1
|
Tanaka Y, Mizutani H, Fujii K, Okubo N. Safety and effectiveness of denosumab in Japanese patients with rheumatoid arthritis: A 2-year post-marketing surveillance study. Mod Rheumatol 2024; 34:927-935. [PMID: 38048431 DOI: 10.1093/mr/road108] [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: 04/03/2023] [Revised: 08/30/2023] [Accepted: 11/15/2023] [Indexed: 12/06/2023]
Abstract
OBJECTIVES To investigate the 2 year safety and effectiveness of denosumab 60 mg in patients with rheumatoid arthritis (RA) in clinical practice in Japan. METHODS This 2 year, prospective, observational cohort study included patients who initiated treatment with denosumab 60 mg for the progression of bone erosion associated with RA. Key endpoints were adverse drug reactions (ADRs), progression of bone erosion, and 28-joint Disease Activity Score based on C-reactive protein or erythrocyte sedimentation rate. Univariate and multivariate analyses were conducted to determine the risk factors for ADRs and the progression of bone erosion. RESULTS In the safety analysis set (n = 1239), the incidence of ADRs was 3.0%; the most common ADRs were hypocalcaemia (1.2%) and osteonecrosis of jaw-related events (0.6%). A history of any drug allergy was a statistically significant risk factor associated with the occurrence of ADRs. In the effectiveness analysis set (n = 815), the incidence of progression of bone erosion was 8.7%. Steinbrocker stage and initial steroid dose were statistically significant risk factors associated with the progression of bone erosion. CONCLUSION Denosumab demonstrated safety and effectiveness over a 2 year period in RA patients without any new safety concerns.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- First Department of Internal Medicine, Faculty of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hideki Mizutani
- Post Marketing Study Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Kunimitsu Fujii
- Post Marketing Study Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Naoki Okubo
- Biostatistics & Data Management Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
2
|
Iwamoto N, Sato S, Furukawa K, Michitsuji T, Shiraishi K, Watanabe K, Chiba K, Osaki M, Kawakami A. Association of denosumab with serum cytokines, chemokines, and bone-related factors in patients with rheumatoid arthritis: A post hoc analysis of a multicentre, open-label, randomised, parallel-group study. Mod Rheumatol 2024; 34:936-946. [PMID: 38226481 DOI: 10.1093/mr/roae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/12/2023] [Accepted: 12/26/2023] [Indexed: 01/17/2024]
Abstract
OBJECTIVES To clarify changes in serum cytokines, chemokines, and bone-related factors during denosumab treatment in rheumatoid arthritis (RA) patients. METHODS This was a post hoc analysis of a multicentre, open-label, randomised, parallel-group study. Patients were randomly assigned to continue treatment with conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) plus receive treatment with denosumab (csDMARDs plus denosumab group) or to continue treatment with csDMARD therapy alone for 12 months. Serum biomarker levels were measured at baseline and at 6 and 12 months. RESULTS Baseline and 6-month data from the csDMARDs plus denosumab (n = 22) and csDMARD therapy alone (n = 22) groups were analysed. Statistically significant changes from baseline were seen: Dickkopf-related protein 1 decreased at 6 and 12 months (both groups); osteopontin decreased at 6 months in the csDMARDs plus denosumab group; osteopontin and soluble CD40 ligand increased at 6 and 12 months in the csDMARD therapy alone group; osteocalcin decreased at 6 and 12 months, epidermal growth factor decreased at 12 months, and macrophage-derived chemokine decreased at 6 months in the csDMARDs plus denosumab group; and interferon gamma-induced protein-10 increased at 12 months in the csDMARD therapy alone group. CONCLUSIONS Denosumab may inhibit bone destruction by suppressing bone-related factors/chemokines.
Collapse
Affiliation(s)
- Naoki Iwamoto
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shuntaro Sato
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Kaori Furukawa
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Toru Michitsuji
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuteru Shiraishi
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kounosuke Watanabe
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ko Chiba
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Makoto Osaki
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
3
|
Jeong YJ, Park SA, Park YH, Kim LK, Lee HR, Kim HJ, Heo TH. Anti-inflammatory effect of the combined treatment of LMT-28 and kaempferol in a collagen-induced arthritis mouse model. PLoS One 2024; 19:e0302119. [PMID: 39083495 PMCID: PMC11290667 DOI: 10.1371/journal.pone.0302119] [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: 03/28/2024] [Accepted: 05/31/2024] [Indexed: 08/02/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by joint inflammation and swelling. Several studies have demonstrated that RA fibroblast-like synovial cells (RA-FLS) play an important role in RA pathogenesis. Activated RA-FLS contribute to synovial inflammation by secreting inflammatory cytokines including interleukin (IL)-1β, IL-6 and tumor necrosis factor-α. LMT-28 is derivative of oxazolidone and exerts anti-inflammatory effects on RA via IL-6 signaling pathway regulation. LMT-28 also regulates T cell differentiation in RA condition. However, the effect of LMT-28 on the migration and invasion of RA-FLS remains unknown. Kaempferol has been reported to have pharmacological effects on various diseases, such as inflammatory diseases, autoimmune diseases, and cancer. Additionally, kaempferol has been reported to inhibit RA-FLS migration and invasion, but it is not known about the therapeutic mechanism including molecular mechanism such as receptor. The present study aimed to investigate the synergistic effects of the combined treatment of LMT-28 and kaempferol on RA-FLS activation and RA pathogenesis in mouse model. LMT-28 and kaempferol co-administration inhibited RA disease severity and histological collapse in the joint tissues of CIA mice, as well as downregulated the levels of pro-inflammatory cytokines in mouse serum. Additionally, the combined treatment inhibited excessive differentiation of T helper 17 cells and osteoclasts. Furthermore, compared with single treatments, combined treatment showed enhanced inhibitory effects on the hyperactivation of IL-6-induced signaling pathway in RA-FLS. Combined treatment also inhibited RA-FLS cell proliferation, migration, and invasion and suppressed the expression of matrix metalloproteinase in RA-FLS. Furthermore, we confirmed that the combined treatment inhibited chondrocyte proliferation, migration, and invasion. In conclusion, our results suggest that the combined treatment of LMT-28 and kaempferol exerts a synergistic effect on the RA development via the regulation of IL-6-induced hyperactivation of RA-FLS. Furthermore, this study suggests that combination therapies can be an effective therapeutic option for arthritis.
Collapse
Affiliation(s)
- Young-Jin Jeong
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Sun-Ae Park
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Yeon-Hwa Park
- Biowave, Anyangcheon-ro, Yangcheon-gu, Seoul, Republic of Korea
| | - Lee Kyung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Hae-Ri Lee
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Hee Jung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| | - Tae-Hwe Heo
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for Smart Pharma Leaders, College of Pharmacy, The Catholic University of Korea, Bucheon-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
4
|
Terashima A, Ono K, Omata Y, Tanaka S, Saito T. Inflammatory diseases causing joint and bone destruction: rheumatoid arthritis and hemophilic arthropathy. J Bone Miner Metab 2024; 42:455-462. [PMID: 38856919 DOI: 10.1007/s00774-024-01520-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
Various diseases and conditions cause joint disorders. Osteoarthritis (OA) is characterized by the degeneration of articular cartilage, synovitis, and anabolic changes in surrounding bone tissues. In contrast, rheumatoid arthritis (RA) and hemophilic arthropathy (HA) display marked destruction of bone tissues caused by synovitis. RA is a representative autoimmune disease. The primary tissue of RA pathogenesis is the synovial membrane and involves various immune cells that produce catabolic cytokines and enzymes. Hemophilia is a genetic disorder caused by a deficiency in blood clotting factors. Recurrent intra-articular bleeding leads to chronic synovitis through excessive iron deposition and results in the destruction of affected joints. Although the triggers for these two joint diseases are completely different, many cytokines and enzymes are common in the pathogenesis of both RA and HA. This review focuses on the similarities between joint and bone destruction in RA and HA. The insights may be useful in developing better treatments for hemophilia patients with arthropathy and osteoporosis by leveraging advanced therapeutics for RA.
Collapse
Affiliation(s)
- Asuka Terashima
- Bone and Cartilage Regenerative Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Kumiko Ono
- Department of Joint Surgery, Research Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo, 108-8639, Japan
- Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Yasunori Omata
- Bone and Cartilage Regenerative Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
- Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Sakae Tanaka
- Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Taku Saito
- Orthopaedic Surgery, Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
5
|
Mochizuki T, Yano K, Ikari K, Okazaki K. Effects of Locomotion Training on Bone Mineral Density in Patients with Rheumatoid Arthritis. Prog Rehabil Med 2024; 9:20240022. [PMID: 38895591 PMCID: PMC11180612 DOI: 10.2490/prm.20240022] [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: 02/01/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
Objectives This study aimed to evaluate the effects of locomotion training on bone mineral density (BMD) and the factors associated with increased BMD in patients with rheumatoid arthritis (RA). Methods We enrolled 85 patients with RA who underwent locomotion training for 6 months after receiving instructions from a physical therapist. We evaluated the BMD of the lumbar spine, total hip, and femoral neck 1 year before baseline (the start of locomotion training) and 1 year after baseline. Results The change in BMD from 1 year before baseline (non-exercise period) and 1 year after baseline (exercise period) were 0.1 ± 3.1% and 1.6 ± 3.7% (P=0.007) for the lumbar spine, -0.2 ± 2.4% and 1.0 ± 2.4% (P=0.005) for the total hip, and -0.6 ± 3.9% and 1.8 ± 3.5% (P<0.001) for the femoral neck, respectively. The Health Assessment Questionnaire Disability Index score at baseline was associated with increased BMD at the femoral neck. No factor was associated with increased BMD in the lumbar spine or total hip. Conclusions Locomotion training increased the BMD of the lumbar spine, total hip, and femoral neck during the exercise period compared with that during the non-exercise period. The current treatment for RA and osteoporosis accompanied by optional therapy with locomotion training might be effective in increasing BMD in patients with RA.
Collapse
Affiliation(s)
- Takeshi Mochizuki
- Department of Orthopaedic Surgery, Kamagaya General Hospital, Kamagaya, Japan
| | - Koichiro Yano
- Department of Orthopaedic Surgery, Tokyo Women’s Medical University, Tokyo, Japan
| | - Katsunori Ikari
- Department of Orthopaedic Surgery, Tokyo Women’s Medical University, Tokyo, Japan
| | - Ken Okazaki
- Department of Orthopaedic Surgery, Tokyo Women’s Medical University, Tokyo, Japan
| |
Collapse
|
6
|
Kaur C, Mishra Y, Kumar R, Singh G, Singh S, Mishra V, Tambuwala MM. Pathophysiology, diagnosis, and herbal medicine-based therapeutic implication of rheumatoid arthritis: an overview. Inflammopharmacology 2024; 32:1705-1720. [PMID: 38528307 PMCID: PMC11136810 DOI: 10.1007/s10787-024-01445-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/10/2024] [Indexed: 03/27/2024]
Abstract
Rheumatoid arthritis (RA) stands as an autoimmune disorder characterized by chronic joint inflammation, resulting in profound physiological alterations within the body. Affecting approximately 0.4-1.3% of the global population, this condition poses significant challenges as current therapeutic approaches primarily offer symptomatic relief, with the prospect of complete recovery remaining elusive. This review delves into the contemporary advancements in understanding the pathophysiology, diagnosis, and the therapeutic potential of herbal medicine in managing RA. Notably, early diagnosis during the initial stages emerges as the pivotal determinant for successful recovery post-treatment. Utilizing tools such as Magnetic Resonance Imaging (MRI), anti-citrullinated peptide antibody markers, and radiography proves crucial in pinpointing the diagnosis of RA with precision. Unveiling the intricate pathophysiological mechanisms of RA has paved the way for innovative therapeutic interventions, incorporating plant extracts and isolated phytoconstituents. In the realm of pharmacological therapy for RA, specific disease-modifying antirheumatic drugs have showcased commendable efficacy. However, this conventional approach is not without its drawbacks, as it is often associated with various side effects. The integration of methodological strategies, encompassing both pharmacological and plant-based herbal therapies, presents a promising avenue for achieving substantive recovery. This integrated approach not only addresses the symptoms but also strives to tackle the underlying causes of RA, fostering a more comprehensive and sustainable path towards healing.
Collapse
Affiliation(s)
- Charanjit Kaur
- 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
| | - Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Gurvinder Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Sukhraj Singh
- Department of Food Civil Supply and Consumer Affairs, Amritsar, 143001, Punjab, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India.
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, England, UK.
| |
Collapse
|
7
|
Al-Saoodi H, Kolahdooz F, Andersen JR, Jalili M. Effect of vitamin D on inflammatory and clinical outcomes in patients with rheumatoid arthritis: a systematic review and dose-response meta-analysis of randomized controlled trials. Nutr Rev 2024; 82:600-611. [PMID: 37437898 DOI: 10.1093/nutrit/nuad083] [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] [Indexed: 07/14/2023] Open
Abstract
CONTEXT Rheumatoid arthritis is a chronic inflammatory disease that causes synovitis. Vitamin D deficiency is common in rheumatoid arthritis. OBJECTIVE This systematic review and meta-analysis investigated whether vitamin D supplementation affects the inflammatory and clinical outcomes in patients with rheumatoid arthritis on the basis of randomized clinical trials. DATA SOURCES A literature search was performed in the Cochrane Central Register of Controlled Trials (CENTRAL), PubMed, MEDLINE, Embase, and Google Scholar for articles published until May 2022. DATA EXTRACTION The studies were selected according to PRISMA guidelines, and the risk of bias was assessed for randomized controlled trials. DATA ANALYSIS A random effects model was used to conduct a meta-analysis, and heterogeneity was assessed using the I2 statistic. Of 464 records, 11 studies were included from 3049 patients. Conclusion: Vitamin D supplementation did not significantly reduce C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), disease activity score in 28 joints (DAS28), or the health assessment questionnaire score; however, the response to supplementation was highly heterogeneous. The pooled analysis showed that vitamin D significantly reduced the pain-visual analogue scale (VAS) weighted mean difference (WMD = -1.30, 95% confidence interval [CI] [-2.34, -27], P = .01), DAS28-CRP (WMD = -.58, 95% CI [-.86, -.31], P < .0001), and DAS28-ESR (WMD = -.58, 95% CI [-.86, -.31], P = .0001). Subgroup analysis for vitamin D doses (>100 µg per day versus <100 µg per day) showed that the higher doses had a more significant effect on CRP than the lower doses (P < .05). CONCLUSIONS There was no significant difference between the effect of 2 vitamin D doses on ESR and DAS28. To minimize the high heterogeneity among studies in this meta-analysis, other confounding factors such as baseline vitamin D, age, dietary vitamin D, time of year, sun exposure, drug interaction, effect dosage, and power of study should be examined.
