic acid and polyethylene glycol (PEG) matrices co-loaded with mesoporous polydopamine nanoparticles (MPDANPs) and MTX. MPDANPs/MTX HA-PEG gel achieved prolonged, staged, and self-regulated MTX release, coupled with ROS scavenging capabilities for enhanced therapeutic efficacy. Due to its optimized MTX release behavior and significant ROS scavenging function, MPDANPs/MTX HA-PEG gel exhibited potent anti-inflammatory effects in collagen-induced arthritis (CIA) rats following a single intra-articular injection. Our findings highlight the potential of MPDANPs/MTX HA-PEG gel as a highly effective treatment strategy for RA, offering a promising avenue for improving patient outcomes.
Collapse
Affiliation(s)
- Mingsheng Xu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Tingting Fu
- Department of Orthopaedics, The Fourth Affiliated of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215001, China
| | - Chenhui Zhang
- Department of Orthopaedics, The Fourth Affiliated of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215001, China
| | - Zhen An
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jincong Yan
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhongzhong Lu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Hanfei Wu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jihuan Liu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Lei Qiu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Lei Shi
- Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan 215300, China
| | - Jun Lin
- Department of Orthopaedics, The Fourth Affiliated of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215001, China.
| | - Yi Cao
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; Jiangxi Institute of Nanotechnology, Nanchang 330200, China.
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| |
Collapse
|
3
|
Zhao X, Wu S, Ni S, Zhong Y, Qin X, Zhang K, Qu K, Zhu L, Wu W. Tannic Acid-Based Biomimetic Nanomedicine with Pathological Reactive Oxygen Species-Responsive Cargo Release for Relieving Inflammation in Rheumatoid Arthritis. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39448903 DOI: 10.1021/acsami.4c11494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic disease characterized by immune cell infiltration and cartilage damage. The local lesion of RA shows severe oxidative stress and proinflammatory cytokine secretion. For drug therapy, the efficacy of agents, such as methotrexate (MTX), may be greatly limited, resulting from the low bioavailability, immune clearance, and toxic side effects. A nanocarrier (TA-PBA NPs) was developed with anti-inflammatory and antioxidant activities, combined with MTX to prepare nanomedicine (MTX NPs) for synergistic treatment of RA. Moreover, inspired by the biological functions homing to inflammation lesion of macrophages, the biomimetic nanomedicine camouflaged with macrophage membrane (MM@MTX NPs) was constructed. TA-PBA NPs could timely promote MTX release in response to the overaccumulated ROS to exhibit high anti-inflammatory and antioxidant activities for alleviating RA progression. The experimental results confirmed that MM@MTX NPs could significantly reduce the secretion of proinflammatory cytokines (TNF-α) while significantly increasing the typical anti-inflammatory cytokines (IL-10), promote the phenotype transformation of macrophages from M1 to M2, and up-regulate the Nrf2-keap1 pathway-related proteins (HO-1 and NRF2) to positively regulate the local inflammation for effectively inhibiting RA development. Thus, MM@MTX NPs represent a possible candidate as a safe and efficient nanotherapy platform for RA management.
Collapse
Affiliation(s)
- Xiong Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Shuai Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Sheng Ni
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yuan Zhong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xian Qin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Kun Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Kai Qu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Li Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wei Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
- JinFeng Laboratory, Chongqing 401329, China
| |
Collapse
|
4
|
Shen S, Wan A, Wang Y, Liu L, Yao Y, Weng J, Zhu T, Yang Q, Yan Q. Flexible microneedles incorporating gold nanorods and tacrolimus for effective synergistic photothermal-chemotherapy of rheumatoid arthritis. Int J Biol Macromol 2024; 276:133797. [PMID: 38992523 DOI: 10.1016/j.ijbiomac.2024.133797] [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: 11/16/2023] [Revised: 11/24/2023] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Transdermal drug delivery systems for rheumatoid arthritis (RA) have garnered substantial attention due to their great potential to overcome limitations observed in conventional oral or injection strategies, including limited selectivity and adverse effects on extra-articular tissues. Microneedles (MNs) appear to be highly desirable carriers for transdermal drug delivery of RA. However, microneedles typically are unable to keep up with the flexibility of joints, which decreases the effectiveness of administration. In this study, we developed a flexible microneedles (FMNs) delivery system. And gelatin was employed for the fabrication of flexible backings for microneedles owing to its excellent ductility and biocompatibility. We achieved synergisticphotothermal-chemotherapy of RA by incorporating the chemical drug Tacrolimus (TAC) and the photothermal agent gold nanorods (AuNRs) into dissolving microneedles. Results showed a high mechanical strength of the proposed FMNs. In the animal model of adjuvant-induced arthritis (AA), it is indicated that the prepared FMNs inhibited the expression of related inflammatory cytokines such as IL-1ß and TNF-α while enhancing bone repair and other related factors. Thus, the combination therapy of FMNs-mediated hyperthermia and chemotherapy can serve as a novel and synergistic treatment option for RA.
