1
|
Ubhe A. IL-1 receptor antagonist: etiological and drug delivery systems overview. Inflamm Res 2024:10.1007/s00011-024-01960-y. [PMID: 39455436 DOI: 10.1007/s00011-024-01960-y] [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: 07/27/2024] [Revised: 09/18/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVE This article is aims to provide an overview of studies reported in the literature to investigate the etiological role of IL-1/IL-1ra in various disease conditions and the different drug delivery systems developed to achieve IL-1ra as a possible therapeutic option. METHODS Studies reported in PubMed, Google scholar, and other open-source literature related to etiological involvement of IL-1ra in pathophysiological conditions and various drug delivery schemes developed for IL-1ra for its efficacy evaluation as a possible treatment for different disease conditions were surveyed. RESULTS AND CONCLUSIONS The pathophysiological conditions involving IL-1/IL-1 ra spanned CNS-related disorders, Diabetes, Cardiac disorders, Ocular disease conditions, Gastrointestinal conditions, Tumor growth & metastasis, and miscellaneous conditions. The drug delivery systems developed for IL-1ra included a commercial drug product, Gene therapy, Antibody fusions, Extended-release delivery systems, and Pegylated-IL-1ra systems.
Collapse
|
2
|
Schlesinger N, Pillinger MH, Simon LS, Lipsky PE. Interleukin-1β inhibitors for the management of acute gout flares: a systematic literature review. Arthritis Res Ther 2023; 25:128. [PMID: 37491293 PMCID: PMC10367374 DOI: 10.1186/s13075-023-03098-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/23/2023] [Indexed: 07/27/2023] Open
Abstract
OBJECTIVES The objective of this systematic review was to assess the effects of interleukin-1β (IL-1β) inhibitors on gout flares. METHODS Studies published between 2011 and 2022 that evaluated the effects of IL-1β inhibitors in adult patients experiencing gout flares were eligible for inclusion. Outcomes including pain, frequency and intensity of gout flares, inflammation, and safety were assessed. Five electronic databases (Pubmed/Medline, Embase, Biosis/Ovid, Web of Science and Cochrane Library) were searched. Two independent reviewers performed study screening, data extraction and risk of bias assessments (Cochrane Risk of Bias Tool 2 for randomised controlled trials [RCTs] and Downs and Black for non-RCTs). Data are reported as a narrative synthesis. RESULTS Fourteen studies (10 RCTs) met the inclusion criteria, with canakinumab, anakinra, and rilonacept being the three included IL-1β inhibitors. A total of 4367 patients with a history of gout were included from the 14 studies (N = 3446, RCTs; N = 159, retrospective studies [with a history of gout]; N = 762, post hoc analysis [with a history of gout]). In the RCTs, canakinumab and rilonacept were reported to have a better response compared to an active comparator for resolving pain, while anakinra appeared to be not inferior to an active comparator for resolving pain. Furthermore, canakinumab and rilonacept reduced the frequency of gout flares compared to the comparators. All three medications were mostly well-tolerated compared to their comparators. CONCLUSION IL-1β inhibitors may be a beneficial and safe medication for patients experiencing gout flares for whom current standard therapies are unsuitable. REVIEW PROTOCOL REGISTRATION PROSPERO ID: CRD42021267670.
Collapse
Affiliation(s)
- Naomi Schlesinger
- Division of the Rheumatology at the Spencer Fox Eccles School of Medicine, University of Utah, Harold J, Ardella T, and Helen T Stevenson Presidential Endowed Chair of Rheumatology, Salt Lake City, UT, 84132, USA.
| | - Michael H Pillinger
- The Division of Rheumatology, NYU Grossman School of Medicine, New York, USA
| | | | | |
Collapse
|
3
|
Mohapatra A, Rajendrakumar SK, Chandrasekaran G, Revuri V, Sathiyamoorthy P, Lee YK, Lee JH, Choi SY, Park IK. Biomineralized Nanoscavenger Abrogates Proinflammatory Macrophage Polarization and Induces Neutrophil Clearance through Reverse Migration during Gouty Arthritis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:3812-3825. [PMID: 36646643 DOI: 10.1021/acsami.2c19684] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The deposition of monosodium urate (MSU) crystals induces the overexpression of reactive oxygen species (ROS) and proinflammatory cytokines in residential macrophages, further promoting the infiltration of inflammatory leukocytes in the joints of gouty arthritis. Herein, a peroxidase-mimicking nanoscavenger was developed by forming manganese dioxide over albumin nanoparticles loaded with an anti-inflammatory drug, indomethacin (BIM), to block the secretion of ROS and COX2-induced proinflammatory cytokines in the MSU-induced gouty arthritis model. In the MSU-induced arthritis mouse model, the BIM nanoparticles alleviated joint swelling, which is attributed to the abrogation of ROS and inflammatory cytokine secretions from proinflammatory macrophages that induces neutrophil infiltration and fluid building up in the inflammation site. Further, the BIM nanoparticle treatment reduced the influx of macrophages and neutrophils in the injured region by blocking migration and inducing reverse migration in the zebrafish larva tail amputation model as well as in MSU-induced peritonitis and air pouch mouse models. Overall, the current strategy of employing biomineralized nanoscavengers for arthritis demonstrates clinical significance in dual blocking of peroxides and COX2 to prevent influx of inflammatory cells into the sites of inflammation.
