1
|
Iwamoto N, Chiba K, Sato S, Tashiro S, Shiraishi K, Watanabe K, Ohki N, Okada A, Koga T, Kawashiri SY, Tamai M, Osaki M, Kawakami A. Preferable effect of CTLA4-Ig on both bone erosion and bone microarchitecture in rheumatoid arthritis revealed by HR-pQCT. Sci Rep 2024; 14:27673. [PMID: 39532911 PMCID: PMC11557861 DOI: 10.1038/s41598-024-77392-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
This exploratory study aimed to examine the impact of abatacept treatment on bone structure in patients with rheumatoid arthritis (RA) using high-resolution peripheral quantitative computed tomography (HR-pQCT). RA patients initiating either abatacept or newly introduced csDMARDs were enrolled in this prospective, non-randomized, two-group study. Bone structure in the 2nd and 3rd metacarpal heads was assessed using HR-pQCT at 0, 6, and 12 months after enrollment. Synovitis was evaluated using musculoskeletal ultrasound and MRI. The adjusted mean between-group differences (abatacept-csDMARDs group) were estimated using a mixed-effect model. Thirty-five patients (abatacept group: n = 15; csDMARDs group: n = 20) were analyzed. Changes in erosion volume, depth and width were numerically smaller in the abatacept group compared to the csDMARDs group (adjusted mean between-group differences: - 1.86 mm3, - 0.02 mm, and - 0.09 mm, respectively). Over a 12-month period, 5 erosions emerged in the csDMARDs group, while only 1 erosion appeared in the abatacept group. Compared to csDMARDs, abatacept better preserved bone microarchitecture; several components of bone microarchitecture were significantly worsened at 6 months in the csDMARDs group, but were not deteriorated at 6 months in the abatacept group. Changes in synovitis scores were similar between the two treatment groups. Our results indicate that abatacept prevented the progression of bone erosion including new occurrence, and also prevented worsening of bone strength independently with synovitis compared to csDMARDs including MTX. Thus, abatacept treatment may provide benefits not only in inhibiting the progress of bone erosion but also in preventing bone microarchitectural deterioration.
Collapse
Affiliation(s)
- Naoki Iwamoto
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Ko Chiba
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shuntaro Sato
- Clinical Research Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shigeki Tashiro
- Clinical Research Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kazuteru Shiraishi
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kounosuke Watanabe
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Nozomi Ohki
- Department of Radiological Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Akitomo Okada
- Department of Rheumatology, National Hospital Organization Nagasaki Medical Center, Kubara 2-1001-1, Omura, Nagasaki, 856-8562, Japan
| | - Tomohiro Koga
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shin-Ya Kawashiri
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Center for Collaborative Medical Education and Development, Nagasaki University Institute of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Mami Tamai
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Makoto Osaki
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Atsushi Kawakami
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
2
|
Mou K, Chan SMH, Vlahos R. Musculoskeletal crosstalk in chronic obstructive pulmonary disease and comorbidities: Emerging roles and therapeutic potentials. Pharmacol Ther 2024; 257:108635. [PMID: 38508342 DOI: 10.1016/j.pharmthera.2024.108635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/13/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a multifaceted respiratory disorder characterized by progressive airflow limitation and systemic implications. It has become increasingly apparent that COPD exerts its influence far beyond the respiratory system, extending its impact to various organ systems. Among these, the musculoskeletal system emerges as a central player in both the pathogenesis and management of COPD and its associated comorbidities. Muscle dysfunction and osteoporosis are prevalent musculoskeletal disorders in COPD patients, leading to a substantial decline in exercise capacity and overall health. These manifestations are influenced by systemic inflammation, oxidative stress, and hormonal imbalances, all hallmarks of COPD. Recent research has uncovered an intricate interplay between COPD and musculoskeletal comorbidities, suggesting that muscle and bone tissues may cross-communicate through the release of signalling molecules, known as "myokines" and "osteokines". We explored this dynamic relationship, with a particular focus on the role of the immune system in mediating the cross-communication between muscle and bone in COPD. Moreover, we delved into existing and emerging therapeutic strategies for managing musculoskeletal disorders in COPD. It underscores the development of personalized treatment approaches that target both the respiratory and musculoskeletal aspects of COPD, offering the promise of improved well-being and quality of life for individuals grappling with this complex condition. This comprehensive review underscores the significance of recognizing the profound impact of COPD on the musculoskeletal system and its comorbidities. By unravelling the intricate connections between these systems and exploring innovative treatment avenues, we can aspire to enhance the overall care and outcomes for COPD patients, ultimately offering hope for improved health and well-being.