Collapse
Affiliation(s)
- Shulin Shen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Aiqun Wan
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Yan Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Linxiao Liu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Yao Yao
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Jiaqi Weng
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China; Institute of Laser Advanced Manufacturing, Zhejiang University of Technology, Hangzhou, Zhejiang 310023, PR China; College of Mechanical Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310023, PR China
| | - Tong Zhu
- School of Education and English, Faculty of Humanities and Social Sciences, University of Nottingham, Ningbo, Zhejiang 315199, PR China
| | - Qingliang Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Qinying Yan
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, PR China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, PR China.
| |
Collapse
|
5
|
Tang B, Xie X, Lu J, Huang W, Yang J, Tian J, Lei L. Designing biomaterials for the treatment of autoimmune diseases. APPLIED MATERIALS TODAY 2024; 39:102278. [DOI: 10.1016/j.apmt.2024.102278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
6
|
Ren S, Xu Y, Dong X, Mu Q, Chen X, Yu Y, Su G. Nanotechnology-empowered combination therapy for rheumatoid arthritis: principles, strategies, and challenges. J Nanobiotechnology 2024; 22:431. [PMID: 39034407 PMCID: PMC11265020 DOI: 10.1186/s12951-024-02670-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with multifactorial etiology and intricate pathogenesis. In RA, repeated monotherapy is frequently associated with inadequate efficacy, drug resistance, and severe side effects. Therefore, a shift has occurred in clinical practice toward combination therapy. However, conventional combination therapy encounters several hindrances, including low selectivity to arthritic joints, short half-lives, and varying pharmacokinetics among coupled drugs. Emerging nanotechnology offers an incomparable opportunity for developing advanced combination therapy against RA. First, it allows for co-delivering multiple drugs with augmented physicochemical properties, targeted delivery capabilities, and controlled release profiles. Second, it enables therapeutic nanomaterials development, thereby expanding combination regimens to include multifunctional nanomedicines. Lastly, it facilitates the construction of all-in-one nanoplatforms assembled with multiple modalities, such as phototherapy, sonodynamic therapy, and imaging. Thus, nanotechnology offers a promising solution to the current bottleneck in both RA treatment and diagnosis. This review summarizes the rationale, advantages, and recent advances in nano-empowered combination therapy for RA. It also discusses safety considerations, drug-drug interactions, and the potential for clinical translation. Additionally, it provides design tips and an outlook on future developments in nano-empowered combination therapy. The objective of this review is to achieve a comprehensive understanding of the mechanisms underlying combination therapy for RA and unlock the maximum potential of nanotechnology, thereby facilitating the smooth transition of research findings from the laboratory to clinical practice.
Collapse
Affiliation(s)
- Shujing Ren
- Department of Pharmacy, Affiliated Hospital 2 of Nantong University, Nantong, 226000, PR China
| | - Yuhang Xu
- School of Pharmacy, Nantong University, Nantong, 226000, PR China
| | - Xingpeng Dong
- School of Pharmacy, Nantong University, Nantong, 226000, PR China
| | - Qingxin Mu
- Department of Pharmaceutics, University of Washington, Seattle, WA, 98195, USA
| | - Xia Chen
- Department of Pharmacy, Affiliated Hospital 2 of Nantong University, Nantong, 226000, PR China.
| | - Yanyan Yu
- School of Pharmacy, Nantong University, Nantong, 226000, PR China.