Collapse
Affiliation(s)
- Adityanarayan Mohapatra
- Department of Biomedical Sciences and Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju61469, Republic of Korea
| | - Santhosh Kalash Rajendrakumar
- Department of Biomedical Sciences and Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju61469, Republic of Korea
- Department of Chemistry, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Gopalakrishnan Chandrasekaran
- Department of Biomedical Sciences and Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju61469, Republic of Korea
| | - Vishnu Revuri
- Department of Green Bioengineering, Korea National University of Transportation, Chungju27470, Republic of Korea
| | - Padmanaban Sathiyamoorthy
- Department of Biomedical Sciences and Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju61469, Republic of Korea
| | - Yong-Kyu Lee
- Department of Green Bioengineering, Korea National University of Transportation, Chungju27470, Republic of Korea
| | - Jae Hyuk Lee
- Department of Pathology, Chonnam National University Medical School, Gwangju61469, Republic of Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences and Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju61469, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences and Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju61469, Republic of Korea
| |
Collapse
|
4
|
Zhou X, Shi Q, Li J, Quan S, Zhang X, Gu L, Li H, Ju Y, Hu M, Li Q. Medicinal fungus Phellinus igniarius alleviates gout in vitro by modulating TLR4/NF-kB/NLRP3 signaling. Front Pharmacol 2022; 13:1011406. [PMID: 36339594 PMCID: PMC9634182 DOI: 10.3389/fphar.2022.1011406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/11/2022] [Indexed: 11/28/2022] Open
Abstract
Background:Phellinus igniarius (P. igniarius) is a valuable medicinal and edible fungus with various biological activities such as anti-inflammation, antioxidation, and immune regulation. In this study, we explored the effects of P. igniarius on a gout model in vitro. Methods: The DPPH, ABTS, and FRAP methods were combined to determine and compare the antioxidant activities of wild P. igniarius total polyphenols (WPP) and cultivated P. igniarius total polyphenols (CPP) in vitro. Spectrophotometry was used to compare the inhibitory effect of WPP and CPP on xanthine oxidase (XO) activity to evaluate anti-hyperuricemia activity in vitro. HUVECs were stimulated with monosodium urate (MSU) crystals for 24 h to establish an acute gouty inflammation model in vitro. The protective effects were compared by measuring cell viability; the contents of ICAM-1, IL-1β, IL-6 and VCAM-1; the protein expressions of TLR4 and NLRP3; reactive oxygen species production; and the nuclear translocation of NF-κB p65. UHPLC-QE-MS technology was used to explore the potential metabolic mechanism of P. igniarius against gout. Results: WPP and CPP had strong antioxidant capacity, and the antioxidant capacity of CPP was similar to that of WPP. In a comparative experiment of xanthine oxidase activity inhibition by WPP and CPP, the IC50 values were 88.19 μg/ml and 108.0 μg/ml, respectively. At a dose of 40 μg/ml, WPP and CPP significantly improved the decrease in cell viability induced by monosodium urate (150 μg/ml) and inhibited the increase in inflammatory factors such as ICAM-1, IL-1β, IL-6, and VCAM-1. The increase in TLR4 and NLRP3 protein expression induced by MSU crystals in HUVECs was also significantly inhibited by total polyphenols from wild and cultivated P. igniarius. In addition, both significantly improved MSU-induced ROS overproduction and NF-κB p65 nuclear translocation. WPP and CPP may primarily be involved in phenylalanine metabolism and lysophosphatidylcholine metabolism in their role in the treatment of gout. Conclusion: CPP and WPP both showed good antioxidant activity and xanthine oxidase inhibitory activity and had good therapeutic effects on the gout model in vitro. Furthermore, this study indicated that cultivated P. igniarius had a protective effect similar to that of wild P. igniarius, which would be expected to improve the shortage of wild P. igniarius and promote the development of the cultivated P. igniarius industry and product development.