Collapse
Affiliation(s)
- Kevin Mou
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Stanley M H Chan
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
3
|
Su YJ, Lin CY, Hsu CY. Analyzing the risk of osteoporosis and fracture in rheumatoid arthritis patients who have been treated with various biologics. Int J Rheum Dis 2024; 27:e15055. [PMID: 38334206 DOI: 10.1111/1756-185x.15055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/31/2023] [Accepted: 01/19/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a major risk factor for osteoporosis/osteoporotic fractures. We aimed to elucidate the role of treatment choices among osteoporosis/osteoporotic fractures. METHODOLOGY We utilized the Chang-Gung Research Database to assess the risks of osteoporosis/osteoporotic fractures among independently treated RA patients, using retrospective time-to-event outcomes analysis. RESULTS A total of 3509 RA patients with a mean of 63.1 ± 8.6 years were analyzed. Among all, 1300 RA patients (37%) were diagnosed with newly diagnosed osteoporosis. The crude incidence of newly diagnosed osteoporosis was the highest among those treated with other conventional disease-modifying anti-rheumatic drugs (cDMARDs; 74.1 events/1000-PYs, 95%CI 66.0-82.3), followed by those with a non-treatment period (68 events/1000-PYs, 95%CI 63.1-72.9), methotrxate (MTX) monotherapy (60.7 events/1000-PYs, 95%CI 41.2-80.3), MTX plus other cDMARDs (51.9 events/1000-PYs, 95%CI 43.4-60.3), and abatacept/rituximab (48.6 events/1000-PYs, 95%CI 14.9-82.3). The lowest crude incidence was found in patients treated with anti-TNFi biologics (40.4 events/1000-PYs, 95%CI 28.6-52.2) and other biologic disease-modifying anti-rheumatic drugs (bDMARDs; 40.1 events/1000-PYs, 95%CI 8.0-72.1). A total of 270 patients (20.8%) suffered from an incident fracture during follow-ups. The crude incidence of fracture was the highest among those treated with abatacept/rituximab (49.0 events/1000-PYs, 95%CI 6.0-91.9), followed by those with non-treatment periods (24.3 events/1000-PYs, 95%CI 19.3-29.4), other cDMARDs (24.2 events/1000-PYs, 95%CI 18.1-30.2), anti-TNFi biologics (20.2 events/1000-PYs, 95%CI 8.8-31.6). Other bDMARDs (13.3 events/1000-PYs, 95%CI 0-39.2), MTX mono (12.5 events/1000-PYs, 95%CI 0.3-24.8), and MTX plus other cDMARDs (11.4 events/1000-PYs, 95%CI 5.4-17.4) were low incidences. CONCLUSION The treatment option has emerged as a critical determinant in the context of future osteoporosis and osteoporotic fracture risks among RA. These findings offer a valuable resource for clinicians, empowering them to tailor bespoke treatment strategies for RA patients, thereby mitigating the potential for future osteoporosis and fractures.
Collapse
Affiliation(s)
- Yu-Jih Su
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chun-Yu Lin
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chung-Yuan Hsu
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
4
|
Cai L, Lv Y, Yan Q, Guo W. Cytokines: The links between bone and the immune system. Injury 2024; 55:111203. [PMID: 38043143 DOI: 10.1016/j.injury.2023.111203] [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: 03/09/2023] [Revised: 11/05/2023] [Accepted: 11/12/2023] [Indexed: 12/05/2023]
Abstract
Osteoporosis results from an imbalance in a highly balanced physiological process called bone remodeling, in which osteoclast-mediated bone resorption and osteoblast-mediated bone formation play important roles. Osteoimmunology is a newly discovered interdisciplinary research field that focuses on the relationship between bone and the immune system. Specifically, bone and the immune system interact through cytokines, immune cells secrete cytokines, and cytokines finely regulate bone metabolism by mediating the differentiation and activity of osteoclasts and osteoblasts. Therefore, understanding the influence of cytokines on bone metabolism is conducive for the development of novel targeted drugs against immune-related bone diseases. This review summarizes the pathophysiological functions of various common cytokines in bone and discusses the potential clinical value of multiple cytokines in immune-mediated bone diseases.