Collapse
Affiliation(s)
- Hagir Al-Saoodi
- Preventive and Clinical Nutrition Group, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Fariba Kolahdooz
- Indigenous Global Health Research Group, Department of Medicine, College of Health Sciences, University of Alberta, Edmonton, Canada
| | - Jens Rikardt Andersen
- Preventive and Clinical Nutrition Group, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Mahsa Jalili
- Preventive and Clinical Nutrition Group, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
8
|
Henning P, Kassem A, Westerlund A, Lundberg P, Engdahl C, Lionikaite V, Wikström P, Wu J, Li L, Lindholm C, de Souza PPC, Movérare-Skrtic S, Lerner UH. Toll-like receptor-2 induced inflammation causes local bone formation and activates canonical Wnt signaling. Front Immunol 2024; 15:1383113. [PMID: 38646530 PMCID: PMC11026618 DOI: 10.3389/fimmu.2024.1383113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/11/2024] [Indexed: 04/23/2024] Open
Abstract
It is well established that inflammatory processes in the vicinity of bone often induce osteoclast formation and bone resorption. Effects of inflammatory processes on bone formation are less studied. Therefore, we investigated the effect of locally induced inflammation on bone formation. Toll-like receptor (TLR) 2 agonists LPS from Porphyromonas gingivalis and PAM2 were injected once subcutaneously above mouse calvarial bones. After five days, both agonists induced bone formation mainly at endocranial surfaces. The injection resulted in progressively increased calvarial thickness during 21 days. Excessive new bone formation was mainly observed separated from bone resorption cavities. Anti-RANKL did not affect the increase of bone formation. Inflammation caused increased bone formation rate due to increased mineralizing surfaces as assessed by dynamic histomorphometry. In areas close to new bone formation, an abundance of proliferating cells was observed as well as cells robustly stained for Runx2 and alkaline phosphatase. PAM2 increased the mRNA expression of Lrp5, Lrp6 and Wnt7b, and decreased the expression of Sost and Dkk1. In situ hybridization demonstrated decreased Sost mRNA expression in osteocytes present in old bone. An abundance of cells expressed Wnt7b in Runx2-positive osteoblasts and ß-catenin in areas with new bone formation. These data demonstrate that inflammation, not only induces osteoclastogenesis, but also locally activates canonical WNT signaling and stimulates new bone formation independent on bone resorption.
Collapse
Affiliation(s)
- Petra Henning
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ali Kassem
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - Anna Westerlund
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Lundberg
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - Cecilia Engdahl
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Vikte Lionikaite
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pernilla Wikström
- Department of Medical Biosciences, Section of Pathology, Umeå University, Umeå, Sweden
| | - Jianyao Wu
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Lei Li
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Catharina Lindholm
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pedro P. C. de Souza
- Innovation in Biomaterials Laboratory, Federal University of Goiás, Goiania, Brazil
| | - Sofia Movérare-Skrtic
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Ulf H. Lerner
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| |
Collapse
|
9
|
Yokota K. Osteoclast differentiation in rheumatoid arthritis. Immunol Med 2024; 47:6-11. [PMID: 37309864 DOI: 10.1080/25785826.2023.2220931] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/30/2023] [Indexed: 06/14/2023] Open
Abstract
Osteoclasts, derived from the monocyte/macrophage line of bone marrow hematopoietic stem cell progenitors, are the sole bone-resorbing cells of the body. Conventional osteoclast differentiation requires macrophage colony-stimulating factor and receptor activator of nuclear factor kappa-B ligand (RANKL) signaling. Rheumatoid arthritis (RA) is the most prevalent systemic autoimmune disease and inflammatory arthritis characterized by bone destruction. Increased levels of proinflammatory cytokines, such as tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), in the serum and joints, cause excessive bone destruction. We have recently reported that stimulation of human peripheral blood monocytes with TNF-α and IL-6 induces the differentiation of osteoclasts with bone resorption activity. This review presents the functional differences between representative osteoclasts, conventional RANKL-induced osteoclasts, and recently identified proinflammatory cytokine (TNF-α and IL-6)-induced osteoclasts in RA patients. We believe novel pathological osteoclasts associated with RA will be identified, and new therapeutic strategies will be developed to target these osteoclasts and prevent the progression of bone destruction.
Collapse
Affiliation(s)
- Kazuhiro Yokota
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
10
|
Yang M, Zhu L. Osteoimmunology: The Crosstalk between T Cells, B Cells, and Osteoclasts in Rheumatoid Arthritis. Int J Mol Sci 2024; 25:2688. [PMID: 38473934 DOI: 10.3390/ijms25052688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/22/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Rheumatoid arthritis (RA) is an ongoing inflammatory condition that affects the joints and can lead to severe damage to cartilage and bones, resulting in significant disability. This condition occurs when the immune system becomes overactive, causing osteoclasts, cells responsible for breaking down bone, to become more active than necessary, leading to bone breakdown. RA disrupts the equilibrium between osteoclasts and osteoblasts, resulting in serious complications such as localized bone erosion, weakened bones surrounding the joints, and even widespread osteoporosis. Antibodies against the receptor activator of nuclear factor-κB ligand (RANKL), a crucial stimulator of osteoclast differentiation, have shown great effectiveness both in laboratory settings and actual patient cases. Researchers are increasingly focusing on osteoclasts as significant contributors to bone erosion in RA. Given that RA involves an overactive immune system, T cells and B cells play a pivotal role by intensifying the immune response. The imbalance between Th17 cells and Treg cells, premature aging of T cells, and excessive production of antibodies by B cells not only exacerbate inflammation but also accelerate bone destruction. Understanding the connection between the immune system and osteoclasts is crucial for comprehending the impact of RA on bone health. By delving into the immune mechanisms that lead to joint damage, exploring the interactions between the immune system and osteoclasts, and investigating new biomarkers for RA, we can significantly improve early diagnosis, treatment, and prognosis of this condition.
Collapse
Affiliation(s)
- Mei Yang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Lei Zhu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, Beijing 100005, China
| |
Collapse
|
11
|
Xu XX, Shao H, Wang QX, Wang ZY. Network Pharmacology and Experimental Validation Explore the Pharmacological Mechanisms of Herb Pair for Treating Rheumatoid Arthritis. Comb Chem High Throughput Screen 2024; 27:1808-1822. [PMID: 38213142 DOI: 10.2174/0113862073263839231129163200] [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: 06/23/2023] [Revised: 10/20/2023] [Accepted: 11/01/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVE This study aimed to elucidate the multitarget mechanism of the Mori Ramulus - Taxilli Herba (MT) herb pair in treating rheumatoid arthritis (RA). METHODS The targets of the herb pair and RA were predicted from databases and screened through cross-analysis. The core targets were obtained using protein-protein interaction (PPI) network analysis. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed. Finally, animal experiments were conducted to validate the anti-RA effect and mechanism of this herb pair. RESULTS This approach successfully identified 9 active compounds of MT that interacted with 6 core targets (AKT1, TNF, IL6, TP53, VEGFA, and IL1β). Pathway and functional enrichment analyses revealed that MT had significant effects on the TNF and IL-17 signaling pathways. The consistency of interactions between active components and targets in these pathways was confirmed through molecular docking. Moreover, the potential therapeutic effect of MT was verified in vivo, demonstrating its ability to effectively relieve inflammation by regulating these targeted genes and pathways. CONCLUSION The present work suggests that the therapeutic effect of MT herb pair on RA may be attributed to its ability to regulate the TNF signaling pathway and IL-17 signaling pathway.
Collapse
Affiliation(s)
- Xi-Xi Xu
- Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Hua Shao
- Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Qiao-Xue Wang
- Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, 210009, P. R. China
| | - Zi-Yuan Wang
- Public Experimental Platform, China Pharmaceutical University, Nanjing, 211100, P. R. China
| |
Collapse
|
12
|
Chiba K, Iwamoto N, Watanabe K, Shiraishi K, Saito K, Okubo N, Kawakami A, Osaki M. Denosumab improves bone mineral density and microarchitecture in rheumatoid arthritis: randomized controlled trial by HR-pQCT. J Bone Miner Metab 2023; 41:797-806. [PMID: 37480398 DOI: 10.1007/s00774-023-01452-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/17/2023] [Indexed: 07/24/2023]
Abstract
INTRODUCTION This pre-specified exploratory analysis investigated the effect of denosumab on bone mineral density (BMD) and bone microarchitecture in patients with rheumatoid arthritis (RA) treated with conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs). MATERIALS AND METHODS In this open-label, parallel-group study, patients were randomly assigned (1:1) to continuous treatment with csDMARDs plus denosumab or continuous treatment with csDMARD therapy alone for 12 months. BMD and bone microarchitecture were measured by dual-energy X-ray absorptiometry (DXA) and high-resolution peripheral quantitative computed tomography (HR-pQCT). RESULTS Of 46 patients enrolled in the primary study, 43 were included in the full analysis set. The mean age was 65.3 years, 88.4% were female, and 60.5% had osteoporosis. Areal BMD of the lumbar spine increased from baseline to 6 and 12 months in both groups, but the increase was higher in the csDMARDs plus denosumab group. Areal BMD of the total hip and femoral neck increased from baseline to 6 and 12 months only in the csDMARDs plus denosumab group. Cortical volumetric BMD and cortical thickness of the distal tibia increased in the csDMARDs plus denosumab group at 6 and 12 months but decreased in the csDMARD therapy alone group. Trabecular bone parameters of the distal tibia improved only in the csDMARDs plus denosumab group at 12 months. CONCLUSION Denosumab may be recommended for patients with RA treated with csDMARDs to increase BMD and improve bone microarchitecture.
Collapse
Affiliation(s)
- Ko Chiba
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Naoki Iwamoto
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kounosuke Watanabe
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuteru Shiraishi
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kengo Saito
- Primary Medical Science Department, Medical Affairs Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Naoki Okubo
- Data Intelligence Department, Digital Transformation Management Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Makoto Osaki
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
13
|
Stefania S, Rotondo C, Mele A, Trotta A, Cantatore FP, Corrado A. Role of denosumab in bone erosions in rheumatoid arthritis. Postgrad Med J 2023; 99:976-984. [PMID: 36841226 DOI: 10.1093/postmj/qgad013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/05/2022] [Accepted: 01/17/2023] [Indexed: 02/27/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by systemic inflammation and synovitis which evolve into joint destruction and deformity. Bone abnormalities are represented by marginal bone erosions and iuxta-articular and generalized osteoporosis. Overactivation of osteoclasts along with dysregulation of osteoblasts are the key events. Bone resorption is mediated by the receptor activator of nuclear factor (NF)-κB (RANK) ligand (RANK-L), responsible for the differentiation, proliferation, and activation of osteoclasts. RANK-L binds its receptor RANK, localized on the surface of preosteoclasts and mature osteoclasts promoting osteoclastogenesis. High levels of RANK-L were demonstrated in active RA patients. Denosumab, a fully human monoclonal antibody, binds RANK-L and suppresses the RANK-RANK-L signaling pathway leading to the inhibition of osteoclastogenesis. A retrospective analysis of published studies such as clinical trials evidenced the efficacy of denosumab in preventing bone erosion progression in RA patients. Key messages Key questions to answer in future include the following: Could denosumab be associated with other biologic therapies in RA patients? Could denosumab block the progression of bone damage in RA? Could denosumab be used for the prevention of bone erosion in RA?
Collapse
Affiliation(s)
- Silvia Stefania
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Cinzia Rotondo
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Angiola Mele
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Antonello Trotta
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Addolorata Corrado
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| |
Collapse
|
14
|
Kim SK, Kim JW, Lee H, Park SH, Choe JY, Kim B. The comparable efficacy of denosumab on bone mineral density in rheumatoid arthritis patients with postmenopausal osteoporosis: A retrospective case-control study. Medicine (Baltimore) 2023; 102:e34219. [PMID: 37390268 PMCID: PMC10313284 DOI: 10.1097/md.0000000000034219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/15/2023] [Indexed: 07/02/2023] Open
Abstract
Little is known about differences in the therapeutic efficacy of denosumab in subjects with and without rheumatoid arthritis (RA). This study compares the changes in bone mineral density (BMD) between RA patients and controls without RA who had been treated with denosumab for 2 years for postmenopausal osteoporosis. A total of 82 RA patients and 64 controls were enrolled, who were refractory to selective estrogen receptor modulators (SERMs) or bisphosphonates and completed the treatment of denosumab 60 mg for 2 years. The efficacy of denosumab in RA patients and controls was assessed using areal BMD (aBMD) and T-score of the lumbar spine, femur neck, and total hip. A general linear model with repeated measures analysis of variance was used to determine differences in aBMD and T-score between 2 study groups. No significant differences in percent changes in aBMD and T-scores by denosumab treatment for 2 years at the lumbar spine, femur neck, and total hip were evident between RA patients and controls (P > .05 of all), except T-score of the total hip (P = .034). Denosumab treatment equally increased aBMD at the lumbar spine and T-scores at the lumbar spine and total hip between RA patients and controls without statistical differences, but RA patients showed less improvement in aBMD at the femur neck (ptime*group = 0.032) and T-scores at the femur neck and total hip than controls (ptime*group = 0.004 of both). Changes in aBMD and T-scores after denosumab treatment in RA patients were not affected by previous use of bisphosphonates or SERMs. Differences of T-score at the femur neck among previous bisphosphonate users and aBMD and T-score at the femur neck and T-scores at the total hip were evident. This study revealed that 2 years of denosumab treatment in female RA patients achieved comparable efficacy on BMD to controls at the lumbar spine, but showed somewhat insufficient improvement at the femur neck and total hip.