Collapse
|
5
|
Zhao J, Guo S, Schrodi SJ, He D. Trends in the Contribution of Genetic Susceptibility Loci to Hyperuricemia and Gout and Associated Novel Mechanisms. Front Cell Dev Biol 2022; 10:937855. [PMID: 35813212 PMCID: PMC9259951 DOI: 10.3389/fcell.2022.937855] [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: 05/06/2022] [Accepted: 05/31/2022] [Indexed: 11/14/2022] Open
Abstract
Hyperuricemia and gout are complex diseases mediated by genetic, epigenetic, and environmental exposure interactions. The incidence and medical burden of gout, an inflammatory arthritis caused by hyperuricemia, increase every year, significantly increasing the disease burden. Genetic factors play an essential role in the development of hyperuricemia and gout. Currently, the search on disease-associated genetic variants through large-scale genome-wide scans has primarily improved our understanding of this disease. However, most genome-wide association studies (GWASs) still focus on the basic level, whereas the biological mechanisms underlying the association between genetic variants and the disease are still far from well understood. Therefore, we summarized the latest hyperuricemia- and gout-associated genetic loci identified in the Global Biobank Meta-analysis Initiative (GBMI) and elucidated the comprehensive potential molecular mechanisms underlying the effects of these gene variants in hyperuricemia and gout based on genetic perspectives, in terms of mechanisms affecting uric acid excretion and reabsorption, lipid metabolism, glucose metabolism, and nod-like receptor pyrin domain 3 (NLRP3) inflammasome and inflammatory pathways. Finally, we summarized the potential effect of genetic variants on disease prognosis and drug efficacy. In conclusion, we expect that this summary will increase our understanding of the pathogenesis of hyperuricemia and gout, provide a theoretical basis for the innovative development of new clinical treatment options, and enhance the capabilities of precision medicine for hyperuricemia and gout treatment.
Collapse
Affiliation(s)
- Jianan Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Shicheng Guo
- Computation and Informatics in Biology and Medicine, University of WI-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of WI-Madison, Madison, WI, United States
| | - Steven J. Schrodi
- Computation and Informatics in Biology and Medicine, University of WI-Madison, Madison, WI, United States
- Department of Medical Genetics, School of Medicine and Public Health, University of WI-Madison, Madison, WI, United States
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, Shanghai, China
- Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Wang Y, Zhu W, Lu D, Zhang C, Wang Y. Tetrahydropalmatine attenuates MSU crystal-induced gouty arthritis by inhibiting ROS-mediated NLRP3 inflammasome activation. Int Immunopharmacol 2021; 100:108107. [PMID: 34482265 DOI: 10.1016/j.intimp.2021.108107] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 02/07/2023]
Abstract
Activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome plays a crucial role in the inflammatory responses of monosodium urate (MSU) crystal-induced gouty arthritis. Therefore, the molecular basis of NLRP3 inflammasome is very valuable in developing potential therapeutic drugs for gout. Tetrahydropalmatine (THP), the main active component of the traditional Chinese medicinal herb Corydalis yanhusuo, has shown prominent anti-inflammatory and analgesic activities, but to date, these effects have not been investigated exhaustively on gout. This study indicated that THP attenuated pain and swelling in an MSU-induced acute gout model by decreasing pro-inflammatory cytokine production and inflammatory cell infiltration. THP exerted its actions by suppressing NLRP3 inflammasome activation and subsequent formation of caspase-1. Furthermore, results showed that THP alleviated MSU-induced reactive oxygen species (ROS) generation, upstream of NLRP3 inflammasome activation, by an increase in the activities of antioxidant enzymes both in vitro and in vivo. In conclusion, our study suggests that THP suppressed ROS-mediated NLRP3 inflammasome activation in MSU-induced inflammatory responses, which highlights its therapeutic potential in gouty arthritis.
Collapse
Affiliation(s)
- Yifan Wang
- The First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Wen Zhu
- The First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Dandan Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Chuzhao Zhang
- The First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Yue Wang
- The First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210029, PR China.