Collapse
Affiliation(s)
- Liping Cai
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China; Department of Endocrinology, Rheumatology and Immunology, Anyang People's Hospital, Anyang, Henan 455000, China
| | - You Lv
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Qihui Yan
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Weiying Guo
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
5
|
Simon D, Minopoulou I, Kemenes S, Bayat S, Tascilar K, Mutlu MY, Valor-Méndez L, Krönke G, Hueber AJ, Schett G, Kleyer A. Baricitinib Improves Bone Properties and Biomechanics in Patients With Rheumatoid Arthritis: Results of the Prospective Interventional BARE BONE Trial. Arthritis Rheumatol 2023; 75:1923-1934. [PMID: 37229650 DOI: 10.1002/art.42617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is characterized by erosive joint damage, deterioration of bone mass, and biomechanics. Preclinical evidence suggests a beneficial effect of Janus kinase inhibition (JAKi) on bone properties, but clinical data are scarce to date. In this study, we evaluated the effect of JAKi through baricitinib (BARI) on 1) volumetric bone mineral density (vBMD), bone microstructure, biomechanics, and erosion repair and 2) synovial inflammation in RA patients. METHODS Prospective, single-arm, interventional, open-label, single-center phase 4 study in RA patients with pathological bone status and clinical indication of JAKi (BARE BONE trial). Participants received BARI (4 mg/day) over 52 weeks. To assess bone properties and synovial inflammation, high-resolution computed tomography scans and magnetic resonance imaging were performed at baseline (BL), week 24, and week 52. Clinical response and safety were monitored. RESULTS Thirty RA patients were included. BARI significantly improved disease activity (Disease Activity Score in 28 joints using the erythrocyte sedimentation rate: 4.82 ± 0.90 to 2.71 ± 0.83) and synovial inflammation (RAMRIS synovitis score: 5.3 [4.2] to 2.7 [3.5]). We observed a significant improvement in trabecular vBMD with a mean change of 6.11 mgHA/mm3 (95% confidence interval [95% CI] 0.01-12.26). Biomechanical properties also improved with mean change from baseline in estimated stiffness of 2.28 kN/mm (95% CI 0.30-4.25) and estimated failure load of 98.8 N (95% CI 15.9-181.7). The number and size of erosions in the metacarpal joints remained stable. No new safety signals with BARI treatment were observed. CONCLUSION Bones of RA patients improve with BARI therapy, as shown by an increase in trabecular bone mass and an improvement of biomechanical properties.
Collapse
Affiliation(s)
- David Simon
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ioanna Minopoulou
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stephan Kemenes
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sara Bayat
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Koray Tascilar
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Melek Yalcin Mutlu
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Larissa Valor-Méndez
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Axel J Hueber
- Division of Rheumatology, Klinikum Nürnberg, Paracelsus Medical University, Nürnberg, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, and Deutsches Zentrum Immuntherapie, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
6
|
Minopoulou I, Kleyer A, Yalcin-Mutlu M, Fagni F, Kemenes S, Schmidkonz C, Atzinger A, Pachowsky M, Engel K, Folle L, Roemer F, Waldner M, D'Agostino MA, Schett G, Simon D. Imaging in inflammatory arthritis: progress towards precision medicine. Nat Rev Rheumatol 2023; 19:650-665. [PMID: 37684361 DOI: 10.1038/s41584-023-01016-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 09/10/2023]
Abstract
Imaging techniques such as ultrasonography and MRI have gained ground in the diagnosis and management of inflammatory arthritis, as these imaging modalities allow a sensitive assessment of musculoskeletal inflammation and damage. However, these techniques cannot discriminate between disease subsets and are currently unable to deliver an accurate prediction of disease progression and therapeutic response in individual patients. This major shortcoming of today's technology hinders a targeted and personalized patient management approach. Technological advances in the areas of high-resolution imaging (for example, high-resolution peripheral quantitative computed tomography and ultra-high field MRI), functional and molecular-based imaging (such as chemical exchange saturation transfer MRI, positron emission tomography, fluorescence optical imaging, optoacoustic imaging and contrast-enhanced ultrasonography) and artificial intelligence-based data analysis could help to tackle these challenges. These new imaging approaches offer detailed anatomical delineation and an in vivo and non-invasive evaluation of the immunometabolic status of inflammatory reactions, thereby facilitating an in-depth characterization of inflammation. By means of these developments, the aim of earlier diagnosis, enhanced monitoring and, ultimately, a personalized treatment strategy looms closer.