Collapse
Affiliation(s)
- Seong-Kyu Kim
- Division of Rheumatology, Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Ji-Won Kim
- Division of Rheumatology, Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Hwajeong Lee
- Division of Rheumatology, Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Sung-Hoon Park
- Division of Rheumatology, Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Jung-Yoon Choe
- Division of Rheumatology, Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Boyoung Kim
- Division of Rheumatology, Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
15
|
Messina OD, Vidal M, Adami G, Vidal LF, Clark P, Torres JAM, Lems W, Zerbini C, Arguissain C, Reginster JY, Lane NE. Chronic arthritides and bone structure: focus on rheumatoid arthritis-an update. Aging Clin Exp Res 2023:10.1007/s40520-023-02432-9. [PMID: 37222927 DOI: 10.1007/s40520-023-02432-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/01/2023] [Indexed: 05/25/2023]
Abstract
Normal bone remodeling depends of a balance between bone forming cells, osteoblasts and bone resorbing cells, the osteoclasts. In chronic arthritides and some inflammatory and autoimmune diseases such as rheumatoid arthritis, there is a great constellation of cytokines produced by pannus that impair bone formation and stimulate bone resorption by inducing osteoclast differentiation and inhibiting osteoblast maturation. Patients with chronic inflammation have multiple causes that lead to low bone mineral density, osteoporosis and a high risk of fracture including circulating cytokines, impaired mobility, chronic administration of glucocorticoids, low vitamin D levels and post-menopausal status in women, among others. Biologic agents and other therapeutic measures to reach prompt remission might ameliorate these deleterious effects. In many cases, bone acting agents need to be added to conventional treatment to reduce the risk of fractures and to preserve articular integrity and independency for daily living activities. A limited number of studies related to fractures in chronic arthritides were published, and future investigation is needed to determine the risk of fractures and the protective effects of different treatments to reduce this risk.
Collapse
Affiliation(s)
- Osvaldo Daniel Messina
- Collaborating Centre WHO, Investigaciones Reumatológicas y Osteológicas (IRO), Buenos Aires, Argentina
- International Osteoporosis Foundation (IOF), Buenos Aires, Argentina
| | - Maritza Vidal
- Centro de Diagnóstico de Osteoporosis y Enfermedades Reumáticas (CEDOR), Lima, Peru.
| | - Giovanni Adami
- Rheumatology Section, Department of Medicine, University of Verona, Verona, Italy
| | - Luis Fernando Vidal
- Centro de Diagnóstico de Osteoporosis y Enfermedades Reumáticas (CEDOR), Lima, Peru
- Regional Advisory Council for Latin America - International Osteoporosis Foundation (IOF), Lima, Peru
| | - Patricia Clark
- International Osteoporosis Foundation (IOF), Buenos Aires, Argentina
- Chief of Clinical Epidemiology Unit-Hospital Federico Gomez School of Medicine UNAM, Mexico City, Mexico
| | | | - William Lems
- Department of Rheumatology, Amsterdam UMC, Location VU University Medical Centre Amsterdam, Amsterdam, North-Holland, The Netherlands
| | | | - Constanza Arguissain
- Collaborating Centre WHO, Investigaciones Reumatológicas y Osteológicas (IRO), Buenos Aires, Argentina
| | - Jean-Yves Reginster
- Division of Public Health, Epidemiology and Health Economics, WHO Collaborating Centre for Public Health, Aspects of Musculoskeletal Health and Ageing, University of Liege, Liege, Belgium
| | - Nancy E Lane
- Center for Musculoskeletal Health, University of California at Davis School of Medicine, Sacramento, CA, 95817, USA
| |
Collapse
|
16
|
Matsuda K, Shiba N, Hiraoka K. New Insights into the Role of Synovial Fibroblasts Leading to Joint Destruction in Rheumatoid Arthritis. Int J Mol Sci 2023; 24:ijms24065173. [PMID: 36982247 PMCID: PMC10049180 DOI: 10.3390/ijms24065173] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Rheumatoid arthritis (RA), one of the most common autoimmune diseases, is characterized by multiple-joint synovitis with subsequent destruction of bone and cartilage. The excessive autoimmune responses cause an imbalance in bone metabolism, promoting bone resorption and inhibiting bone formation. Preliminary studies have revealed that receptor activator of NF-κB ligand (RANKL)-mediated osteoclast induction is an important component of bone destruction in RA. Synovial fibroblasts are the crucial producers of RANKL in the RA synovium; novel analytical techniques, primarily, single-cell RNA sequencing, have confirmed that synovial fibroblasts include heterogeneous subsets of both pro-inflammatory and tissue-destructive cell types. The heterogeneity of immune cells in the RA synovium and the interaction of synovial fibroblasts with immune cells have recently received considerable attention. The current review focused on the latest findings regarding the crosstalk between synovial fibroblasts and immune cells, and the pivotal role played by synovial fibroblasts in joint destruction in RA.
Collapse
Affiliation(s)
- Kotaro Matsuda
- Department of Orthopedic Surgery, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Fukuoka, Japan
| | - Naoto Shiba
- Department of Orthopedic Surgery, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Fukuoka, Japan
| | - Koji Hiraoka
- Department of Orthopedic Surgery, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Fukuoka, Japan
| |
Collapse
|
17
|
Kobayakawa T, Miyazaki A, Kanayama Y, Hirano Y, Takahashi J, Suzuki T, Nakamura Y. Comparable efficacy of denosumab and romosozumab in patients with rheumatoid arthritis receiving glucocorticoid administration. Mod Rheumatol 2023; 33:96-103. [PMID: 35234889 DOI: 10.1093/mr/roac014] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/16/2021] [Accepted: 01/28/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Romosozumab is a newly released and widely known molecular-targeted drug for severe osteoporosis treatment with comparable effectiveness to denosumab. However, there have been no reports discussing the efficacy of those treatments for rheumatoid arthritis (RA) patients, especially those receiving glucocorticoids. This retrospective observational registry study compared the efficacy of 12-month treatment of denosumab and romosozumab in RA patients under the influence of glucocorticoid intake. METHODS Following propensity score matching, 36 patients each in the denosumab and romosozumab groups were analysed in this study. Drug effectiveness was evaluated by measuring bone mineral density (BMD) at the lumbar spine, total hip, and femoral neck at baseline, 6 and 12 months as well as alterations in P1NP, TRACP-5b, and simplified disease activity index (SDAI). The occurrence of adverse events and new fractures was also assessed. RESULTS At 12 months of treatment, BMD at the lumbar spine was increased by 7.5% in the denosumab group and 8.7% in the romosozumab group, which were both significantly and comparably elevated over baseline. At the total hip and femoral neck, romosozumab tended to exhibit favourable efficacy to increase BMD versus denosumab. Both P1NP and TRACP-5b were significantly lower in the denosumab group as compared with the baseline. Conversely in the romosozumab group, P1NP was increased over baseline, while TRACP-5b was decreased. Regarding SDAI alterations, both the romosozumab and denosumab groups exhibited comparable improvements in RA disease activity over time during treatment. Recorded adverse events and new fractures during treatment were few and minor in both groups. CONCLUSIONS Romosozumab exhibited comparable efficacy to denosumab for increasing BMD even under the influence of glucocorticoids for treating RA. Both drugs may be therefore suitable for managing osteoporosis in patients with RA and glucocorticoid intake.
Collapse
Affiliation(s)
| | - Akiko Miyazaki
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yasuhide Kanayama
- Department of Orthopedic Surgery and Rheumatology, Toyota Kosei Hospital, Toyota, Aichi, Japan
| | - Yuji Hirano
- Department of Rheumatology, Toyohashi Municipal Hospital, Toyohashi, Aichi, Japan
| | - Jun Takahashi
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takako Suzuki
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan.,Department of Human Nutrition, Faculty of Human Nutrition, Tokyo Kasei Gakuin University, Tokyo, Japan
| | - Yukio Nakamura
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| |
Collapse
|
18
|
Kawahito Y, Morinobu A, Kaneko Y, Kohno M, Hirata S, Kishimoto M, Seto Y, Sugihara T, Tanaka E, Ito H, Kojima T, Matsushita I, Nishida K, Mori M, Murashima A, Yamanaka H, Nakayama T, Kojima M, Harigai M. Drug treatment algorithm and recommendations from the 2020 update of the Japan College of Rheumatology clinical practice guidelines for the management of rheumatoid arthritis-secondary publication. Mod Rheumatol 2023; 33:21-35. [PMID: 35297492 DOI: 10.1093/mr/roac017] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/05/2022] [Accepted: 02/09/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The aim of this study was to update the Japan College of Rheumatology (JCR) clinical practice guidelines (CPG) for the management of rheumatoid arthritis (RA; JCR CPG for RA) according to recent changes in the medical environment in Japan. This article is a digest version of the guidance. METHODS We used the Grading of Recommendations, Assessment, Development, and Evaluation method to update the 2014 JCR CPG for RA. A consensus was formed by CPG panel members. RESULTS We identified 36 important clinical questions regarding drug treatment and developed corresponding recommendations for RA. The recommendations included the following RA medications: non-steroidal anti-inflammatory drugs, corticosteroids, conventional synthetic disease-modifying antirheumatic drugs, biological disease-modifying antirheumatic drugs, anti-receptor activator for nuclear factor-κB ligand antibodies, and Janus kinase inhibitors, as well as the tapering and discontinuation of these medications. Recommendations regarding the efficacy and safety of treatments in the elderly and patients with comorbidities were also developed. Finally, we used these recommendations to create an original algorithm for drug treatment for RA based on the Treat-to-Target approach. CONCLUSION The 2020 JCR CPG for RA provides a useful tool for rheumatologists, health care professionals, and patients with RA, enabling shared decision-making in a variety of clinical situations.
Collapse
Affiliation(s)
- Yutaka Kawahito
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuko Kaneko
- Department of Internal Medicine, Division of Rheumatology, Keio University School of Medicine, Tokyo, Japan
| | - Masataka Kohno
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shintaro Hirata
- Department of Clinical Immunology and Rheumatology, Hiroshima University Hospital, Hiroshima, Japan
| | - Mitsumasa Kishimoto
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Tokyo, Japan
| | - Yohei Seto
- Department of Rheumatology, Yachiyo Medical Center, Tokyo Women's Medical University, Yachiyo, Japan
| | - Takahiko Sugihara
- Division of Rheumatology and Allergology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Eiichi Tanaka
- Department of Internal Medicine, Division of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Hiromu Ito
- Department of Orthopedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Orthopaedic Surgery, Kurashiki Central Hospital, Kurashiki, Japan
| | - Toshihisa Kojima
- Department of Orthopedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Isao Matsushita
- Department of Rehabilitation Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Keiichiro Nishida
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama, Japan
| | - Masaaki Mori
- Division of Rheumatology and Allergology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Atsuko Murashima
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine/Japan Drug Information Institute in Pregnancy, National Center for Child Health and Development, Tokyo, Japan
| | | | - Takeo Nakayama
- Department of Health Informatics, Kyoto University School of Public Health, Kyoto, Japan
| | - Masayo Kojima
- Department of Frailty Research, Center for Gerontology and Social Science, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Masayoshi Harigai
- Department of Internal Medicine, Division of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Takeuchi T. Cytokines and cytokine receptors as targets of immune-mediated inflammatory diseases-RA as a role model. Inflamm Regen 2022; 42:35. [PMID: 36451227 PMCID: PMC9713106 DOI: 10.1186/s41232-022-00221-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/15/2022] [Indexed: 12/05/2022] Open
Abstract
Recent advances in our understanding in the immune-mediated inflammatory diseases (IMID) are explored and promoted by the targeted treatment. Among these targets, cytokines and cytokine receptors have become the good candidates for the drug development. In this review, the cytokine and cytokine receptors, which are approved in IMID, are overviewed, and modalities of the treatment, the role of cytokines and cytokine receptors in each disease, and the updated molecular information by modern technologies in rheumatoid arthritis as a role model are shown and discussed for the future perspectives.
Collapse
Affiliation(s)
- Tsutomu Takeuchi
- Saitama Medical University, 38 Morohongo, Moroyama, Iruma, Saitama, 350-0495, Japan.