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong, 226000, PR China.
| |
Collapse
|
7
|
Hu Y, Liu J, Qi Y, Zhou Q, Li Y, Cong C, Chen Y. Integrating clinical data mining, network analysis and experimental validation reveal the anti-inflammatory mechanism of Huangqin Qingre Chubi Capsule in rheumatoid arthritis treatment. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118077. [PMID: 38556141 DOI: 10.1016/j.jep.2024.118077] [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: 01/16/2024] [Revised: 03/06/2024] [Accepted: 03/18/2024] [Indexed: 04/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqin Qingre Chubi Capsule (HQC) is a Chinese medicinal compound used for the treatment of damp-heat pattern rheumatism, guided by the traditional Chinese medicine syndrome differentiation practice. HQC has been used in the clinical treatment of rheumatic diseases for more than 20 years with remarkable efficacy. HQC has been experimentally shown to exert anti-arthritic effects via the Wnt signaling pathway. AIM OF THE STUDY This study used clinical data mining, network analysis, and in vitro and in vivo tests to investigate the anti-arthritic and possible anti-inflammatory mechanism of HQC. Specifically, emphasis was placed on the function of the hsa_circ_0091,685/EIF4A3/IL-17 axis in the anti-inflammatory process. MATERIALS AND METHODS A random walk model was used to evaluate the effects of HQC on clinical immune inflammatory marker function in patients with RA. Network analysis was used to predict the potential target genes and pathways of HQC. Hematoxylin & eosin, safranin O-fast green and toluidine blue staining, immunohistochemistry, and transmission electron microscopy were performed to evaluate the anti-arthritic effects of HQC in rat models. Cell Counting Kit-8 assay, quantitative real-time polymerase chain reaction, western blotting, enzyme-linked immunosorbent assay, and RNA pull-down were used to study the anti-proliferation and anti-inflammatory mechanisms of HQC. RESULTS Patients with RA who underwent HQC treatment showed a significant reduction in inflammatory response levels, according to retrospective clinical study. Network analysis revealed that HQC potentially targeted genes and pathways related to inflammation, especially IL-6, IL-17, TNF-α, IL-23, and IL-17 signaling pathway. Animal experiments showed that HQC inhibits inflammation through the IL-17 signaling pathway in rat models. Cellular experiments showed that HQC-containing serum inhibited the inflammatory response in patients with RA-FLS or RA by blocking hsa_circ_0091,685 and EIF4A3 expression. CONCLUSION In RA patients, HQC reduces the inflammatory response. The antiproliferative and anti-inflammatory qualities of HQC are responsible for its therapeutic impact. The suppression of the hsa_circ_0091,685/EIF4A3/IL-17 axis was linked to these favorable outcomes.
Collapse
Affiliation(s)
- Yuedi Hu
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, No.117, Meishan Road, Hefei, Anhui, China; College of Chinese Medicine, Anhui University of Traditional Chinese Medicine, No.350, Longzihu Road, Hefei, Anhui, China.
| | - Jian Liu
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, No.117, Meishan Road, Hefei, Anhui, China; Institute of Rheumatology, Anhui University of Traditional Chinese Medicine, No.350, Longzihu Road, Hefei, Anhui, China.
| | - Yajun Qi
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, No.117, Meishan Road, Hefei, Anhui, China.
| | - Qiao Zhou
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, No.117, Meishan Road, Hefei, Anhui, China.
| | - Yang Li
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, No.117, Meishan Road, Hefei, Anhui, China.
| | - Chengzhi Cong
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, No.117, Meishan Road, Hefei, Anhui, China.