| |
Collapse
|
7
|
Shi L, Zhao F, Zhu F, Liang Y, Yang F, Zhang G, Xu L, Yin L. Traditional Chinese Medicine Formula "Xiaofeng granules" suppressed gouty arthritis animal models and inhibited the proteoglycan degradation on chondrocytes induced by monosodium urate. JOURNAL OF ETHNOPHARMACOLOGY 2016; 191:254-263. [PMID: 27267827 DOI: 10.1016/j.jep.2016.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 05/10/2016] [Accepted: 06/04/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaofeng Granules (XF) is a kind of granules prepared by the famous traditional Chinese medicine formula for its efficiency in treating gouty diseases. AIM OF THE STUDY We investigated the relevance between XF that made from Modified simiaowan (MSW) as the anti-gouty arthritis drugs and protective mechanisms for cartilage matrix in order to provide the evidence for new drug application. MATERIALS AND METHODS In the present study, we evaluated the anti-gouty arthritis activity of XF in rats and rabbits models induced by MSU together with chondrocytes focusing on the link to proteoglycan degradation in vitro studies. RESULTS The results demonstrated that XF significantly reduced the swelling rate and attenuated the pathological changes in joints. The XF-containing serum were used medicated serum in cellular experiments. The in vitro data were in accordance with the in vivo results, showing that the constituents in XF-containing serum had obvious inhibitory effects on the activation of pro-inflammatory mediators in chondrocytes. Moreover, XF-containing serum substantially inhibited MSU-induced expression of glycosaminoglycans(GAG) and hydroxyproline(Hyp), and up regulated proteoglycan, which might be associated with the regulation of the balance of MMP-3/TIMP-1and ADAMTS-4/TIMP-3 inchondrocytes. CONCLUSION In conclusion, XF that made from MSW showed obvious effects on acute gouty arthritis, which also provided an effective protection on cartilage matrix degradation.
Collapse
Affiliation(s)
- Le Shi
- College of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing 210023 PR China
| | - Fangli Zhao
- College of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing 210023 PR China
| | - Fangfang Zhu
- College of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing 210023 PR China
| | - Yuqiong Liang
- College of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing 210023 PR China
| | - Fan Yang
- College of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing 210023 PR China
| | - Guangji Zhang
- College of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing 210023 PR China
| | - Li Xu
- College of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing 210023 PR China.
| | - Lian Yin
- College of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing 210023 PR China.
| |
Collapse
|
8
|
Zhao F, Guochun L, Yang Y, Shi L, Xu L, Yin L. A network pharmacology approach to determine active ingredients and rationality of herb combinations of Modified-Simiaowan for treatment of gout. JOURNAL OF ETHNOPHARMACOLOGY 2015; 168:1-16. [PMID: 25824593 DOI: 10.1016/j.jep.2015.03.035] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 01/25/2015] [Accepted: 03/10/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Modified Simiaowan (MSW) is a traditional Chinese medicine (TCM) formula and is widely used as a clinically medication formula for its efficiency in treating gouty diseases.To predict the active ingredients in MSW and uncover the rationality of herb combinations of MSW. MATERIALS AND METHODS Three drug-target networks including the "candidate ingredient-target network" (cI-cT) that links the candidate ingredients and targets, the "core ingredient-target-pathway network" connecting core potential ingredients and targets through related pathways, and the "rationality of herb combinations of MSW network", which was derived from the cI-cT network, were developed to dissect the active ingredients in MSW and relationship between ingredients in herb combinations and their targets for gouty diseases. On the other hand, herbal ingredients comparisons were also conducted based on six physicochemical properties to investigate whether the herbs in MSW are similar in chemicals. Moreover, HUVEC viability and expression levels of ICAM-1 induced by monosodium urate (MSU) crystals were assessed to determine the activities of potential ingredients in MSW. RESULTS Predicted by the core ingredient-target-pathway network, we collected 30 core ingredients in MSW and 25 inflammatory cytokines and uric acid synthetase or transporters, which are effective for gouty treatment through some related pathways. Experimental results also confirmed that those core ingredients could significantly increase HUVEC viability and attenuate the expression of ICAM-1, which supported the effectiveness of MSW in treating gouty diseases. Moreover, heat-clearing and dampness-eliminating herbs in MSW have similar physicochemical properties, which stimulate all the inflammatory and uric acid-lowing targets respectively, while the core drug and basic prescription in MSW stimulate the major and almost all the core targets, respectively. CONCLUSION Our work successfully predicts the active ingredients in MSW and explains the cooperation between these ingredients and corresponding targets through related pathways for gouty diseases, and provides basis for an alternative approach to investigate the rationality of herb combinations of MSW on the network pharmacology level, which might be beneficial to drug development and applications.