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Keio University, 35 Shinanomachi, Shinjuku, Tokyo, 160-8582, Japan.
| |
Collapse
|
20
|
Kovalenko PS, Dydykina IS, Smirnov AV, Nasonov EL. Bone resorption during therapy with denosumab in patients with rheumatoid arthritis, positive for the main immunological markers. RHEUMATOLOGY SCIENCE AND PRACTICE 2022. [DOI: 10.47360/1995-4484-2022-573-579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objective. Inflammation in rheumatoid arthritis (RA) leads to the development of local and generalized bone loss. Rheumatoid factor (RF) and anti-cyclic citrullinated peptide antibodies (ACCP) are believed to play a negative role in the radiological progression of RA. The use of such antiresorptive therapy as denosumab – monoclonal antibodies to RANKL (receptor activator of nuclear factor kappa-B ligand), reduces the activity of osteoclasts, increases bone mineral density (BMD), and also potentially affects the erosive process at RA. The aim of the study is to evaluate the effect of denosumab therapy on BMD and erosion count in patients with RA and osteoporosis (OP) in consideration of the positivity in the RF and ACCP in serum and the baseline RA activity. Materials and methods. The 12-month prospective study of the efficacy of denosumab therapy (60 mg subcutaneously every 6 months) in patients with RA and OP included 66 postmenopausal women; age – 59.4±7.5 years, duration of RA – 17.8±10.6 years, RF-positive – 47 (72%) patients, ACCP-positive – 48 (74%) patients. At baseline and after 12 months, dual-energy X-ray absorptiometry was performed with an assessment of BMD in the lumbar spine (L1– L4), proximal femur (hip neck and total hip), distal forearm; X-ray of the hands and distal parts of the feet in direct projection, followed by assessment of erosive-destructive changes according to the Sharp/van der Heijde method. Results. A significant increase in BMD was established in all studied sites of the skeleton despite the positivity of the RF and ACCP (for the hip neck p=0.05), while a significant increase in BMD in the RF- and ACCP-negative group was detected only in L1–L4 site. The progression of the increase in erosion count was noted in the RF- and ACCP-positive group while in the RF- and ACCP-negative group this indicator did not change. Regardless of the baseline activity of RA (by DAS28 (Disease Activity Score 28)) the BMD of most parts of the skeleton were stabilized. In patients with moderate RA activity, BMD increased significantly in L1–L4 in both groups: RF- and ACCP-positive and -negative , as well as in the total hip – in RF- and ACCP-positive group. The dynamics of the erosion count in RA patients did not depend on the baseline degree of DAS28 activity, no significant changes were detected in the analyzed groups. Conclusions. RF and ACCP positivity in serum in patients with RA and OP treated with denosumab did not have a negative effect on the dynamics of BMD, while the number of erosions increased. The baseline RA activity level did not affect the dynamics of the erosion count and the dynamics of BMD in most subgroups – BMD levels have been increased or stabilized.
Collapse
Affiliation(s)
| | | | | | - E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology; I.M. Sechenov First Moscow State Medical University of the Ministry
of Health Care of Russian Federation (Sechenov University)
| |
Collapse
|
21
|
Hauser B, Raterman H, Ralston SH, Lems WF. The Effect of Anti-rheumatic Drugs on the Skeleton. Calcif Tissue Int 2022; 111:445-456. [PMID: 35771255 PMCID: PMC9560949 DOI: 10.1007/s00223-022-01001-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/11/2022] [Indexed: 12/27/2022]
Abstract
The therapeutic armamentarium for rheumatoid arthritis has increased substantially over the last 20 years. Historically antirheumatic treatment was started late in the disease course and frequently included prolonged high-dose glucocorticoid treatment which was associated with accelerated generalised bone loss and increased vertebral and non-vertebral fracture risk. Newer biologic and targeted synthetic treatments and a combination of conventional synthetic DMARDs prevent accelerated systemic bone loss and may even allow repair of cortical bone erosions. Emerging data also gives new insight on the impact of long-term conventional synthetic DMARDs on bone health and fracture risk and highlights the need for ongoing studies for better understanding of "established therapeutics". An interesting new antirheumatic treatment effect is the potential of erosion repair with the use of biologic DMARDs and janus kinase inhibitors. Although several newer anti-rheumatic drugs seem to have favorable effects on bone mineral density in RA patients, these effects are modest and do not seem to influence the fracture risk thus far. We summarize recent developments and findings of the impact of anti-rheumatic treatments on localized and systemic bone integrity and health.
Collapse
Affiliation(s)
- B Hauser
- Rheumatic Disease Unit, Western General Hospital, Edinburgh, UK.
- Rheumatology and Bone Disease Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| | - H Raterman
- Department of Rheumatology, Northwest Clinics, Alkmaar, The Netherlands
| | - S H Ralston
- Rheumatic Disease Unit, Western General Hospital, Edinburgh, UK
- Rheumatology and Bone Disease Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - W F Lems
- Amsterdam Rheumatology and Immunology Centre, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Okawa T, Okawa M, Koike T. Risk factors for poor response to denosumab treatment in Japanese postmenopausal women with osteoporosis. J Bone Miner Metab 2022; 40:960-967. [PMID: 35939236 DOI: 10.1007/s00774-022-01357-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/05/2022] [Indexed: 10/15/2022]
Abstract
INTRODUCTION Denosumab has been reported to increase bone mineral density (BMD) and suppress fractures, but poor responders are not uncommon. This study aimed to identify risk factors for poor response to denosumab treatment. This is the first study to explore risk factors for poor response to denosumab. MATERIALS AND METHODS This retrospective observational study investigated 227 Japanese postmenopausal women who received denosumab with monitoring of BMD by dual-energy X-ray absorptiometry at 6-month intervals. Risk factors were identified using Cox's proportional hazard modeling. Poor responders were defined as not exceeding the least significant change of BMD from baseline for 3 years. RESULTS Mean relative change from baseline for 3 years in lumbar spine (LS)-BMD, femoral neck (FN)-BMD, and total hip (TH)-BMD were 12.6%, 6.8%, and 6.1%, respectively. Numbers of poor responders were 10 in LS-BMD, 47 in FN-BMD, 38 in TH-BMD. Risk factors for poor response were concomitant glucocorticoid use for LS-BMD, low body mass index or initiation at higher BMD for FN-BMD, and pretreatment with bisphosphonates or initiation at higher BMD for TH-BMD. CONCLUSION Risk factors for insufficient denosumab effect differed between BMD measurement sites. These results should be taken into consideration when selecting denosumab in clinical practice.
Collapse
Affiliation(s)
- Tokutaro Okawa
- Okawa Orthopaedic Surgery Hospital, Domyojimachi 6-12-34, Fujiidera, Osaka, 583-0012, Japan.
| | - Motomi Okawa
- Okawa Orthopaedic Surgery Hospital, Domyojimachi 6-12-34, Fujiidera, Osaka, 583-0012, Japan
| | - Tatsuya Koike
- Center for Senile Degenerative Disorders (CSDD), Osaka Metropolitan University Graduate School of Medicine, Abenoku Asahimachi 1-4-3, Osaka, 545-8585, Japan
- Search Institute for Bone and Arthritis Disease (SINBAD), Shirahama Foundation for Health and Welfare, Nishimurogun Shirahamacho 1447, Wakayama, 649-2211, Japan
| |
Collapse
|
23
|
Nawata T, Sakai H, Honda T, Otsuka M, Fujita H, Uchinoumi H, Kobayashi S, Yamamoto T, Asagiri M, Yano M. Dantrolene, a stabilizer of the ryanodine receptor, prevents collagen-induced arthritis. Biochem Biophys Res Commun 2022; 624:141-145. [PMID: 35940127 DOI: 10.1016/j.bbrc.2022.07.111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022]
Abstract
Dantrolene inhibits Ca2+ leakage from destabilized ryanodine receptors and therefore may serve as a therapeutic agent against endoplasmic reticulum stress-associated diseases. However, its effectiveness in treating autoimmune diseases remains unclear. Here, we investigated the effect of dantrolene on collagen-induced arthritis (CIA) in mice. Oral administration of dantrolene resulted in significantly lower arthritic scores in both male and female CIA mice than in the control mice. Micro-computed tomographic and histological analyses showed that dantrolene suppressed bone and chondral destruction. The serum levels of anti-type II collagen (CII) IgG were positively correlated with the arthritic scores (r = 0.704, p < 0.01). In addition, the serum levels of anti-CII IgG were significantly lower in the dantrolene group than in the control group (p < 0.05). These results demonstrate that oral administration of dantrolene to CIA mice inhibits the production of serum anti-CII IgG and consequently prevents arthritis. Therefore, dantrolene may be a potential anti-rheumatic drug.
Collapse
Affiliation(s)
- Takashi Nawata
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan.
| | - Hiroki Sakai
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan
| | - Takeshi Honda
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan
| | - Marina Otsuka
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan
| | - Hina Fujita
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan
| | - Hitoshi Uchinoumi
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan
| | - Shigeki Kobayashi
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan
| | - Takeshi Yamamoto
- Faculty of Health Sciences, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan
| | - Masataka Asagiri
- Department of Pharmacology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan
| | - Masafumi Yano
- Department of Medicine and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, 755-8505, Japan
| |
Collapse
|
24
|
Andreev D, Kachler K, Schett G, Bozec A. Rheumatoid arthritis and osteoimmunology: The adverse impact of a deregulated immune system on bone metabolism. Bone 2022; 162:116468. [PMID: 35688359 DOI: 10.1016/j.bone.2022.116468] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 11/20/2022]
Abstract
The term osteoimmunology describes an interdisciplinary research field that links the investigation of osteology (bone cells) with immunology. The crosstalk between innate and adaptive immune cells and cells involved in bone remodeling, mainly bone-resorbing osteoclasts and bone-forming osteoblasts, becomes particularly obvious in the inflammatory autoimmune disease rheumatoid arthritis (RA). Besides striking inflammation of the joints, RA causes bone loss, leading to joint damage and disabilities as well as generalized osteoporosis. Mechanistically, RA-associated immune cells (macrophages, T cells, B cells etc.) produce high levels of pro-inflammatory cytokines, receptor activator of nuclear factor κB ligand (RANKL) and autoantibodies that promote bone degradation and at the same time counteract new bone formation. Today, antirheumatic therapy effectively ceases joint inflammation and arrests bone erosion. However, the repair of established bone lesions still presents a challenging task and requires improved treatment options. In this review, we outline the knowledge gained over the past years about the immunopathogenesis of RA and the impact of a dysregulated immune system on bone metabolism.
Collapse
Affiliation(s)
- Darja Andreev
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Katerina Kachler
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany.
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany.
| |
Collapse
|
25
|
ETS1 is a key transcription factor that drives RANKL-expressing, tissue-destructive fibroblasts. Nat Immunol 2022; 23:1303-1304. [PMID: 35999397 DOI: 10.1038/s41590-022-01298-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Yan M, Komatsu N, Muro R, Huynh NCN, Tomofuji Y, Okada Y, Suzuki HI, Takaba H, Kitazawa R, Kitazawa S, Pluemsakunthai W, Mitsui Y, Satoh T, Okamura T, Nitta T, Im SH, Kim CJ, Kollias G, Tanaka S, Okamoto K, Tsukasaki M, Takayanagi H. ETS1 governs pathological tissue-remodeling programs in disease-associated fibroblasts. Nat Immunol 2022; 23:1330-1341. [PMID: 35999392 DOI: 10.1038/s41590-022-01285-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 07/08/2022] [Indexed: 02/04/2023]
Abstract
Fibroblasts, the most abundant structural cells, exert homeostatic functions but also drive disease pathogenesis. Single-cell technologies have illuminated the shared characteristics of pathogenic fibroblasts in multiple diseases including autoimmune arthritis, cancer and inflammatory colitis. However, the molecular mechanisms underlying the disease-associated fibroblast phenotypes remain largely unclear. Here, we identify ETS1 as the key transcription factor governing the pathological tissue-remodeling programs in fibroblasts. In arthritis, ETS1 drives polarization toward tissue-destructive fibroblasts by orchestrating hitherto undescribed regulatory elements of the osteoclast differentiation factor receptor activator of nuclear factor-κB ligand (RANKL) as well as matrix metalloproteinases. Fibroblast-specific ETS1 deletion resulted in ameliorated bone and cartilage damage under arthritic conditions without affecting the inflammation level. Cross-tissue fibroblast single-cell data analyses and genetic loss-of-function experiments lent support to the notion that ETS1 defines the perturbation-specific fibroblasts shared among various disease settings. These findings provide a mechanistic basis for pathogenic fibroblast polarization and have important therapeutic implications.
Collapse
Affiliation(s)
- Minglu Yan
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Noriko Komatsu
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryunosuke Muro
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nam Cong-Nhat Huynh
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,Laboratory of Oral-Maxillofacial Biology, Faculty of Odonto-Stomatology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Yoshihiko Tomofuji
- Department of Statistical Genetics, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University, Graduate School of Medicine, Osaka, Japan.,Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Hiroshi I Suzuki
- Division of Molecular Oncology, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Hiroyuki Takaba
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Riko Kitazawa
- Division of Diagnostic Pathology, Ehime University Hospital, Toon City, Japan
| | - Sohei Kitazawa
- Department of Molecular Pathology, Graduate School of Medicine, Ehime University, Toon City, Japan
| | - Warunee Pluemsakunthai
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuichi Mitsui
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Innate Cell Therapy, Osaka, Japan
| | - Takashi Satoh
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Innate Cell Therapy, Osaka, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), POSTECH Biotech Center, Pohang, Republic of Korea.,ImmunoBiome, Pohang, Republic of Korea.,Institute of Convergence Science, Yonsei University, Seoul, Republic of Korea
| | - Chan Johng Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), POSTECH Biotech Center, Pohang, Republic of Korea
| | - George Kollias
- Institute for Bioinnovation, Biomedical Sciences Research Center (BSRC) 'Alexander Fleming,' Vari, Attika, Greece.,Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masayuki Tsukasaki
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
27
|
Rodríguez-Trillo A, Pena C, García S, Pérez-Pampín E, Rodríguez-López M, Mera-Varela A, González A, Conde C. ROCK inhibition with Y-27632 reduces joint inflammation and damage in serum-induced arthritis model and decreases in vitro osteoclastogenesis in patients with early arthritis. Front Immunol 2022; 13:858069. [PMID: 36032152 PMCID: PMC9410766 DOI: 10.3389/fimmu.2022.858069] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
Rheumatoid arthritis (RA) is a common chronic inflammatory disease affecting primarily peripheral joints, which is only partially controlled with current treatments. RA leads to pain, disability, deformities, and life expectancy shortening. Its pathogenesis is complex involving multiple cell types and signaling pathways that we incompletely understand. One of the pathways we have elucidated starts with WNT5A signaling and contributes to the aggressive phenotype of the RA synoviocytes through RYK-RhoA/ROCK signaling. Now, we have explored the contribution of ROCK to arthritis in vivo, using the K/BxN serum-transfer arthritis model; and to osteoclastogenesis, using the arthritis model and cells from patients with inflammatory arthritis. The mice and cells were treated with the ROCK inhibitor Y-27632 that caused a significant improvement of arthritis and reduction of osteoclastogenesis. The improvement in mouse arthritis was observed in the clinical evaluation and, histologically, in synovial inflammation, cartilage damage, bone erosion, and the abundance of multinucleated TRAP+ cells. Expression of inflammatory mediators in the arthritic joints, as assessed by real-time PCR, was also significantly reduced. The effect on bone was confirmed with in vitro assays using bone marrow precursors of arthritic mice and peripheral blood monocytes of patients with inflammatory arthritis. These assays showed dramatically reduced osteoclastogenesis and bone resorption. Overall, our findings suggest that ROCK inhibition could be part of a therapeutic strategy for RA by its dual action on inflammation and bone erosion.