| | - Yiming Chen
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, No.117, Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
8
|
Ban Z, Li Z, Xing S, Ye Y. IGF2BP3 regulates the expression of RRM2 and promotes the progression of rheumatoid arthritis via RRM2/Akt/MMP-9 pathway. PLoS One 2024; 19:e0303593. [PMID: 38820515 PMCID: PMC11142689 DOI: 10.1371/journal.pone.0303593] [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] [Received: 02/14/2024] [Accepted: 04/28/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a common inflammatory and autoimmune disease. Ribonucleotide Reductase Regulatory Subunit M2 (RRM2) is a crucial and a rate-limiting enzyme responsible for deoxynucleotide triphosphate(dNTP) production. We have found a high expression level of RRM2 in patients with RA, but the molecular mechanism of its action remains unclear. METHODS We analyzed the expression of hub genes in RA using GSE77298 datasets downloaded from Gene Expression Omnibus database. RRM2 and insulin-like growth factor-2 messenger ribonucleic acid (mRNA)-binding protein 3 (IGF2BP3) gene knockdown was achieved by infection with lentiviruses. The expression of RRM2, IGF2BP3, matrix metalloproteinase (MMP)-1, and MMP-9 were detected via western blotting assay. Cell viability was detected via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. MeRIP-qRT-PCR was performed to test the interaction of IGF2BP3 and RRM2 mRNA via m6A modification. Cell proliferation was determined by clone formation assay. Migration and invasion assays were performed using transwell Boyden chamber. RESULTS RRM2 and IGF2BP3 were highly expressed in clinical specimens and tumor necrosis factor alpha (TNF-α) and interleukin (IL)-1β-stimulated synovial cells. RRM2 and IGF2BP3 knockdown inhibited the proliferation, migration, and invasion of MH7A cells. The inhibitory effects of IGF2BP3 knockdown were effectively reversed by simultaneously overexpressing RRM2 in MH7A cells. By analyzing N6-methyladenosine (m6A)2Target database, five m6A regulatory target binding sites for IGF2BP3 were identified in RRM2 mRNA, suggesting a direct relationship between IGF2BP3 and RRM2 mRNA. Additionally, in RRM2 small hairpin (sh)RNA lentivirus-infected cells, the levels of phosphorylated Akt and MMP-9 were significantly decreased compared with control shRNA lentivirus-infected cells. CONCLUSION The present study demonstrated that RRM2 promoted the Akt phosphorylation leading to high expression of MMP-9 to promote the migration and invasive capacities of MH7A cells. Overall, IGF2BP promotes the expression of RRM2, and regulates the migration and invasion of MH7A cells via Akt/MMP-9 pathway to promote RA progression.
Collapse
Affiliation(s)
- Zhaonan Ban
- Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, 611130, China
| | - Zhengjiang Li
- Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, 611130, China
| | - Shuxing Xing
- Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, 611130, China
| | - Yongjie Ye
- Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, 611130, China
| |
Collapse
|
9
|
Aljabali AAA, Obeid MA, Gammoh O, El-Tanani M, Mishra V, Mishra Y, Kapre S, Srivatsa Palakurthi S, Hassan SS, Nawn D, Lundstrom K, Hromić-Jahjefendić A, Serrano-Aroca Á, Redwan EM, Uversky VN, Tambuwala MM. Nanomaterial-Driven Precision Immunomodulation: A New Paradigm in Therapeutic Interventions. Cancers (Basel) 2024; 16:2030. [PMID: 38893150 PMCID: PMC11171400 DOI: 10.3390/cancers16112030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Immunotherapy is a rapidly advancing field of research in the treatment of conditions such as cancer and autoimmunity. Nanomaterials can be designed for immune system manipulation, with precise targeted delivery and improved immunomodulatory efficacy. Here, we elaborate on various strategies using nanomaterials, including liposomes, polymers, and inorganic NPs, and discuss their detailed design intricacies, mechanisms, and applications, including the current regulatory issues. This type of nanomaterial design for targeting specific immune cells or tissues and controlling release kinetics could push current technological frontiers and provide new and innovative solutions for immune-related disorders and diseases without off-target effects. These materials enable targeted interactions with immune cells, thereby enhancing the effectiveness of checkpoint inhibitors, cancer vaccines, and adoptive cell therapies. Moreover, they allow for fine-tuning of immune responses while minimizing side effects. At the intersection of nanotechnology and immunology, nanomaterial-based platforms have immense potential to revolutionize patient-centered immunotherapy and reshape disease management. By prioritizing safety, customization, and compliance with regulatory standards, these systems can make significant contributions to precision medicine, thereby significantly impacting the healthcare landscape.
Collapse
Affiliation(s)
- Alaa A. A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan; (A.A.A.A.); (M.A.O.)
| | - Mohammad A. Obeid
- Faculty of Pharmacy, Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan; (A.A.A.A.); (M.A.O.)