Collapse
Affiliation(s)
- Ioanna Minopoulou
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Arnd Kleyer
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Melek Yalcin-Mutlu
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Filippo Fagni
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Kemenes
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christian Schmidkonz
- Department of Nuclear Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Institute for Medical Engineering, University of Applied Sciences Amberg-Weiden, Weiden, Germany
| | - Armin Atzinger
- Department of Nuclear Medicine, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Milena Pachowsky
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Lukas Folle
- Pattern Recognition Lab, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Frank Roemer
- Institute of Radiology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Radiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Maximilian Waldner
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Internal Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Maria-Antonietta D'Agostino
- Division of Rheumatology, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Université Paris-Saclay, UVSQ, Inserm U1173, Infection et Inflammation, Laboratory of Excellence Inflamex, Montigny-Le-Bretonneux, France
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - David Simon
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
7
|
Miyata M, Hirabayashi Y, Munakata Y, Urata Y, Saito K, Okuno H, Yoshida M, Kodera T, Watanabe R, Miyamoto S, Ishii T, Nakazawa S, Takemori H, Ando T, Kanno T, Komagamine M, Kato I, Takahashi Y, Komatsuda A, Endo K, Murai C, Takakubo Y, Miura T, Sato Y, Ichikawa K, Konta T, Chiba N, Muryoi T, Kobayashi H, Fujii H, Sekiguchi Y, Hatakeyama A, Ogura K, Sakuraba H, Asano T, Kanazawa H, Suzuki E, Takasaki S, Asakura K, Suzuki Y, Takagi M, Nakayama T, Watanabe H, Miura K, Mori Y. Feasibility of methotrexate discontinuation following tocilizumab and methotrexate combination therapy in patients with long-standing and advanced rheumatoid arthritis: a 3-year observational cohort study. Fukushima J Med Sci 2023; 69:11-20. [PMID: 36990790 PMCID: PMC10122970 DOI: 10.5387/fms.2022-06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 01/30/2023] [Indexed: 03/31/2023] Open
Abstract
OBJECTIVES Methotrexate (MTX) is associated with extensive side effects, including myelosuppression, interstitial pneumonia, and infection. It is, therefore, critical to establish whether its administration is required after achieving remission with tocilizumab (TCZ) and MTX combination therapy in patients with rheumatoid arthritis (RA). Therefore, the aim of this multicenter, observational, cohort study was to evaluate the feasibility of MTX discontinuation for the safety of these patients. METHODS Patients with RA were administered TCZ, with or without MTX, for 3 years; those who received TCZ+MTX combination therapy were selected. After remission was achieved, MTX was discontinued without flare development in one group (discontinued [DISC] group, n = 33) and continued without flare development in another group (maintain [MAIN] group, n = 37). The clinical efficacy of TCZ+MTX therapy, patient background characteristics, and adverse events were compared between groups. RESULTS The disease activity score in 28 joints-erythrocyte sedimentation rate (DAS28-ESR) at 3, 6, and 9 months was significantly lower in the DISC group (P < .05, P < .01, and P < .01, respectively). Further, the DAS28-ESR remission rate at 6 and 9 months and Boolean remission rate at 6 months were significantly higher in the DISC group (P < .01 for all). Disease duration was significantly longer in the DISC group (P < .05). Furthermore, the number of patients with stage 4 RA was significantly higher in the DISC group (P < .01). CONCLUSIONS Once remission was achieved, MTX was discontinued in patients who responded favorably to TCZ+MTX therapy, despite the prolonged disease duration and stage progression.