Collapse
Affiliation(s)
- Angela Rodríguez-Trillo
- Laboratorio de Reumatología Experimental y Observacional y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Carmen Pena
- Laboratorio de Reumatología Experimental y Observacional y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Samuel García
- Laboratorio de Reumatología y Enfermedades Inmunomediadas (IRIDIS), Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur), Hospital Álvaro Cunqueiro, Vigo, Spain
| | - Eva Pérez-Pampín
- Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Marina Rodríguez-López
- Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Antonio Mera-Varela
- Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Antonio González
- Laboratorio de Reumatología Experimental y Observacional y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Carmen Conde
- Laboratorio de Reumatología Experimental y Observacional y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
- *Correspondence: Carmen Conde,
| |
Collapse
|
28
|
Komatsu N, Takayanagi H. Mechanisms of joint destruction in rheumatoid arthritis - immune cell-fibroblast-bone interactions. Nat Rev Rheumatol 2022; 18:415-429. [PMID: 35705856 DOI: 10.1038/s41584-022-00793-5] [Citation(s) in RCA: 194] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2022] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is characterized by inflammation and destruction of bone and cartilage in affected joints. Autoimmune responses lead to increased osteoclastic bone resorption and impaired osteoblastic bone formation, the imbalance of which underlies bone loss in RA, which includes bone erosion, periarticular bone loss and systemic osteoporosis. The crucial role of osteoclasts in bone erosion has been demonstrated in basic studies as well as by the clinical efficacy of antibodies targeting RANKL, an important mediator of osteoclastogenesis. Synovial fibroblasts contribute to joint damage by stimulating both pro-inflammatory and tissue-destructive pathways. New technologies, such as single-cell RNA sequencing, have revealed the heterogeneity of synovial fibroblasts and of immune cells including T cells and macrophages. To understand the mechanisms of bone damage in RA, it is important to clarify how the immune system promotes the tissue-destructive properties of synovial fibroblasts and influences bone cells. The interaction between immune cells and fibroblasts underlies the imbalance between regulatory T cells and T helper 17 cells, which in turn exacerbates not only inflammation but also bone destruction, mainly by promoting RANKL expression on synovial fibroblasts. An improved understanding of the immune mechanisms underlying joint damage and the interplay between the immune system, synovial fibroblasts and bone will contribute to the identification of novel therapeutic targets in RA.
Collapse
Affiliation(s)
- Noriko Komatsu
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
29
|
Mochizuki T, Yano K, Ikari K, Hiroshima R, Okazaki K. Comparison of Romosozumab Versus Denosumab Treatment on Bone Mineral Density After One Year in Rheumatoid Arthritis Patients with Severe Osteoporosis: A Randomized Clinical Pilot Study. Mod Rheumatol 2022; 33:490-495. [PMID: 35689558 DOI: 10.1093/mr/roac059] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVES To investigate the effect of romosozumab versus denosumab treatment on bone mineral density (BMD), disease activity, and joint damage in patients with rheumatoid arthritis (RA) and severe osteoporosis. METHODS Fifty-one postmenopausal women were enrolled and randomized equally into two groups to receive either romosozumab or the denosumab. Changes (Δ) in the BMD (at lumbar spine, total hip, and femoral neck), disease activity score in 28 joints (DAS28)-erythrocyte sedimentation rate (ESR), and van der Heijde-modified Total Sharp Score (TSS) from baseline to 12 months after treatment were evaluated. RESULTS The ΔBMD at 12 months in the romosozumab and denosumab groups were 10.2 ± 5.6% and 5.0 ± 3.1% (p = 0.002) for the lumbar spine, 3.7 ± 4.9% and 3.5 ± 3.0% (p = 0.902) for total hip, and 3.6 ± 4.7% and 3.2 ± 4.9% (p = 0.817) for femoral neck, respectively. The ΔDAS28-ESR at 12 months in the romosozumab and denosumab groups was 0.14 and 0.22 (p = 0.643), respectively, whereas, the ΔTSS at 12 months was 0.33 and 0.29 (p = 0.927), respectively. CONCLUSIONS Our results suggest that romosozumab treatment was more effective in increasing the BMD at the lumbar spine than denosumab, and may be selected for patients who require a significant increase in the lumbar spine BMD. Moreover, romosozumab may be not affect disease activity and joint damage in patients with RA.
Collapse
Affiliation(s)
- Takeshi Mochizuki
- Department of Orthopaedic Surgery, Kamagaya General Hospital, Chiba, Japan
| | - Koichiro Yano
- Department of Orthopaedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Katsunori Ikari
- Department of Orthopaedic Surgery, Tokyo Women's Medical University, Tokyo, Japan.,Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women's Medical University, Tokyo, Japan
| | - Ryo Hiroshima
- Department of Orthopaedic Surgery, Kamagaya General Hospital, Chiba, Japan
| | - Ken Okazaki
- Department of Orthopaedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
30
|
Personalized Therapeutic Strategies in the Management of Osteoporosis in Patients with Autoantibody-Positive Rheumatoid Arthritis. J Clin Med 2022; 11:jcm11092341. [PMID: 35566466 PMCID: PMC9104810 DOI: 10.3390/jcm11092341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/02/2022] [Accepted: 04/20/2022] [Indexed: 12/22/2022] Open
Abstract
Bone mineral density (BMD) reduction and fragility fractures still represent a major source of morbidity in rheumatoid arthritis (RA) patients, despite adequate control of the disease. An increasing number of clinical and experimental evidence supports the role of autoantibodies, especially anti-citrullinated protein antibodies (ACPAs), in causing localized and generalised bone loss in ways that are both dependent on and independent of inflammation and disease activity. The human receptor activator of nuclear factor kappa B and its ligand—the so-called RANK-RANKL pathway—is known to play a key role in promoting osteoclasts’ activation and bone depletion, and RANKL levels were shown to be higher in ACPA-positive early untreated RA patients. Thus, ACPA-positivity can be considered a specific risk factor for systemic and periarticular bone loss. Through the inhibition of the RANK-RANKL system, denosumab is the only antiresorptive drug currently available that exhibits both a systemic anti-osteoporotic activity and a disease-modifying effect when combined with conventional synthetic or biologic disease-modifying anti-rheumatic drugs (DMARDs). Thus, the combination of DMARD and anti-RANKL therapy could be beneficial in the prevention of fragility fractures and structural damage in the subset of RA patients at risk of radiographic progression, as in the presence of ACPAs.
Collapse
|
31
|
Morikawa N, Kato Y, Takeshita N, Shimizu Y. Pharmacological characterization of AS2690168, a novel small molecule RANKL signal transduction inhibitor. Eur J Pharmacol 2022; 924:174941. [PMID: 35398031 DOI: 10.1016/j.ejphar.2022.174941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022]
Abstract
Pathological osteolysis is associated with excessive bone resorption by activated osteoclasts. Given that receptor activator of NF-kB and its ligand (RANKL) are key players in the differentiation and activation of osteoclasts, the RANKL/RANK signaling pathway is considered a promising target for the development of effective osteoclastogenesis inhibitors. We previously found that the orally available compound, AS2690168, suppresses RANKL-induced osteoclastogenesis of RAW264 cells. In this report, we further characterized the pharmacological profiles of AS2690168 in vitro and in vivo. AS2690168 suppressed soluble RANKL (sRANKL)-induced NFATc1 mRNA expression in RAW264 cells at 0.3 and 3.0 μM. It also suppressed calcium release from parathyroid hormone-stimulated mouse calvaria with an IC50 value of 0.46 μM. Oral administration of AS2690168 completely suppressed the decrease in femoral bone mineral content in an sRANKL-induced osteopenic mice model at 3.0 mg/kg. It also significantly suppressed the decrease in femoral bone mineral density and increase in serum tartrate-resistant acid phosphatase-5b levels in ovariectomized rats at doses of 0.3, 1 and 3 mg/kg. Finally, AS260168 suppressed the increase in urine deoxypyridinoline in a rat prednisolone-induced osteoporosis model at 10 mg/kg. These results suggest that AS2690168 is a promising treatment for bone disorders with excessive bone resorption.
Collapse
Affiliation(s)
- Noriyuki Morikawa
- Drug Discovery Research, Astellas Pharma Inc. 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - Yasuko Kato
- Drug Discovery Research, Astellas Pharma Inc. 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| | - Nobuaki Takeshita
- Drug Discovery Research, Astellas Pharma Inc. 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan.
| | - Yasuaki Shimizu
- Drug Discovery Research, Astellas Pharma Inc. 21 Miyukigaoka, Tsukuba, Ibaraki, 305-8585, Japan
| |
Collapse
|
32
|
Ebeling PR, Nguyen HH, Aleksova J, Vincent AJ, Wong P, Milat F. Secondary Osteoporosis. Endocr Rev 2022; 43:240-313. [PMID: 34476488 DOI: 10.1210/endrev/bnab028] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 02/07/2023]
Abstract
Osteoporosis is a global public health problem, with fractures contributing to significant morbidity and mortality. Although postmenopausal osteoporosis is most common, up to 30% of postmenopausal women, > 50% of premenopausal women, and between 50% and 80% of men have secondary osteoporosis. Exclusion of secondary causes is important, as treatment of such patients often commences by treating the underlying condition. These are varied but often neglected, ranging from endocrine to chronic inflammatory and genetic conditions. General screening is recommended for all patients with osteoporosis, with advanced investigations reserved for premenopausal women and men aged < 50 years, for older patients in whom classical risk factors for osteoporosis are absent, and for all patients with the lowest bone mass (Z-score ≤ -2). The response of secondary osteoporosis to conventional anti-osteoporosis therapy may be inadequate if the underlying condition is unrecognized and untreated. Bone densitometry, using dual-energy x-ray absorptiometry, may underestimate fracture risk in some chronic diseases, including glucocorticoid-induced osteoporosis, type 2 diabetes, and obesity, and may overestimate fracture risk in others (eg, Turner syndrome). FRAX and trabecular bone score may provide additional information regarding fracture risk in secondary osteoporosis, but their use is limited to adults aged ≥ 40 years and ≥ 50 years, respectively. In addition, FRAX requires adjustment in some chronic conditions, such as glucocorticoid use, type 2 diabetes, and HIV. In most conditions, evidence for antiresorptive or anabolic therapy is limited to increases in bone mass. Current osteoporosis management guidelines also neglect secondary osteoporosis and these existing evidence gaps are discussed.
Collapse
Affiliation(s)
- Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia
| | - Hanh H Nguyen
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Department of Endocrinology and Diabetes, Western Health, Victoria 3011, Australia
| | - Jasna Aleksova
- Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Amanda J Vincent
- Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Monash Centre for Health Research and Implementation, School of Public Health and Preventative Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Phillip Wong
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Frances Milat
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| |
Collapse
|
33
|
Yagita M, Morita T, Kumanogoh A. Therapeutic efficacy of denosumab for rheumatoid arthritis: a systematic review and meta-analysis. Rheumatol Adv Pract 2022; 5:rkab099. [PMID: 34988358 PMCID: PMC8693364 DOI: 10.1093/rap/rkab099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/28/2021] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES Denosumab is used for osteoporosis because it inhibits osteoclast maturation and suppresses bone resorption. Although denosumab is expected to inhibit the bone erosion in RA, its therapeutic efficacy is not well established. The aim of this study was to estimate the effects of denosumab on RA through a meta-analysis. METHODS A systematic review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses. PubMed, Web of Science and Scopus were searched for original studies providing information on BMD, joint destruction and disease activity in denosumab-treated RA. A random-effects model was used in the meta-analysis. RESULTS Of the 367 studies identified, 18 met the selection criteria. The BMDs of the lumbar spine, total hip and femoral neck at 12 months after denosumab treatment increased by 5.27% (95% CI: 4.37, 6.18), 2.82% (2.46, 3.18) and 3.07% (2.66, 3.48), respectively. In the sensitivity analysis, age and sex tended to influence the effect of denosumab therapy on the rate of variation of BMD, but not glucocorticoid use. The changes in the modified total sharp, erosion and joint space narrowing scores at 12 months after denosumab treatment were significantly smaller with denosumab than with placebo, although the DAS did not change after denosumab treatment. CONCLUSION Although denosumab has an inhibitory effect on the bone resorption in RA, its effects might be influenced by the age and sex of RA patients, but not by glucocorticoid use.