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Sumedha Kapre
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.K.); (S.S.P.)
| | - Sushesh Srivatsa Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.K.); (S.S.P.)
| | - Sk. Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram, Paschim Medinipur 721140, West Bengal, India;
| | - Debaleena Nawn
- Indian Research Institute for Integrated Medicine (IRIIM), Unsani, Howrah 711302, West Bengal, India;
| | | | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| | - Elrashdy M. Redwan
- Department of Biological Science, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia;
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21934, Egypt
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Murtaza M. Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| |
Collapse
|
10
|
Zakeri Z, Heiderzadeh M, Kocaarslan A, Metin E, Hosseini Karimi SN, Saghati S, Vural A, Akyoldaş G, Baysal K, Yağcı Y, Gürsoy-Özdemir Y, Taşoğlu S, Rahbarghazi R, Sokullu E. Exosomes encapsulated in hydrogels for effective central nervous system drug delivery. Biomater Sci 2024; 12:2561-2578. [PMID: 38602364 DOI: 10.1039/d3bm01055d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
The targeted delivery of pharmacologically active molecules, metabolites, and growth factors to the brain parenchyma has become one of the major challenges following the onset of neurodegeneration and pathological conditions. The therapeutic effect of active biomolecules is significantly impaired after systemic administration in the central nervous system (CNS) because of the blood-brain barrier (BBB). Therefore, the development of novel therapeutic approaches capable of overcoming these limitations is under discussion. Exosomes (Exo) are nano-sized vesicles of endosomal origin that have a high distribution rate in biofluids. Recent advances have introduced Exo as naturally suitable bio-shuttles for the delivery of neurotrophic factors to the brain parenchyma. In recent years, many researchers have attempted to regulate the delivery of Exo to target sites while reducing their removal from circulation. The encapsulation of Exo in natural and synthetic hydrogels offers a valuable strategy to address the limitations of Exo, maintaining their integrity and controlling their release at a desired site. Herein, we highlight the current and novel approaches related to the application of hydrogels for the encapsulation of Exo in the field of CNS tissue engineering.
Collapse
Affiliation(s)
- Ziba Zakeri
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
| | - Morteza Heiderzadeh
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
| | - Azra Kocaarslan
- Chemistry Department, Faculty of Science, İstanbul Technical University, İstanbul, Turkey
| | - Ecem Metin
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
| | | | - Sepideh Saghati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atay Vural
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Department of Neurology, School of Medicine, KoÒ« University, Istanbul 34450, Turkey
| | - Göktuğ Akyoldaş
- Department of Neurosurgery, Koç University Hospital, Istanbul 34450, Turkey
| | - Kemal Baysal
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Department of Biochemistry, School of Medicine, Koç University, Istanbul 34450, Turkey
| | - Yusuf Yağcı
- Chemistry Department, Faculty of Science, İstanbul Technical University, İstanbul, Turkey
| | - Yasemin Gürsoy-Özdemir
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Department of Neurology, School of Medicine, KoÒ« University, Istanbul 34450, Turkey
| | - Savaş Taşoğlu
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Mechanical Engineering Department, School of Engineering, Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Emel Sokullu
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Biophysics Department, Koç University School of Medicine, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey
| |
Collapse
|
11
|
Dai R, Zhao M, Zheng X, Li D, Kang W, Hao H, Chen X, Jin Y, Li J, Liu Q, Zheng Z, Zhang R. Homology-Activated Ultrasensitive Nanomedicines for Precise NIR-II FL/MRI Imaging-Guided "Knock-On" Dynamic Therapy in Rheumatoid Arthritis. Adv Healthc Mater 2024; 13:e2303892. [PMID: 38219028 DOI: 10.1002/adhm.202303892] [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: 11/07/2023] [Revised: 12/25/2023] [Indexed: 01/15/2024]
Abstract
Stimuli-responsive nanomedicines represent a pivotal technology for in situ on-demand drug release and offer multiple advantages over conventional drug delivery systems to combat rheumatoid arthritis(RA). However, the lack of sensitivity to a single-stimuli source or the inability to synchronize multi-stimuli responses can easily lead to challenges in achieving precise-theranostics of RA. Herein, a homology-activated ultrasensitive nanomedicines MnO2-CQ4T-GOx(MCG NMs) is designed for NIR-II fluorescence(NIR-II FL)/magnetic resonance imaging(MRI)-guided effective "knock-on" dynamic anti-RA therapy. Building upon the characteristics of the RA-microenvironment, the MCG innovatively construct a MnO2-Mn2+ system, which can normalized activation sites. The ultrasensitive-responsive degradation is achieved using the multi-stimuli processes in the RA-microenvironment, triggering release of functional small molecules. The produced Mn2+ can exert Fenton-like activity to generate •OH from H2O2, thus providing the effective chemodynamic therapy(CDT). Moreover, the up-regulation of H2O2 by GOx-catalysis not only sensitizes the MnO2-Mn2+ system but also achieves self-enhancing CDT efficacy. The NIR-II FL quenching of CQ4T-BSA in the aggregated state occurs in MCG NMs, which can be rapidly and precisely "turn-on" via the MnO2-Mn2+ system. Meanwhile, the integration of activated Mn2+-based MRI imaging has successfully developed an activatable dual-modal imaging. Feedback imaging-guided precise photodynamic therapy of CQ4T-BSA can achieve efficient "knock-on" dynamic therapy for RA.