Collapse
Affiliation(s)
- Fangli Zhao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Li Guochun
- College of Preclinical Medical, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Yanhua Yang
- Changzhou Seventh People's Hospital, Changzhou 213011, Jiangsu, China
| | - Le Shi
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Li Xu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Lian Yin
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210046, China.
| |
Collapse
|
9
|
Shi L, Xu L, Yang Y, Song H, Pan H, Yin L. Suppressive effect of modified Simiaowan on experimental gouty arthritis: an in vivo and in vitro study. JOURNAL OF ETHNOPHARMACOLOGY 2013; 150:1038-1044. [PMID: 24184191 DOI: 10.1016/j.jep.2013.10.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 09/24/2013] [Accepted: 10/05/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Modified Simiaowan (MSW) is frequently prescribed in traditional Chinese medicine and is famous for its efficiency in treating gouty diseases. We investigated the effectiveness of MSW as an anti-gouty inflammation medicine and its mechanism of action in monosodium urate (MSU) crystal-induced gouty rat in vivo and human umbilical vein endothelial cells (HUVECs) in vitro. MATERIALS AND METHODS Rats were orally administered with the water extract of MSW (2.5, 5.0, and 10 g/kg body weight), and indomethacin (12.5 mg/kg body weight) was given as a positive control. An intra-articular injection of 0.1 ml (10 mg) of MSU crystals was used to generate the gout model to assess paw volume at 1, 3, and 5h after MSU crystal injection and to analyze the histopathology of joint synovial tissues in the control and MSU crystal-treated rats at the end of the experiment. The HUVEC viability, expression levels of endothelial cell intercellular adhesion molecule-1 (ICAM-1), and apoptotic HUVECs were assessed in MSU crystal-induced HUVECs treated with (75 μg/ml to 300 μg/ml) MSW and (20 μg/ml) indomethacin by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide test, reverse transcriptase PCR, and acridine orange/ethidium bromide staining, respectively. RESULTS MSW could significantly prevent the paw swelling and neutrophil infiltration induced by intra-articular MSU injection in rats. MSW also showed potent analgesic effects at (5.0, 10, and 20 g/kg body weight) in acetic acid-induced mice depending on the dosage. Moreover, MSW could significantly increase HUVEC viability, attenuate the expression of ICAM-1, and prevent apoptosis of HUVECs in MSU-induced HUVECs. CONCLUSION These results provide evidence for the anti-inflammatory effect of MSW by preventing neutrophil infiltration and apoptosis of HUVECs. These mechanisms of action of MSW are similar to that by indomethacin. Therefore, the results support the effectiveness of MSW in treating gouty diseases.
Collapse
Affiliation(s)
- Le Shi
- College of Pharmacy, Nanjing University of Traditional Chinese Medicine, Nanjing 210046, PR China
| | | | | | | | | | | |
Collapse
|
10
|
Abstract
Gout prevalence is increasing, yet management remains suboptimal. Fortunately, new insights into gout biology are permitting the development of novel, potentially more effective strategies for both gouty inflammation and urate lowering. Colchicine, a drug long used for gout, has been recently approved (for the first time ever) by the FDA, based on a new, safer dosing regimen. The recently appreciated centrality of IL-1β in acute gouty inflammation has prompted studies of agents blocking the IL-1β receptor or soluble IL-1β signaling (canakinumab, rilonacept, anakinra). Novel approaches to urate lowering have led to mechanism-based therapies such as urate synthesis inhibitors (febuxostat is already FDA approved and BCX4208 is in development), URAT-1 inhibitors promoting renal uric acid excretion (lesinurad), and recombinant uricase to directly catabolize urate (pegloticase). These new treatments do not obviate the need for lifestyle and dietary management, another area in which significant scientific and clinical progress has recently been made.
Collapse
Affiliation(s)
- Daria B Crittenden
- Crystal Diseases Study Group, Division of Rheumatology, Department of Medicine, NYU School of Medicine/NYU Langone Medical Center, New York, New York 10003, USA.
| | | |
Collapse
|
11
|
Mechanistic aspects of inflammation and clinical management of inflammation in acute gouty arthritis. J Clin Rheumatol 2013; 19:19-29. [PMID: 23319019 DOI: 10.1097/rhu.0b013e31827d8790] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It has been recently demonstrated that interleukin 1β (IL-1β) plays a central role in monosodium urate crystal-induced inflammation and that the NALP3 inflammasome plays a major role in IL-1β production. These discoveries have offered new insights into the pathogenesis of acute gouty arthritis. In this review, we discuss the molecular mechanisms by which monosodium urate crystals induce acute inflammation and examine the mechanisms of action (MOAs) of traditional anti-inflammatory drugs (e.g., nonsteroidal anti-inflammatory drugs, colchicine, and glucocorticoids) and biologic agents (e.g., the IL-1β antagonists anakinra, rilonacept, and canakinumab) to understand how their MOAs contribute to their safety profiles. Traditional anti-inflammatory agents may act on the IL-1β pathway at some level; however, their MOAs are broad-ranging, unspecific, and biologically complex. This lack of specificity may explain the range of systemic adverse effects associated with them. The therapeutic margins of nonsteroidal anti-inflammatory drugs, colchicine, and glucocorticoids are particularly low in elderly patients and in patients with cardiovascular, metabolic, or renal comorbidities that are frequently associated with gouty arthritis. In contrast, the IL-1β antagonists act on very specific targets of inflammation, which may decrease the potential for systemic adverse effects, although infrequent but serious adverse events (including infection and administration reactions) have been reported. Because these IL-1β antagonists target an early event immediately downstream from NALP3 inflammasome activation, they may provide effective alternatives to traditional agents with minimal systemic adverse effects. Results of ongoing trials of IL-1β antagonists will likely provide clarification of their potential role in the management of acute gouty arthritis.