Collapse
Affiliation(s)
- Mayu Yagita
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine
| | - Takayoshi Morita
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine.,Laboratory of Immunopathology, World Premier International Immunology Frontier Research Center.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University.,Center for Infectious Disease for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
34
|
Hu Q, Zhong X, Tian H, Liao P. The Efficacy of Denosumab in Patients With Rheumatoid Arthritis: A Systematic Review and Pooled Analysis of Randomized or Matched Data. Front Immunol 2022; 12:799575. [PMID: 35069583 PMCID: PMC8766643 DOI: 10.3389/fimmu.2021.799575] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/13/2021] [Indexed: 12/19/2022] Open
Abstract
Objective The purpose of this study was to evaluate the efficacy of denosumab treatment in patients with rheumatoid arthritis (RA). Methods The Medline, Embase and Cochrane Library databases were searched for relevant clinical studies. Studies that assessed the efficacy of denosumab in patients with RA were identified. The primary endpoints were the percent changes in bone mineral density (BMD), and the changes in modified total Sharp score (mTSS), modified Sharp erosion score and joint space narrowing (JSN) score. Pooled analyses were calculated using random-effect models. Results After searching the literature and performing further detailed assessments, 10 studies with a total of 1758 patients were included in the quantitative analysis. Pooled analyses showed that denosumab treatment significantly increased the percent changes in lumbar spine BMD [mean difference (MD): 5.12, confidence intervals (CI): 4.15 to 6.09], total hip BMD (MD: 2.72, 95% CI: 1.80 to 3.64) and femoral neck BMD (MD: 2.20, 95% CI: 0.94 to 3.46) compared with controls. Moreover, denosumab treatment significantly decreased the changes in mTSS (MD: -0.63, 95% CI: -0.86 to -0.41) and modified Sharp erosion score (MD: -0.62, 95% CI: -0.88 to -0.35). Subgroup analysis indicated that denosumab was superior to bisphosphonates for the improvement of BMD and the mitigation of joint destruction. Conclusion Denosumab treatment was associated with increased BMD and alleviated progression of joint destruction in RA patients, even when compared with bisphosphonates.
Collapse
Affiliation(s)
- Qiongwen Hu
- Department of Medical Laboratory, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Xue Zhong
- Department of Nephrology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Hua Tian
- Department of Medical Laboratory, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| | - Pu Liao
- Department of Medical Laboratory, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| |
Collapse
|
35
|
Iwamoto N, Chiba K, Sato S, Shiraishi K, Watanabe K, Oki N, Okada A, Koga T, Kawashiri SY, Tamai M, Hosogaya N, Furuyama M, Kobayashi M, Saito K, Okubo N, Uetani M, Osaki M, Kawakami A. Inhibition of bone erosion, determined by high-resolution peripheral quantitative computed tomography (HR-pQCT), in rheumatoid arthritis patients receiving a conventional synthetic disease-modifying anti-rheumatic drug (csDMARD) plus denosumab vs csDMARD therapy alone: an open-label, randomized, parallel-group study. Arthritis Res Ther 2022; 24:264. [PMID: 36476479 PMCID: PMC9727996 DOI: 10.1186/s13075-022-02957-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/20/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND This exploratory study compared the inhibition of bone erosion progression in rheumatoid arthritis (RA) patients treated with a conventional synthetic disease-modifying anti-rheumatic drug (csDMARD) plus denosumab versus csDMARD therapy alone and investigated the effects of denosumab on bone micro-architecture and other bone-related parameters using high-resolution peripheral quantitative computed tomography (HR-pQCT). METHODS In this open-label, randomized, parallel-group study, patients with RA undergoing treatment with a csDMARD were randomly assigned (1:1) to continue csDMARD therapy alone or to continue csDMARDs with denosumab (60-mg subcutaneous injection once every 6 months) for 12 months. The primary endpoint was the change from baseline in the depth of bone erosion, measured by HR-pQCT, in the second and third metacarpal heads at 6 months after starting treatment. Exploratory endpoints were also evaluated, and adverse events (AEs) were monitored for safety. RESULTS In total, 46 patients were enrolled, and 43 were included in the full analysis set (csDMARDs plus denosumab, N = 21; csDMARD therapy alone, N = 22). Most patients were female (88.4%), and the mean age was 65.3 years. The adjusted mean (95% confidence interval) change from baseline in the depth of bone erosion, measured by HR-pQCT, in the 2-3 metacarpal heads at 6 months was - 0.57 mm (- 1.52, 0.39 mm) in the csDMARDs plus denosumab group vs - 0.22 mm (- 0.97, 0.53 mm) in the csDMARD therapy alone group (between-group difference: - 0.35 mm [- 1.00, 0.31]; P = 0.2716). Similar results were shown for the adjusted mean between-group difference in the width and volume of bone erosion of the 2-3 metacarpal heads. Significant improvements in bone micro-architecture parameters were shown. The incidence of AEs and serious AEs was similar between the csDMARDs plus denosumab and the csDMARD therapy alone groups (AEs: 52.2% vs 56.5%; serious AEs: 4.3% vs 8.7%). CONCLUSIONS Although the addition of denosumab to csDMARDs did not find statistically significant improvements in bone erosion after 6 months of treatment, numerical improvements in these parameters suggest that the addition of denosumab to csDMARDs may be effective in inhibiting the progression of bone erosion and improving bone micro-architecture. TRIAL REGISTRATION University Hospital Medical Information Network Clinical Trials Registry, UMIN000030575. Japan Registry for Clinical Trials, jRCTs071180018.
Collapse
Affiliation(s)
- Naoki Iwamoto
- grid.174567.60000 0000 8902 2273Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Ko Chiba
- grid.174567.60000 0000 8902 2273Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Shuntaro Sato
- grid.411873.80000 0004 0616 1585Clinical Research Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Kazuteru Shiraishi
- grid.174567.60000 0000 8902 2273Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Kounosuke Watanabe
- grid.174567.60000 0000 8902 2273Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Nozomi Oki
- grid.174567.60000 0000 8902 2273Department of Radiological Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Akitomo Okada
- grid.415640.2Department of Rheumatology, National Hospital Organization Nagasaki Medical Center, 2-1001-1 Kubara, Omura, Nagasaki, 856-8562 Japan
| | - Tomohiro Koga
- grid.174567.60000 0000 8902 2273Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Shin-ya Kawashiri
- grid.174567.60000 0000 8902 2273Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan ,grid.174567.60000 0000 8902 2273Departments of Community Medicine, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Mami Tamai
- grid.174567.60000 0000 8902 2273Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Naoki Hosogaya
- grid.411873.80000 0004 0616 1585Clinical Research Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Masako Furuyama
- Department of Rheumatology, Nagasaki Kita Hospital, 800 Motomurago, Nishisonogigun Togitsucho, Nagasaki, 851-2103 Japan
| | - Makiko Kobayashi
- grid.410844.d0000 0004 4911 4738Primary Medical Science Department, Medical Affairs Division, Daiichi Sankyo Co., Ltd, 3-5-1 Nihonbashi-Honcho, Chuo-ku, Tokyo, 103-8426 Japan
| | - Kengo Saito
- grid.410844.d0000 0004 4911 4738Primary Medical Science Department, Medical Affairs Division, Daiichi Sankyo Co., Ltd, 3-5-1 Nihonbashi-Honcho, Chuo-ku, Tokyo, 103-8426 Japan
| | - Naoki Okubo
- grid.410844.d0000 0004 4911 4738Data Intelligence Department, Digital Transformation Management Division, Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710 Japan
| | - Masataka Uetani
- grid.174567.60000 0000 8902 2273Department of Radiological Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Makoto Osaki
- grid.174567.60000 0000 8902 2273Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Atsushi Kawakami
- grid.174567.60000 0000 8902 2273Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| |
Collapse
|
36
|
Wu H, Cheng K, Guo Q, Yang W, Tong L, Wang Y, Sun Z. Mapping Knowledge Structure and Themes Trends of Osteoporosis in Rheumatoid Arthritis: A Bibliometric Analysis. Front Med (Lausanne) 2021; 8:787228. [PMID: 34888333 PMCID: PMC8650090 DOI: 10.3389/fmed.2021.787228] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/18/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Rheumatoid arthritis is a chronic disabling disease characterized by chronic inflammation, articular cartilage destruction, and reduced bone mass. Multiple studies have revealed that the development of osteoporosis in rheumatoid arthritis (RA; ORA) patients could be led to a reduced quality of life and increased healthcare costs. Nevertheless, no attempt has been made to analyze the field of ORA research with the bibliometric method. This study aimed to provide a comprehensive overview of the knowledge structure and theme trends in the field of ORA research from a bibliometric perspective. Methods: Articles and reviews regarding ORA from 1998 to 2021 were identified from the Web of Science database. An online bibliometric platform, CiteSpace, and VOSviewer software were used to generate visualization knowledge maps including co-authorship, co-citation, and co-occurrence analysis. SPSS, R, and Microsoft Excel software were used to conduct curve fitting and correlation analysis, and to analyze quantitative indicators, such as publication and citation counts, h-index, and journal citation reports. Results: A total of 1,081 papers with 28,473 citations were identified. Publications were mainly concentrated in North America, Western Europe, and Eastern Asia. Economic strength is an important factor affecting scientific output. The United States contributed the most publications (213) with the highest h-index value (46) as of September 14, 2021. Diakonhjemmet Hospital and professor Haugeberg G were the most prolific institution and influential authors, respectively. Journal of Rheumatology was the most productive journal concerning ORA research. According to the burst references, “anti-citrullinated protein antibodies” and “preventing joint destruction” have been recognized as the hot research issues in the domain. The keywords co-occurrence analysis identified “teriparatide,” “interleukin-6,” “Wnt,” and “vertebral fractures” as the important future research directions. Conclusion: This was the first bibliometric study comprehensively summarizing the trends and development of ORA research. Our findings could offer practical sources for scholars to understand the key information in this field, and identify the potential research frontiers and hot directions in the near future.
Collapse
Affiliation(s)
- Haiyang Wu
- Department of Clinical Medicine, Graduate School of Tianjin Medical University, Tianjin, China
| | - Kunming Cheng
- Department of Intensive Care Unit, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiang Guo
- Department of Orthopaedic Surgery, Baodi Clinical College of Tianjin Medical University, Tianjin, China
| | - Weiguang Yang
- Department of Clinical Medicine, Graduate School of Tianjin Medical University, Tianjin, China
| | - Linjian Tong
- Department of Clinical Medicine, Graduate School of Tianjin Medical University, Tianjin, China
| | - Yulin Wang
- Department of Clinical Medicine, Graduate School of Tianjin Medical University, Tianjin, China
| | - Zhiming Sun
- Department of Clinical Medicine, Graduate School of Tianjin Medical University, Tianjin, China.,Department of Orthopaedic Surgery, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
37
|
Kim B, Cho YJ, Lim W. Osteoporosis therapies and their mechanisms of action (Review). Exp Ther Med 2021; 22:1379. [PMID: 34650627 PMCID: PMC8506919 DOI: 10.3892/etm.2021.10815] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Osteoporosis is a common disease that affects millions of patients worldwide and is most common in menopausal women. The main characteristics of osteoporosis are low bone density and increased risk of fractures due to deterioration of the bone architecture. Osteoporosis is a chronic disease that is difficult to treat; thus, investigations into novel effective therapeutic methods are required. A number of studies have focused on determining the most effective treatment options for this disease. There are several treatment options for osteoporosis that differ depending on the characteristics of the disease, and these include both well-established and newly developed drugs. The present review focuses on the various drugs available for osteoporosis, the associated mechanisms of action and the methods of administration.
Collapse
Affiliation(s)
- Beomchang Kim
- Laboratory of Orthopaedic Research, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Yong Jin Cho
- Department of Orthopaedic Surgery, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Wonbong Lim
- Laboratory of Orthopaedic Research, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
- Department of Orthopaedic Surgery, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
- Department of Premedical Sciences, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
38
|
Mori Y, Izumiyama T, Mori N, Aizawa T. Interstitial Lung Disease in a Woman with Rheumatoid Arthritis Treated with Denosumab: A Case Report. Mod Rheumatol Case Rep 2021; 6:155-159. [PMID: 34791403 DOI: 10.1093/mrcr/rxab046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/30/2021] [Accepted: 10/21/2021] [Indexed: 11/12/2022]
Abstract
The present report describes the case of an 84-year-old female Japanese patient with rheumatoid arthritis (RA) who experienced exacerbation of interstitial lung disease (ILD) after denosumab (Dmab) treatment. The onset of RA occurred in 2008, and the patient had been treated with intravenous or subcutaneous injection of tocilizumab (TCZ) since 2009. In July 2013, she experienced a lumbar vertebral fracture and began treatment with 60-mg Dmab injection every 6 months in January 2014. The patient had a history of mild ILD and was evaluated for ILD by chest computed tomography (CT) imaging prior to the start of Dmab use. The vertebral fracture did not recur after the initiation of Dmab treatment, and her osteoporosis was successfully treated. However, she expressed a concern of exacerbations of cough and respiratory discomfort that had occurred since September 2019. The chest CT image in November 2015 showed minor ILD progression, whereas the image in September 2019 showed severe exacerbation of ILD. To treat this exacerbation, 10 mg of methylprednisolone (mPSL) and 2.5 mg of tacrolimus (TAC) were administered, and Dmab was discontinued. The patient was subsequently switched to oral bisphosphonate. The patient's respiratory discomfort and the finding of interstitial lung lesion in CT imaging improved after Dmab discontinuation. This case showed that exacerbation of ILD may occur after Dmab treatment, and physicians should consider the risks of Dmab-related ILD in patients with RA complicated by ILD.
Collapse
Affiliation(s)
- Yu Mori
- Department of Orthopedic Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Takuya Izumiyama
- Department of Orthopedic Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Naoko Mori
- Department of Diagnostic Radiology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Toshimi Aizawa
- Department of Orthopedic Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
39
|
Mochizuki T, Yano K, Ikari K, Okazaki K. Effects of romosozumab or denosumab treatment on the bone mineral density and disease activity for 6 months in patients with rheumatoid arthritis with severe osteoporosis: An open-label, randomized, pilot study. Osteoporos Sarcopenia 2021; 7:110-114. [PMID: 34632114 PMCID: PMC8486617 DOI: 10.1016/j.afos.2021.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/10/2021] [Accepted: 08/15/2021] [Indexed: 12/24/2022] Open
Abstract
Objectives To investigate effects of romosozumab treatment on disease activity and bone mineral density (BMD) in patients with rheumatoid arthritis (RA) and severe osteoporosis in comparison with effects of denosumab treatment. Methods A total of 50 women were enrolled in this study. The subjects were randomized equally into 2 groups: the romosozumab group or the denosumab group. Disease activity score in 28 joints (DAS28)-erythrocyte sedimentation rate (ESR) and BMD at lumbar spine were evaluated. Results The percent changes (Δ) in the BMD values at 3 and 6 months for the lumbar spine were as follows: romosozumab; 4.9% and 5.2%, denosumab: 2.3% and 3.2%. The ΔBMD for the lumbar spine at 3 months was significantly higher in the romosozumab group than in the denosumab group (P = 0.044). The DAS28-ESR at baseline, 3 and 6 months in the romosozumab group were 2.88, 2.60 (P = 0.427) and 2.58 (P = 0.588), respectively. The change from baseline in DAS28-ESR did not differ significantly between these 2 groups at any time point. Conclusions The present study revealed that romosozumab treatment is more effective than denosumab treatment in increasing BMD of the lumbar spine at 3 months. Furthermore, the present study suggested that romosozumab treatment has no effects on the disease activity of RA in patients with RA and severe osteoporosis for 6 months.