Collapse
Affiliation(s)
- Masayuki Miyata
- Department of Internal Medicine, Fukushima Red Cross Hospital
| | | | | | | | | | - Hiroshi Okuno
- Department Orthopedic Surgery, Tohoku Rosai Hospital
| | | | - Takao Kodera
- Center for Arthritis and Rheumatic Diseases, Tohoku Pharmaceutical University Hospital
| | - Ryu Watanabe
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University
| | - Seiya Miyamoto
- Department of Orthopedic Surgery, Tohoku Orthopedic Clinic
| | - Tomonori Ishii
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine
| | | | | | - Takanobu Ando
- Department of Orthopedics, General Hanamaki Hospital
| | - Takashi Kanno
- Department of Rheumatology, Ohta Nishinouchi Hospital
| | | | - Ichiro Kato
- Department Orthopedic Surgery, Tohoku Rosai Hospital
| | | | | | - Kojiro Endo
- Department of Rheumatology and Department of Orthopedic Surgery, Hoshi General Hospital
| | | | - Yuya Takakubo
- Department of Orthopedic Surgery, Yamagata University Faculty of Medicine
| | - Takao Miura
- Department of Orthopedic Surgery, Hirosaki Memorial Hospital
| | - Yukio Sato
- Department of Rheumatology, Kaiyama Central Hospital
| | - Kazunobu Ichikawa
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine
| | - Tsuneo Konta
- Department of Public Health and Hygiene/Nephrology and Collagen Disease
| | - Noriyuki Chiba
- Department of Internal Medicine, Morioka National Hospital
| | | | | | - Hiroshi Fujii
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine
| | | | | | | | - Hirotake Sakuraba
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine
| | - Tomoyuki Asano
- Department of Rheumatology and Collagen Disease, Fukushima Medical University Hospital
| | | | - Eiji Suzuki
- Department of Rheumatology, Ohta Nishinouchi Hospital
| | | | - Kenichi Asakura
- Department of Internal Medicine, Yuri Kumiai General Hospital
| | | | - Michiaki Takagi
- Department of Orthopedic Surgery, Yamagata University Faculty of Medicine
| | | | - Hiroshi Watanabe
- Department of Rheumatology and Collagen Disease, Fukushima Medical University Hospital
| | - Keiki Miura
- Department of Orthopedic Surgery, Wakuya-Town National Health Insurance Hospital
| | - Yu Mori
- Department of Orthopedic Surgery, Tohoku University Graduate School of Medicine
| | - the Michinoku Tocilizumab Study Group
- Department of Internal Medicine, Fukushima Red Cross Hospital
- Department of Rheumatology, Hikarigaoka Spellman Hospital
- Munakata Yasuhiko Clinic
- Department of Rheumatology, Tsugaru General Hospital
- Suminoya Rheumatism & Orthopedics Clinic
- Department Orthopedic Surgery, Tohoku Rosai Hospital
- Yoshida Orthopedic Surgery and Rheumatology Clinic
- Center for Arthritis and Rheumatic Diseases, Tohoku Pharmaceutical University Hospital
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University
- Department of Orthopedic Surgery, Tohoku Orthopedic Clinic
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine
- Nakazawa Sports Clinic
- Department of Rheumatology, Aomori Prefectural Central Hospital
- Department of Orthopedics, General Hanamaki Hospital
- Department of Rheumatology, Ohta Nishinouchi Hospital
- Komagamine Orthopedic and Rheumatology Clinic
- Yu Family Clinic
- Department of Internal Medicine, Ogachi Central Hospital
- Department of Rheumatology and Department of Orthopedic Surgery, Hoshi General Hospital
- Murai Clinic
- Department of Orthopedic Surgery, Yamagata University Faculty of Medicine
- Department of Orthopedic Surgery, Hirosaki Memorial Hospital
- Department of Rheumatology, Kaiyama Central Hospital
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine
- Department of Public Health and Hygiene/Nephrology and Collagen Disease
- Department of Internal Medicine, Morioka National Hospital
- Muryoi Clinic
- Minami-Fukushima Clinic
- NTT East Tohoku Hospital
- Ogura Orthopedic Surgery Clinic
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine
- Department of Rheumatology and Collagen Disease, Fukushima Medical University Hospital