Collapse
|
12
|
|
13
|
Tran TH, Pham JT, Shafeeq H, Manigault KR, Arya V. Role of Interleukin-1 Inhibitors in the Management of Gout. Pharmacotherapy 2013; 33:744-53. [DOI: 10.1002/phar.1265] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Tran H. Tran
- New York Presbyterian Hospital/Columbia Medical Center; College of Pharmacy and Health Sciences; St. John's University; Queens; New York
| | | | - Hira Shafeeq
- New York Presbyterian Hospital/Weill Cornell Medical Center; College of Pharmacy and Health Sciences; St. John's University; Queens; New York
| | | | - Vibhuti Arya
- NYC Department of Health and Mental Hygiene; College of Pharmacy and Health Sciences; St. John's University; Primary Care Information Project; Queens; New York
| |
Collapse
|
14
|
Schumacher HR, Evans RR, Saag KG, Clower J, Jennings W, Weinstein SP, Yancopoulos GD, Wang J, Terkeltaub R. Rilonacept (interleukin-1 trap) for prevention of gout flares during initiation of uric acid-lowering therapy: results from a phase III randomized, double-blind, placebo-controlled, confirmatory efficacy study. Arthritis Care Res (Hoboken) 2013; 64:1462-70. [PMID: 22549879 DOI: 10.1002/acr.21690] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To evaluate the efficacy and safety of the interleukin-1 inhibitor rilonacept (interleukin-1 Trap) for gout flare prevention during initiation of uric acid-lowering therapy (ULT). METHODS In total, 241 adult patients with gout, ≥2 gout flares within the past year, and a serum urate level ≥7.5 mg/dl were initiated on allopurinol 300 mg daily and randomly allocated in a 1:1:1 ratio to receive 16 once-weekly subcutaneous injections of placebo, rilonacept 80 mg, or rilonacept 160 mg, with a double (loading) dose on day 1. Allopurinol was titrated to achieve a serum urate level of <6.0 mg/dl. The study was powered for the primary efficacy end point, the number of gout flares per patient through week 16. RESULTS More patients in the rilonacept groups (80.0% in the rilonacept 80 mg group, 86.4% in the rilonacept 160 mg group) completed the study than in the placebo group (72.5%; P < 0.05 for the rilonacept 160 mg group versus the placebo group). Over 16 weeks, the mean number of gout flares per patient was significantly reduced by rilonacept treatment (placebo: 1.06, rilonacept 80 mg: 0.29 [P < 0.001], rilonacept 160 mg: 0.21 [P < 0.001]). Significantly lower proportions of patients reported ≥1 gout flares with rilonacept 80 mg (18.8%) and rilonacept 160 mg (16.3%) relative to placebo (46.8%; P < 0.001 for both). Except for injection site reactions (1.3% in the placebo group versus 8.8% in the rilonacept 80 mg group [P = 0.0635, post hoc analysis] and 19.8% in the rilonacept 160 mg group [P = 0.0001, post hoc analysis]), the incidence of adverse events was generally balanced among the treatment groups. CONCLUSION Rilonacept markedly reduced the occurrence of gout flares associated with the initiation of ULT. The efficacy and safety profile suggests that rilonacept may have the potential to improve long-term disease control for some patients by improving adherence to ULT by reducing flares during the first months after ULT initiation.