Collapse
Affiliation(s)
- Takeshi Mochizuki
- Department of Orthopedic Surgery, Kamagaya General Hospital, Chiba, Japan
- Corresponding author. Department of Orthopedic Surgery, Kamagaya General Hospital, 929-6 Hatsutomi, Kamagaya, Chiba, 273-0121, Japan.
| | - Koichiro Yano
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Katsunori Ikari
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
- Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women's Medical University, Tokyo, Japan
| | - Ken Okazaki
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
40
|
Abdulkhaliq A, Cheikh M, Almuntashri F, Alzahrani H, Nadwi H, Kadi E, Abed M, Janaini M, Monjed A, Janoudi N, Almoallim H. A Comparison of Demographics, Disease Activity, Disability, and Treatment Among Rheumatoid Arthritis Patients with and without Osteoporosis. Open Access Rheumatol 2021; 13:275-283. [PMID: 34548823 PMCID: PMC8449680 DOI: 10.2147/oarrr.s318810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/27/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Osteoporosis (OP) is one of the most common comorbidities associated with rheumatoid arthritis (RA). Literatures reported that the risk for developing OP was strongly associated with duration and severity of RA. We aim to elaborate on the consequences of OP on disease activity and management plan in patients with RA. Patients and Methods A retrospective cohort study recruited 408 patients, including those with RA alone and with RA plus OP. The RA disease activity in the patients was assessed using disease activity score in 28 joints (DAS28-CRP). A statistical analysis was performed to compare data between the two groups of patients and determine any significant risk factor associated with the development of OP in RA patients. Results Of 408 patients who were included in this study, 353 patients (86.5%) had only RA, while 55 patients (13.5%) had RA with OP and showed significant difference (P = 0.04) concerning age categories. Patients diagnosed with RA and OP had RA duration longer than RA-only patients (independent t-test, P = 0.01). The two groups had almost similar disease activity at the three clinical visits, as well, had nearly similar disability at their first visit, whereas RA with OP patients had significant greater disability at their 2nd and 3rd visits (independent t-test, P = 0.001). Both groups were treated with the same biologic and non-biologic medication of similar frequency, although RA patients with OP received steroid more frequently than patients had RA only (61.7% vs. 41.7%, chi square test, P = 0.03). Conclusion There was no significant difference in disease activity at both groups of patients. However, RA with OP group had longer duration of RA, were more frequently treated with steroids, and had greater disability. We recommend physicians focus on controlling RA disease activity, early screening for and treating of OP.
Collapse
Affiliation(s)
- Altaf Abdulkhaliq
- Department of Clinical Biochemistry, Faculty of Medicine, Umm Al Qura University, Makkah, Saudi Arabia
| | - Mohamed Cheikh
- Internal Medicine Department, Doctor Soliman Fakeeh Hospital, Jeddah, Saudi Arabia.,Department of Medicine, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Fahad Almuntashri
- Department of Medicine, Faculty of Medicine, Umm Al Qura University, Makkah, Saudi Arabia
| | - Haneen Alzahrani
- Department of Medicine, Faculty of Medicine, Umm Al Qura University, Makkah, Saudi Arabia
| | - Huda Nadwi
- Department of Medicine, Faculty of Medicine, Umm Al Qura University, Makkah, Saudi Arabia
| | - Eithar Kadi
- Department of Medicine, Faculty of Medicine, Umm Al Qura University, Makkah, Saudi Arabia
| | - Mutasem Abed
- Department of Medicine, Faculty of Medicine, Umm Al Qura University, Makkah, Saudi Arabia
| | - Murad Janaini
- Department of Medicine, Faculty of Medicine, Umm Al Qura University, Makkah, Saudi Arabia
| | - Alaa Monjed
- Department of Medicine, Faculty of Medicine, Umm Al Qura University, Makkah, Saudi Arabia
| | - Nahed Janoudi
- Internal Medicine Department, Doctor Soliman Fakeeh Hospital, Jeddah, Saudi Arabia
| | - Hani Almoallim
- Department of Medicine, Faculty of Medicine, Umm Al Qura University, Makkah, Saudi Arabia.,Alzaidi Chair of Research in Rheumatic Diseases, Umm Al Qura University, Makkah, Saudi Arabia
| |
Collapse
|
41
|
Takayanagi H. Osteoimmunology as an intrinsic part of immunology. Int Immunol 2021; 33:673-678. [PMID: 34415037 DOI: 10.1093/intimm/dxab057] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/19/2021] [Indexed: 11/13/2022] Open
Abstract
Osteoimmunology has emerged as a field linking immunology and bone biology, but it has yet to be recognized as belonging to mainstream immunology. However, the extent of the research fields immunology actually covers has been enormously widened, and it is now ready to include such an interdisciplinary subject. One of the most obvious examples of an interaction between the immune and bone systems is the pathogenesis of rheumatoid arthritis, where bone resorption is increased by the autoimmune response. Moreover, the regulation of the immune system by bone cells has been clearly demonstrated by the finding that osteoprogenitor cells contribute to hematopoietic stem cell maintenance as well as the suppression of hematopoietic malignancy. Thus, the bidirectional dialogue has been established and inevitably will lead to the union of bone and immunity. Here I summarize the history and concept of osteoimmunology, providing a perspective on the future of immunology.
Collapse
Affiliation(s)
- Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
42
|
Tanaka S, Kobayashi M, Saito K, Takita A. Impact of denosumab discontinuation on changes in bone mineral density and bone erosion in rheumatoid arthritis patients. Mod Rheumatol 2021; 32:284-291. [PMID: 34850085 DOI: 10.1093/mr/roab022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/03/2021] [Accepted: 06/11/2021] [Indexed: 11/12/2022]
Abstract
OBJECTIVES This study investigated changes in bone mineral density (BMD) and erosion after denosumab discontinuation in rheumatoid arthritis (RA) patients without osteoporosis who participated in the DESIRABLE study. METHODS This multicentre observational study consisted of a prediscontinuation visit (date of final assessment in DESIRABLE) and a postdiscontinuation visit (2.5 years after the last administered dose of denosumab). Percentage change in lumbar spine (LS) BMD from baseline was assessed as the primary endpoint. RESULTS Fifty-nine patients were enrolled. The percentage change in LS BMD decreased to baseline levels at the postdiscontinuation visit. Compared with baseline, C-telopeptide of type I collagen levels increased after denosumab discontinuation but most patients had levels within the reference range. Bone erosion scores were not significantly different between the on-treatment period and after denosumab discontinuation (p = .0666) but there was a numerical increase postdiscontinuation. The progression in bone erosion score was significantly reduced in patients whose disease activity was in remission versus those not in remission (p = .0195). CONCLUSIONS In RA patients without osteoporosis, denosumab discontinuation can be explored while considering patient background factors (disease activity and risk of fracture) and accounting for progression of bone erosion and LS BMD decrease after withdrawal.
Collapse
Affiliation(s)
- Sakae Tanaka
- Department of Orthopedic Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makiko Kobayashi
- Primary Medical Science Department, Medical Affairs Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Kengo Saito
- Primary Medical Science Department, Medical Affairs Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Atsushi Takita
- Data Intelligence Department, Digital Transformation Management Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
43
|
Murata K, Uozumi R, Hashimoto M, Ebina K, Akashi K, Onishi A, Nagai K, Yoshikawa A, Katayama M, Son Y, Amuro H, Hara R, Yamamoto W, Watanabe R, Murakami K, Tanaka M, Ito H, Morinobu A, Matsuda S. The real-world effectiveness of anti-RANKL antibody denosumab on the clinical fracture prevention in patients with rheumatoid arthritis: The ANSWER cohort study. Mod Rheumatol 2021; 32:834-838. [PMID: 34910199 DOI: 10.1093/mr/roab043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 11/14/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by localized and generalized bone loss. The risk of fractures is doubled in patients with RA. Denosumab, an anti-RANKL monoclonal antibody, is used for those with osteoporosis at high risk fracture and it has inhibitory effect of progressive bone erosion in patients with RA. While the increase in bone mineral density by denosumab has been reported in patients with RA, preventive effect of fracture by denosumab remains unknown. This study aimed to evaluate the efficacy of denosumab in treating clinical fracture risk in patients with RA. METHODS Patients with RA who received denosumab treatment between 2013 and 2019 were retrospectively evaluated using the ANSWER (Kansai Consortium for the Well-Being of Rheumatic Disease Patients) cohort data. Fracture rates were evaluated between 0 and 6 months (reference period) versus > 6 months (post-reference period) of denosumab use. RESULTS A total of 873 patients with RA received denosumab, and their characteristics were as follows: 88% females, mean age 68 years, and average disease duration 14.5 years. The hazard rates of all clinical fractures were 0.69 (per 100 person-years) in the reference period and 0.35 in the post-reference period, indicating a 49.2% decrease (p = 0.03). CONCLUSIONS Denosumab suppresses the risk of clinical fractures in patients with RA.
Collapse
Affiliation(s)
- Koichi Murata
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryuji Uozumi
- Department of Biomedical and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Motomu Hashimoto
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kosuke Ebina
- Department of Musculoskeletal Regenerative Medicine, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Kengo Akashi
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Akira Onishi
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Koji Nagai
- Department of Internal Medicine (IV), Osaka Medical College, Osaka, Japan
| | - Ayaka Yoshikawa
- Department of Internal Medicine (IV), Osaka Medical College, Osaka, Japan
| | - Masaki Katayama
- Department of Rheumatology, Osaka Red Cross Hospital, Osaka, Japan
| | - Yonsu Son
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Hideki Amuro
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Ryota Hara
- The Center for Rheumatic Diseases, Department of Orthopaedic Surgery, Nara Medical University, Nara, Japan
| | - Wataru Yamamoto
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Health Information Management, Kurashiki Sweet Hospital, Okayama, Japan
| | - Ryu Watanabe
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kosaku Murakami
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masao Tanaka
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiromu Ito
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
44
|
Tanaka Y, Soen S, Ishiguro N, Yamanaka H, Yoneda T, Tanaka S, Ohira T, Nitta T, Okubo N, Genant H, van der Heijde D, Takeuchi T. Identifying the preferable rheumatoid arthritis subgroups for intervention with the anti-RANKL antibody denosumab to reduce progression of joint destruction. RMD Open 2021; 6:rmdopen-2020-001249. [PMID: 32732353 PMCID: PMC7722273 DOI: 10.1136/rmdopen-2020-001249] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/06/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES To clarify which rheumatoid arthritis (RA) patients benefit most from the anti-receptor activator of nuclear factor-κB ligand antibody denosumab to reduce the progression of joint destruction. METHODS We pooled patient data from the 12-month, double-blind, placebo-controlled DRIVE (phase II) and DESIRABLE (phase III) studies. In DRIVE, concomitant treatment was limited to methotrexate, salazosulfapyridine and bucillamine. In DESIRABLE, patients could receive any disease-modifying antirheumatic drug. RA patients were randomised to denosumab 60 mg every 6 months (Q6M), every 3 months (Q3M) or placebo. Efficacy was assessed by van der Heijde-modified total Sharp score (mTSS), bone erosion score (ES) and joint space narrowing score (JSNS). Change in mTSS was assessed in subgroups stratified by risk factors for radiographic damage if the interaction factor was significant. RESULTS The pooled analysis included 909 patients. Denosumab reduced worsening of mTSS (mean (SD)) at 12 months in the Q6M (0.88 (3.30), p=0.0024) and Q3M (0.66 (2.16), p=0.0002) groups versus placebo (1.50 (3.73)). This reduction in mTSS progression was due to the change in ES (Q6M, 0.44 (1.89), p=0.0006; Q3M, 0.20 (0.86), p<0.0001) versus placebo (0.98 (2.54)); no effect was observed on JSNS. Anti-cyclic citrullinated peptide (CCP) antibodies, glucocorticoid use and baseline ES showed a significant interaction. Denosumab was particularly effective in patients who were anti-CCP antibody positive (p<0.05). Changes in mTSS versus placebo were observed in all denosumab dose groups, regardless of glucocorticoid use and baseline ES. CONCLUSIONS Denosumab broadly reduced the progression of joint destruction in RA patients with risk factors for radiographic damage such as especially anti-CCP antibody positivity.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Satoshi Soen
- Department of Orthopaedic Surgery and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan
| | - Naoki Ishiguro
- Department of Orthopaedics, Nagoya University Graduate School of Medicine, Aichi, Japan
| | | | - Toshiyuki Yoneda
- Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, The University of Tokyo, Tokyo, Japan
| | - Takeshi Ohira
- Clinical Development Department, Daiichi Sankyo Co Ltd, Tokyo, Japan
| | - Takaya Nitta
- Clinical Development Department, Daiichi Sankyo Co Ltd, Tokyo, Japan
| | - Naoki Okubo
- Data Intelligence Department, Daiichi Sankyo Co Ltd, Tokyo, Japan
| | - Harry Genant
- Departments of Radiology, University of California San Francisco, California, USA
| | | | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
45
|
Park YJ, Gherghe AM, van der Heijde D. Radiographic progression in clinical trials in rheumatoid arthritis: a systemic literature review of trials performed by industry. RMD Open 2021; 6:rmdopen-2020-001277. [PMID: 32669455 PMCID: PMC7425197 DOI: 10.1136/rmdopen-2020-001277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/28/2020] [Indexed: 11/29/2022] Open
Abstract
Objectives To summarise radiographic data in randomised controlled trials (RCTs) as part of the radiographic inhibition claim of disease-modifying antirheumatic drugs (DMARDs) approved for patients with rheumatoid arthritis (RA). Methods A systemic literature review was performed using the Medline database from 1994 to February 2020. The results were grouped based on the scoring methods (Sharp, Genant modification, van der Heijde modification) and RA patient populations. Results One hundred sixty-eight publications were selected. After detailed assessment, 52 RCTs (7 methotrexate (MTX)-naive, 23 MTX inadequate response (IR), 9 DMARDs IR and 3 tumour necrosis factor-alpha inhibitors (TNFi) IR studies) were finally included. Information on patient population, scoring method used, reader reliability, statistical analyses and detailed radiographic data on baseline and change scores over multiple follow-up periods are presented. Conclusion The data gathered in this review serve as a repository for the design of future trials with radiographic damage as an outcome.