- Department of Internal Medicine, Saiseikai Yamagata Hospital
- Department of Internal Medicine, Yuri Kumiai General Hospital
- Izumi Himawari Clinic
- Department of Internal Medicine, Nihonkai General Hospital
- Department of Orthopedic Surgery, Wakuya-Town National Health Insurance Hospital
- Department of Orthopedic Surgery, Tohoku University Graduate School of Medicine
| |
Collapse
|
8
|
Azadeh H. Association between disease-modifying antirheumatic drugs and bone turnover biomarkers. Int J Rheum Dis 2023; 26:437-445. [PMID: 36573666 DOI: 10.1111/1756-185x.14550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/24/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022]
Abstract
Rheumatoid arthritis (RA) has been linked to an increased risk of osteoporosis as well as fractures. Patients diagnosed with RA had a 25% increased risk of osteoporotic fracture, according to a recent population-based cohort study that compared them to people without RA. Several studies have found a correlation between osteoporosis and the presence of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1, and 6. These cytokines play a crucial part in the process of bone resorption by boosting osteoclast activation and encouraging osteoclast differentiation. Based on the correlation between RA, osteoporosis, and inflammation, it is possible that systemic immunosuppression with disease-modifying antirheumatic drugs (DMARDs) can help individuals with RA have a lower chance of developing osteoporosis and osteoporotic fractures. There is little information on how different DMARDs, biologic or non-biologic, affect RA patients' bone metabolism. In this study, we present an overview of the influence that targeted therapies, such as biologics, non-biologics, and small molecule inhibitors, have on bone homeostasis in RA patients.
Collapse
Affiliation(s)
- Hossein Azadeh
- Department of Internal Medicine, Rheumatology Division, Orthopedic Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
9
|
Serum cytokines and bone metabolic markers in patients with rheumatoid arthritis treated with biological disease modifying anti-rheumatic drugs. Clin Rheumatol 2023; 42:721-730. [PMID: 36163441 DOI: 10.1007/s10067-022-06390-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/30/2022] [Accepted: 09/21/2022] [Indexed: 11/03/2022]
Abstract
INTRODUCTION /objectives Several biological disease-modifying anti-rheumatic drugs (bDMARDs) have been widely used for the management of rheumatoid arthritis (RA). These drugs target different molecules important for the pathophysiology of RA; however, only a few studies have compared the effects of these biological drugs on cytokines and bone metabolic markers. The main aim of this study is to clarify the effects of bDMARDs with different modes of action on the cytokine and bone metabolic marker levels in patients with RA. METHODS Patients with RA who were initiated on infliximab, tocilizumab, or abatacept as the first bDMARD were prospectively enrolled in this study. Serum cytokine and bone metabolic marker levels were measured longitudinally, and changes in their levels were compared. RESULTS A total of 174 patients were enrolled in this study, with 55, 70, and 49 patients in the infliximab, tocilizumab, and abatacept groups, respectively. At six months, despite the similar clinical effectiveness of the three drugs, changes in the cytokine and bone metabolic marker levels were distinct; interferon-γ and tumor necrosis factor-α levels were significantly increased with infliximab, interleukin-6 levels were increased with tocilizumab, and interleukin-1β and interleukin-8 levels were increased with abatacept treatment. Bone-specific alkaline phosphatase and osteocalcin levels increased more significantly with tocilizumab than with infliximab, while osteopontin and osteonectin levels decreased with infliximab treatment. CONCLUSIONS bDMARDs with different modes of action exert different effects on the cytokine and bone metabolic marker levels in patients with RA.