Collapse
Affiliation(s)
- Rong Dai
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Mingxin Zhao
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaochun Zheng
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Dongsheng Li
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Weiwei Kang
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Huifang Hao
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Xuejiao Chen
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Yarong Jin
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Juan Li
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Qin Liu
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Ziliang Zheng
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
- Institute of Medical Technology, Shanxi Medical University, Taiyuan, 030001, China
| | - Ruiping Zhang
- Department of Radiology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, 030000, China
| |
Collapse
|
12
|
Xu H, Wang Y, Rong X, Wang D, Xie J, Huang Z, Zeng W, Fu X, Li J, Zhou Z. Ingenious Synergy of a Pathology-Specific Biomimetic Multifunctional Nanoplatform for Targeted Therapy in Rheumatoid Arthritis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305197. [PMID: 37914665 DOI: 10.1002/smll.202305197] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/11/2023] [Indexed: 11/03/2023]
Abstract
Based on the pathological characteristics of rheumatoid arthritis, including the overproduction of reactive oxygen species (ROS), inflammatory responses, and osteoclast differentiation, a biomimetic multifunctional nanomedicine (M-M@I) is designed. Iguratimod (IGU) is loaded, which inhibits inflammatory responses and osteoclast differentiation, into mesoporous polydopamine (MPDA), which scavenges ROS. Subsequently, the nanoparticles are coated with a cell membrane of macrophages to achieve actively targeted delivery of the nanoparticles to inflamed joints. It is shown that the M-M@I nanoparticles are taken up well by lipopolysaccharide-induced RAW 264.7 macrophages or bone marrow-derived macrophages (BMDMs). In vitro, the M-M@I nanoparticles effectively scavenge ROS, downregulate genes related to inflammation promotion and osteoclast differentiation, and reduce the proinflammatory cytokines and osteoclast-related enzymes. They also reduce the polarization of macrophages to a pro-inflammatory M1 phenotype and inhibit differentiation into osteoclasts. In mice with collagen-induced arthritis, the M-M@I nanoparticles accumulate at arthritic sites and circulate longer, significantly mitigating arthritis symptoms and bone destruction. These results suggest that the pathology-specific biomimetic multifunctional nanoparticles are effective against rheumatoid arthritis, and they validate the approach of developing multifunctional therapies that target various pathological processes simultaneously.
Collapse
Affiliation(s)
- Hong Xu
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuemin Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Xiao Rong
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Duan Wang
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinwei Xie
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zeyu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weinan Zeng
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoxue Fu
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu, 610041, China
| | - Zongke Zhou
- Department of Orthopedic Surgery and Orthopedic Research Institution, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
13
|
Li P, Wang C, Huo H, Xu C, Sun H, Wang X, Wang L, Li L. Prodrug-based nanomedicines for rheumatoid arthritis. DISCOVER NANO 2024; 19:9. [PMID: 38180534 PMCID: PMC10769998 DOI: 10.1186/s11671-023-03950-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/25/2023] [Indexed: 01/06/2024]
Abstract
Most antirheumatic drugs with high toxicity exhibit a narrow therapeutic window due to their nonspecific distribution in the body, leading to undesirable side effects and reduced patient compliance. To in response to these challenges, prodrug-based nanoparticulate drug delivery systems (PNDDS), which combines prodrug strategy and nanotechnology into a single system, resulting their many advantages, including stability for prodrug structure, the higher drug loading capacity of the system, improving the target activity and bioavailability, and reducing their untoward effects. PNDDS have gained attention as a method for relieving arthralgia syndrome of rheumatoid arthritis in recent years. This article systematically reviews prodrug-based nanocarriers for rheumatism treatment, including Nano systems based on prodrug-encapsulated nanomedicines and conjugate-based nanomedicines. It provides a new direction for the clinical treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Pei Li
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Cong Wang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Hongjie Huo
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Chunyun Xu
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Huijun Sun
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xinyu Wang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Li Wang
- College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Lei Li
- College of Pharmacy, Dalian Medical University, Dalian, China.