Collapse
Affiliation(s)
- Yune-Jung Park
- Internal Medicine, The Catholic University of Korea, Seoul, Korea (the Republic of).,Rheumatology, St. Vincent's Hospital, The Catholic University of Korea, Suwon, Korea (the Republic of)
| | - Ana Maria Gherghe
- Internal Medicine and Rheumatology, Cantacuzino Hospital, Bucharest, Romania
| | | |
Collapse
|
46
|
Bertoldo E, Adami G, Rossini M, Giollo A, Orsolini G, Viapiana O, Gatti D, Fassio A. The Emerging Roles of Endocrine Hormones in Different Arthritic Disorders. Front Endocrinol (Lausanne) 2021; 12:620920. [PMID: 34093428 PMCID: PMC8177688 DOI: 10.3389/fendo.2021.620920] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 04/20/2021] [Indexed: 12/17/2022] Open
Abstract
The relationship between endocrine hormones and the spectrum of rheumatic conditions has long been discussed in the literature, focusing primarily on sexual hormones, such as estrogens, androgens, prolactin (PRL). Estrogens are indeed involved in the pathogenesis of the main inflammatory arthritis thanks to their effects on the immune system, both stimulatory and inhibitory. The PRL system has been discovered in synovial tissue of rheumatoid arthritis (RA) and psoriatic arthritis (PsA), patients and has been propose as a new potential therapeutic target. Besides sexual hormones, in the last years scientific interest about the crosstalk of immune system with other class of hormones has grown. Hormones acting on the bone tissue (i.e. parathyroid hormone, vitamin D) and modulators of the Wnt pathway (i.e. Dickkopf-1) have been demonstrated to play active role in inflammatory arthritis course, defining a new field of research named osteoimmunology. PTH, which is one of the main determinants of Dkkopf-1, plays a crucial role in bone erosions in RA and a correlation between PTH, Trabecular Bone Score (TBS) and disease activity has been found in ankylosing spondylitis (AS). In PSA is under studying the interaction among IL-17 and bone metabolism. The purpose of this review is to discuss and summarize the recent data about the interaction between endocrine hormone and immune system in the main rheumatic disorders, covering in particular the role of bone-related hormones and cytokines. We will describe this relationship from a biochemical, diagnostic and therapeutic perspective, with a particular focus on RA, PsA and AS.
Collapse
Affiliation(s)
- Eugenia Bertoldo
- Rheumatology Unit, Department of Medicine, University of Verona, Verona, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Tanaka Y, Takeuchi T, Soen S, Yamanaka H, Yoneda T, Tanaka S, Nitta T, Okubo N, Genant HK, van der Heijde D. Effects of Denosumab in Japanese Patients With Rheumatoid Arthritis Treated With Conventional Antirheumatic Drugs: 36-month Extension of a Phase III Study. J Rheumatol 2021; 48:1663-1671. [PMID: 33858976 DOI: 10.3899/jrheum.201376] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To evaluate the safety and efficacy of long-term denosumab 60 mg every 6 months (Q6M) or every 3 months (Q3M) in patients with rheumatoid arthritis (RA). METHODS This 12-month, randomized, double-blind, placebo-controlled, multicenter, phase III trial with an open-label extension period from 12 to 36 months (DESIRABLE) enrolled Japanese patients with RA treated with placebo (P) for 12 months followed by either denosumab Q6M (P/Q6M) or denosumab Q3M (P/Q3M) for 24 months; denosumab Q6M for 36 months (Q6M/Q6M); or denosumab Q3M for 36 months (Q3M/Q3M). Efficacy was assessed by van der Heijde modified total Sharp score (mTSS), bone erosion score (BES), and joint space narrowing ( JSN) score. RESULTS Long-term treatment better maintained mTSS and BES suppression in the P/Q3M and Q3M/ Q3M vs P/Q6M and Q6M/Q6M groups; changes from baseline in total mTSS (standard error) at 36 months were 2.8 (0.4) and 1.7 (0.3) vs 3.0 (0.4) and 2.4 (0.3), respectively, and corresponding changes in BES were 1.3 (0.2) and 0.4 (0.2) vs 1.4 (0.2) and 1.1 (0.2), respectively. No JSN effect was observed. Bone mineral density consistently increased in all groups after denosumab initiation, regardless of concomitant glucocorticoid administration. Serum C-terminal telopeptide of type I collagen decreased rapidly at 1 month postdenosumab administration (in both the initial 12-month [Q3M and Q6M groups] and long-term treatment [P/Q3M and P/Q6M groups] phases). Adverse event incidence leading to study drug discontinuation was similar across treatment groups. CONCLUSION Denosumab treatment maintained inhibition of progression of joint destruction up to 36 months. Based on effects on BES progression, higher dosing frequency at an earlier treatment stage may be needed to optimize treatment. Denosumab was generally well tolerated. (ClinicalTrials.gov: NCT01973569).
Collapse
Affiliation(s)
- Yoshiya Tanaka
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Tsutomu Takeuchi
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Satoshi Soen
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Hisashi Yamanaka
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Toshiyuki Yoneda
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Sakae Tanaka
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Takaya Nitta
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Naoki Okubo
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Harry K Genant
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Désirée van der Heijde
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| |
Collapse
|
48
|
So H, Cheng IT, Lau SL, Chow E, Lam T, Hung VW, Li EK, Griffith JF, Lee VW, Shi L, Huang J, Kwok KY, Yim CW, Li TK, Lo V, Lee J, Lee JJW, Qin L, Tam LS. Effects of RANKL inhibition on promoting healing of bone erosion in rheumatoid arthritis using HR-pQCT: a 2-year, randomised, double-blind, placebo-controlled trial. Ann Rheum Dis 2021; 80:981-988. [PMID: 33811034 DOI: 10.1136/annrheumdis-2021-219846] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/21/2021] [Accepted: 03/18/2021] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To evaluate the effects of denosumab on erosion healing at 2-4 metacarpophalangeal (MCP) head as determined by high-resolution peripheral quantitative CT (HR-pQCT) in patients with rheumatoid arthritis (RA) with stable disease. METHODS This was a randomised, placebo-controlled, double-blind study. Patients with RA with disease activity score 28 joints (DAS28) ≤5.1 were randomised (1:1) to subcutaneous denosumab 60 mg or placebo once every 6 months for 24 months. The primary outcome was erosion healing at MCP 2-4 on HR-pQCT at 12 months. The effects of denosumab on erosion and joint space parameters on HR-pQCT and radiographs, disease activity and health assessment questionnaire-disability index (HAQ-DI) were also examined. RESULTS At 24 months, HR-pQCT images were analysed in 98 patients. One-third of the patients achieved sustained low disease activity throughout the study. At 12 months, changes in erosion parameters on HR-pQCT were similar between the two groups. At 24 months, new erosions (19% vs 9%, p=0.009) and erosion progression (18% vs 8%, p=0.019) were more common in the placebo group than the denosumab group. Erosion healing was seen in a significantly higher proportion of patients in the denosumab group (20% vs 6%, p=0.045) at 24 months. No significant changes in joint space parameters on HR-pQCT, van der Heijde-Sharp erosion score, DAS28 and HAQ-DI were observed in the two groups at 12 and 24 months. CONCLUSION Although no differences in erosion parameters were observed at 12 months, denosumab was more efficacious than placebo in erosion repair on HR-pQCT after 24 months. TRIAL REGISTRATION NUMBER NCT03239080.
Collapse
Affiliation(s)
- Ho So
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Isaac T Cheng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Sze-Lok Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Evelyn Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tommy Lam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Vivian W Hung
- Bone Quality and Health Centre of the Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Edmund K Li
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - James F Griffith
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Vivian W Lee
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Lin Shi
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Junbin Huang
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Kitty Yan Kwok
- Department of Medicine, Queen Elizabeth Hospital, Hong Kong, Hong Kong
| | - Cheuk-Wan Yim
- Department of Medicine, Tseung Kwan O Hospital, Hong Kong, Hong Kong
| | - Tena K Li
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Vincent Lo
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Jolie Lee
- Department of Medicine, Tai Po Hospital, Hong Kong, Hong Kong
| | - Jack Jock-Wai Lee
- Division of Biostatistics, The Jockey Club School of Public Health & Primary Care, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ling Qin
- Bone Quality and Health Centre of the Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
49
|
Managing Osteoporosis and Joint Damage in Patients with Rheumatoid Arthritis: An Overview. J Clin Med 2021; 10:jcm10061241. [PMID: 33802804 PMCID: PMC8002542 DOI: 10.3390/jcm10061241] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/06/2021] [Accepted: 03/12/2021] [Indexed: 01/04/2023] Open
Abstract
In rheumatoid arthritis, a representative systemic autoimmune disease, immune abnormality and accompanying persistent synovitis cause bone and cartilage destruction and systemic osteoporosis. Biologics targeting tumor necrosis factor, which plays a central role in the inflammatory process, and Janus kinase inhibitors have been introduced in the treatment of rheumatoid arthritis, making clinical remission a realistic treatment goal. These drugs can prevent structural damage to bone and cartilage. In addition, osteoporosis, caused by factors such as menopause, aging, immobility, and glucocorticoid use, can be treated with bisphosphonates and the anti-receptor activator of the nuclear factor-κB ligand antibody. An imbalance in the immune system in rheumatoid arthritis induces an imbalance in bone metabolism. However, osteoporosis and bone and cartilage destruction occur through totally different mechanisms. Understanding the mechanisms underlying osteoporosis and joint destruction in rheumatoid arthritis leads to improved care and the development of new treatments.
Collapse
|
50
|
Mahatma MM, Jayasuriya RL, Hughes D, Hoggard N, Buckley SC, Gordon A, Hamer AJ, Tomouk MW, Kerry RM, Eastell R, Wilkinson JM. Effect of denosumab on osteolytic lesion activity after total hip arthroplasty: a single-centre, randomised, double-blind, placebo-controlled, proof of concept trial. THE LANCET. RHEUMATOLOGY 2021; 3:e195-e203. [PMID: 38279382 DOI: 10.1016/s2665-9913(20)30394-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/27/2020] [Accepted: 11/03/2020] [Indexed: 01/28/2024]
Abstract
BACKGROUND Osteolysis causes recurrent pain and disability after total hip arthroplasty. We investigated the effect of the human monoclonal antibody denosumab on osteolytic lesion activity in patients undergoing revision total hip arthroplasty surgery to show the biological proof of concept for a non-surgical treatment for the disease. METHODS We did a phase 2, randomised, double-blind, placebo-controlled, proof of concept superiority trial at Sheffield Teaching Hospitals, Sheffield, UK. Eligible patients aged 30 years or older and scheduled for revision surgery for symptomatic, radiographically confirmed osteolysis were randomly allocated (1:1) to subcutaneous denosumab (60 mg single-dose) or placebo by an independent pharmacist using a random number table. The primary outcome was the between-group difference in osteoclast number per mm of bone surface of biopsies taken from the osteolytic membrane-bone interface at surgery 8 weeks later, measured by quantitative histomorphometry in all patients who underwent revision surgery. Adverse events were analysed in all randomly assigned participants. This trial is registered with the EU Clinical Trials Register (EudraCT 2011-000541-20). FINDINGS Between Dec 12, 2012, and June 24, 2018, 51 patients were assessed for eligibility, of whom 24 were randomly assigned to study treatment. Two patients had their revision surgery cancelled for unrelated reasons, leaving 22 patients (ten in the denosumab group) for analysis of the primary outcome. There were 83% fewer osteoclasts at the osteolysis membrane-bone interface in the denosumab versus the placebo group (median 0·05 per mm [IQR 0·11] vs 0·30 mm [0·40], p=0·011). No deaths or treatment-related serious adverse events occurred. Seven adverse events, including one severe adverse event, occurred in four (36%) of 11 patients in the denosumab group. In the placebo group ten adverse events, including three severe adverse events, occurred in five (38%) of 13 patients. INTERPRETATION To our knowledge, this is the first clinical trial of an investigational drug for osteolysis that shows tissue-specific biological efficacy. These results justify the need for future trials that target earlier-stage disease to test for clinical efficacy in reducing the need for revision surgery. FUNDING Amgen.
Collapse
Affiliation(s)
- Mohit M Mahatma
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - Raveen L Jayasuriya
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK
| | - David Hughes
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Nigel Hoggard
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Simon C Buckley
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Andrew Gordon
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Andrew J Hamer
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | | - Robert M Kerry
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Richard Eastell
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK
| | - Jeremy M Wilkinson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK; Mellanby Centre for Musculoskeletal Research, University of Sheffield, Sheffield, UK.
| |
Collapse
|