Collapse
|
15
|
Gout as autoinflammatory disease: New mechanisms for more appropriated treatment targets. Autoimmun Rev 2012; 12:66-71. [DOI: 10.1016/j.autrev.2012.07.024] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
16
|
|
17
|
|
18
|
Schlesinger N, Mysler E, Lin HY, De Meulemeester M, Rovensky J, Arulmani U, Balfour A, Krammer G, Sallstig P, So A. Canakinumab reduces the risk of acute gouty arthritis flares during initiation of allopurinol treatment: results of a double-blind, randomised study. Ann Rheum Dis 2011; 70:1264-71. [PMID: 21540198 PMCID: PMC3103669 DOI: 10.1136/ard.2010.144063] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Objective This study assessed the efficacy and safety of canakinumab, a fully human anti-interleukin 1β monoclonal antibody, for prophylaxis against acute gouty arthritis flares in patients initiating urate-lowering treatment. Methods In this double-blind, double-dummy, dose-ranging study, 432 patients with gouty arthritis initiating allopurinol treatment were randomised 1:1:1:1:1:1:2 to receive: a single dose of canakinumab, 25, 50, 100, 200, or 300 mg subcutaneously; 4×4-weekly doses of canakinumab (50+50+25+25 mg subcutaneously); or daily colchicine 0.5 mg orally for 16 weeks. Patients recorded details of flares in diaries. The study aimed to determine the canakinumab dose having equivalent efficacy to colchicine 0.5 mg at 16 weeks. Results A dose-response for canakinumab was not apparent with any of the four predefined dose-response models. The estimated canakinumab dose with equivalent efficacy to colchicine was below the range of doses tested. At 16 weeks, there was a 62% to 72% reduction in the mean number of flares per patient for canakinumab doses ≥50 mg versus colchicine based on a negative binomial model (rate ratio: 0.28–0.38, p≤0.0083), and the percentage of patients experiencing ≥1 flare was significantly lower for all canakinumab doses (15% to 27%) versus colchicine (44%, p<0.05). There was a 64% to 72% reduction in the risk of experiencing ≥1 flare for canakinumab doses ≥50 mg versus colchicine at 16 weeks (hazard ratio (HR): 0.28–0.36, p≤0.05). The incidence of adverse events was similar across treatment groups. Conclusions Single canakinumab doses ≥50 mg or four 4-weekly doses provided superior prophylaxis against flares compared with daily colchicine 0.5 mg.
Collapse
Affiliation(s)
- Naomi Schlesinger
- Division of Rheumatology, Department of Medicine, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, New Brunswick, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Schlesinger N, De Meulemeester M, Pikhlak A, Yücel AE, Richard D, Murphy V, Arulmani U, Sallstig P, So A. Canakinumab relieves symptoms of acute flares and improves health-related quality of life in patients with difficult-to-treat Gouty Arthritis by suppressing inflammation: results of a randomized, dose-ranging study. Arthritis Res Ther 2011; 13:R53. [PMID: 21439048 PMCID: PMC3132043 DOI: 10.1186/ar3297] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 02/11/2011] [Accepted: 03/25/2011] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION We report the impact of canakinumab, a fully human anti-interleukin-1β monoclonal antibody, on inflammation and health-related quality of life (HRQoL) in patients with difficult-to-treat Gouty Arthritis. METHODS In this eight-week, single-blind, double-dummy, dose-ranging study, patients with acute Gouty Arthritis flares who were unresponsive or intolerant to--or had contraindications for--non-steroidal anti-inflammatory drugs and/or colchicine were randomized to receive a single subcutaneous dose of canakinumab (10, 25, 50, 90, or 150 mg) (N = 143) or an intramuscular dose of triamcinolone acetonide 40 mg (N = 57). Patients assessed pain using a Likert scale, physicians assessed clinical signs of joint inflammation, and HRQoL was measured using the 36-item Short-Form Health Survey (SF-36) (acute version). RESULTS At baseline, 98% of patients were suffering from moderate-to-extreme pain. The percentage of patients with no or mild pain was numerically greater in most canakinumab groups compared with triamcinolone acetonide from 24 to 72 hours post-dose; the difference was statistically significant for canakinumab 150 mg at these time points (P < 0.05). Treatment with canakinumab 150 mg was associated with statistically significant lower Likert scores for tenderness (odds ratio (OR), 3.2; 95% confidence interval (CI), 1.27 to 7.89; P = 0.014) and swelling (OR, 2.7; 95% CI, 1.09 to 6.50, P = 0.032) at 72 hours compared with triamcinolone acetonide. Median C-reactive protein and serum amyloid A levels were normalized by seven days post-dose in most canakinumab groups, but remained elevated in the triamcinolone acetonide group. Improvements in physical health were observed at seven days post-dose in all treatment groups; increases in scores were highest for canakinumab 150 mg. In this group, the mean SF-36 physical component summary score increased by 12.0 points from baseline to 48.3 at seven days post-dose. SF-36 scores for physical functioning and bodily pain for the canakinumab 150 mg group approached those for the US general population by seven days post-dose and reached norm values by eight weeks post-dose. CONCLUSIONS Canakinumab 150 mg provided significantly greater and more rapid reduction in pain and signs and symptoms of inflammation compared with triamcinolone acetonide 40 mg. Improvements in HRQoL were seen in both treatment groups with a faster onset with canakinumab 150 mg compared with triamcinolone acetonide 40 mg. TRIAL REGISTRATION clinicaltrials.gov: NCT00798369.