| |
Collapse
|
14
|
Hu Y, Liu J, Xin L, Wan L, Qi Y, Li Y, Chen Y. Huangqin Qingre Chubi Capsule is Associated with Reduced Risk of Readmission in Patients with Rheumatoid Arthritis: A Real-World Retrospective Cohort Study. Int J Gen Med 2023; 16:4819-4834. [PMID: 37908759 PMCID: PMC10615257 DOI: 10.2147/ijgm.s431124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/13/2023] [Indexed: 11/02/2023] Open
Abstract
Purpose The therapeutic effects of Huangqin Qingre Chubi (HQC) in rheumatoid arthritis (RA) have been documented. However, there is a lack of real-world clinical evidence supporting its efficacy. Methods Patients diagnosed with RA were recruited from the First Affiliated Hospital of the Anhui University of Chinese Medicine. Patient information was obtained from the hospital's database. Propensity score matching (PSM), Kaplan-Meier curve, and Cox proportional hazards model were used to control confounding factors and analyze the factors influencing readmission. Association rule analysis and random walk evaluation models were used to evaluate the correlations among HQC treatment, inflammation indicators, and self-perception of patients (SPP) scale. Results After PSM, 3423 patients were enrolled, with 1142 in the HQC group and 2281 in the non-HQC group. The readmission risk of the HQC group was significantly lower than that of the non-HQC group. Combined univariate and multivariate analysis results revealed that risk factors for readmission were age >60 years, female sex, hypertension, chronic gastritis, and elevated levels of laboratory indices, including anticyclic citrullinated peptide and complement component 3 (C3) and C4. HQC, disease-modifying antirheumatic drugs, nonsteroidal anti-inflammatory drugs, and glucocorticoid therapy were protective factors for readmission. HQC treatment was closely associated with improvements in many factors, including erythrocyte sedimentation rate, C-reactive protein, C3, rheumatoid factor levels, visual analog scale, depression self-assessment scale, and patient-reported activity index scores with RA. Conclusion HQC treatment can reduce the risk of readmission and significantly improve immune inflammatory indicators and SPP in patients with RA, with no risk of hepatorenal toxicity.
Collapse
Affiliation(s)
- Yuedi Hu
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
- College of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
| | - Jian Liu
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
- Department of Internal Medicine Application Foundation Research and Development, Anhui Province—Key Laboratory of Modern Chinese Medicine, Hefei, Anhui, People’s Republic of China
| | - Ling Xin
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
- Department of Internal Medicine Application Foundation Research and Development, Anhui Province—Key Laboratory of Modern Chinese Medicine, Hefei, Anhui, People’s Republic of China
| | - Lei Wan
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
- Institute of Rheumatology, Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
- Department of Internal Medicine Application Foundation Research and Development, Anhui Province—Key Laboratory of Modern Chinese Medicine, Hefei, Anhui, People’s Republic of China
| | - Yajun Qi
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
| | - Yang Li
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
| | - Yiming Chen
- Department of Rheumatology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, People’s Republic of China
| |
Collapse
|
15
|
Műzes G, Sipos F. CAR-Based Therapy for Autoimmune Diseases: A Novel Powerful Option. Cells 2023; 12:1534. [PMID: 37296654 PMCID: PMC10252902 DOI: 10.3390/cells12111534] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
The pervasive application of chimeric antigen receptor (CAR)-based cellular therapies in the treatment of oncological diseases has long been recognized. However, CAR T cells can target and eliminate autoreactive cells in autoimmune and immune-mediated diseases. By doing so, they can contribute to an effective and relatively long-lasting remission. In turn, CAR Treg interventions may have a highly effective and durable immunomodulatory effect via a direct or bystander effect, which may have a positive impact on the course and prognosis of autoimmune diseases. CAR-based cellular techniques have a complex theoretical foundation and are difficult to implement in practice, but they have a remarkable capacity to suppress the destructive functions of the immune system. This article provides an overview of the numerous CAR-based therapeutic options developed for the treatment of immune-mediated and autoimmune diseases. We believe that well-designed, rigorously tested cellular therapies could provide a promising new personalized treatment strategy for a significant number of patients with immune-mediated disorders.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary;
| | | |
Collapse
|