Collapse
Affiliation(s)
- Naomi Schlesinger
- Division of Rheumatology, Department of Medicine, Robert Wood Johnson Medical School, 125 Patterson Street, New Brunswick, NJ 089010, USA
| | - Marc De Meulemeester
- Pratique Médicale, Cabinet de Là-Haut, Rue de Marchienne 113, 6534 Gozée, Belgium
| | - Andrey Pikhlak
- Moscow State University of Medicine and Dentistry, Clinical-Diagnostic Center MSMSU, Dolgorukovskaya Street 4, Moscow 127006, Russia
| | - A Eftal Yücel
- Baskent University, Faculty of Medicine, Baglica Kampusu, Eskisehir Yolu 20.km, Baglica 06530, Ankara, Turkey
| | - Dominik Richard
- Immunology & Infectious Disease Therapeutic Area, Novartis Pharma AG, Lichtstrasse 35, CH-4056, Basel, Switzerland
| | - Valda Murphy
- Immunology & Infectious Disease Therapeutic Area, Novartis Pharma AG, Lichtstrasse 35, CH-4056, Basel, Switzerland
| | - Udayasankar Arulmani
- Immunology & Infectious Disease Therapeutic Area, Novartis Pharma AG, Lichtstrasse 35, CH-4056, Basel, Switzerland
| | - Peter Sallstig
- Immunology & Infectious Disease Therapeutic Area, Novartis Pharma AG, Lichtstrasse 35, CH-4056, Basel, Switzerland
| | - Alexander So
- Service de Rhumatologie, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Av. Pierre-Decker 5, CH-1005, Lausanne, Switzerland
| |
Collapse
|
20
|
Lachmann HJ, Quartier P, So A, Hawkins PN. The emerging role of interleukin-1β in autoinflammatory diseases. ACTA ACUST UNITED AC 2011; 63:314-24. [PMID: 20967858 DOI: 10.1002/art.30105] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Helen J Lachmann
- National Amyloidosis Centre, University College London Medical School, London, UK.
| | | | | | | |
Collapse
|
21
|
Affiliation(s)
- Tuhina Neogi
- Section of Clinical Epidemiology Research and Training Unit, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
22
|
Quartier P. Interleukin-1 antagonists in the treatment of autoinflammatory syndromes, including cryopyrin-associated periodic syndrome. Open Access Rheumatol 2011; 3:9-18. [PMID: 27790000 PMCID: PMC5074783 DOI: 10.2147/oarrr.s6696] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Cryopyrin-associated periodic syndrome (CAPS) include a group of rare autoinflammatory disorders, the spectrum of which ranges from the mildest form, ie, familial cold autoinflammatory syndrome to more severe phenotypes, ie, Muckle-Wells syndrome, and chronic infantile neurological cutaneous and articular syndrome, also known as neonatal-onset multisystem inflammatory disease. Three interleukin (IL)-1 antagonists have been tested in adults and children with CAPS, ie, anakinra, a recombinant homolog of the human IL-1 receptor antagonist; rilonacept, a fusion protein comprising the extracellular domains of IL-1 receptor I and the IL-1 adaptor protein, IL-1RAcP, attached to a human immunoglobulin G molecule; and canakinumab, the anti-IL-1β monoclonal antibody. Following rapid clinical development, rilonacept and canakinumab were approved by both the US Food and Drug Administration and the European Medicines Agency for use in adults and children. This review describes how the study of CAPS has helped us to understand better the way the innate immune system works, the pathogenesis of autoinflammatory syndromes, and the key role of IL-1. It also reviews the effects of IL-1 blockade in CAPS and other disorders, in particular systemic juvenile idiopathic arthritis, adult-onset Still's disease, and gout. Finally, this review covers some issues addressed by very recent and ongoing work regarding treatment indications, from orphan diseases to common disorders, continuous versus intermittent treatment, the pharmacokinetics, pharmacodynamics, and optimal dosages of the different drugs, as well as the need for Phase IV trials, exhaustive registries, and long-term follow-up of several patient cohorts.
Collapse
Affiliation(s)
- Pierre Quartier
- Unité d’Immunologie-Hématologie et Rhumatologie pédiatriques, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|