Collapse
|
10
|
Biesemann N, Margerie D, Asbrand C, Rehberg M, Savova V, Agueusop I, Klemmer D, Ding-Pfennigdorff D, Schwahn U, Dudek M, Heyninck K, De Tavernier E, Cornelis S, Kohlmann M, Nestle FO, Herrmann M. Additive efficacy of a bispecific anti-TNF/IL-6 nanobody compound in translational models of rheumatoid arthritis. Sci Transl Med 2023; 15:eabq4419. [PMID: 36724239 DOI: 10.1126/scitranslmed.abq4419] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Rheumatoid arthritis (RA) is one of the most common autoimmune diseases affecting primarily the joints. Despite successful therapies including antibodies against tumor necrosis factor (TNF) and interleukin-6 (IL-6) receptor, only 20 to 30% of patients experience remission. We studied whether inhibiting both TNF and IL-6 would result in improved efficacy. Using backtranslation from single-cell RNA sequencing (scRNA-seq) data from individuals with RA, we hypothesized that TNF and IL-6 act synergistically on fibroblast-like synoviocytes (FLS) and T cells. Coculture of FLS from individuals with RA and T cells supported this hypothesis, revealing effects on both disease-driving pathways and biomarkers. Combining anti-TNF and anti-IL-6 antibodies in collagen-induced arthritis (CIA) mouse models resulted in sustained long-term remission, improved histology, and effects on bone remodeling pathways. These promising data initiated the development of an anti-TNF/IL-6 bispecific nanobody compound 1, with similar potencies against TNF and IL-6. We observed additive efficacy of compound 1 in a FLS/T cell coculture affecting arthritis and T helper 17 (TH17) pathways. This nanobody compound transcript signature inversely overlapped with described RA endotypes, indicating a potential efficacy in a broader patient population. In summary, we showed superiority of a bispecific anti-TNF/IL-6 nanobody compound or combination treatment over monospecific treatments in both in vitro and in vivo models. We anticipate improved efficacy in upcoming clinical studies.
Collapse
Affiliation(s)
- Nadine Biesemann
- Sanofi R&D, Immunology and Inflammation Therapeutic Area, Type 1/17 Immunology Cluster, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Daniel Margerie
- Sanofi R&D, Digital and Data Science, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Christian Asbrand
- Sanofi R&D, Immunology and Inflammation Therapeutic Area, Type 1/17 Immunology Cluster, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Markus Rehberg
- Sanofi R&D, Digital and Data Science, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Virginia Savova
- Precision Medicine and Computational Biology, Sanofi R&D, 350 Water St., Cambridge, MA 02141, USA
| | - Inoncent Agueusop
- Sanofi R&D, Biostatistics and Programming, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Daniel Klemmer
- Sanofi R&D, Biostatistics and Programming, Non-Clinical Efficacy and Safety, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Danping Ding-Pfennigdorff
- Sanofi R&D, Immunology and Inflammation Therapeutic Area, Type 1/17 Immunology Cluster, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Uwe Schwahn
- Sanofi R&D, Translational Medicine and Early Development, Biomarkers and Clinical Bioanalysis, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Markus Dudek
- Sanofi R&D, Translational Medicine and Early Development, Biomarkers and Clinical Bioanalysis, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | - Karen Heyninck
- Sanofi R&D, NANOBODY Research Platform, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Evelyn De Tavernier
- Sanofi R&D, NANOBODY Research Platform, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Sigrid Cornelis
- Sanofi R&D, NANOBODY Research Platform, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Markus Kohlmann
- Sanofi R&D, Early Clinical Development Therapeutic Area Immunology and Inflammation, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| | | | - Matthias Herrmann
- Sanofi R&D, Immunology and Inflammation Therapeutic Area, Type 1/17 Immunology Cluster, Industriepark Hoechst, 65926 Frankfurt am Main, Germany
| |
Collapse
|
11
|
Drobinski PJ, Nissen NI, Sinkeviciute D, Willumsen N, Karsdal MA, Bay-Jensen AC. In Contrast to Anti-CCP, MMP-Degraded and Citrullinated Vimentin (VICM) Is Both a Diagnostic and a Treatment Response Biomarker. Int J Mol Sci 2022; 24:ijms24010321. [PMID: 36613765 PMCID: PMC9820189 DOI: 10.3390/ijms24010321] [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: 11/01/2022] [Revised: 11/25/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
Protein citrullination and degradation by matrix metalloproteinases (MMP) plays a central role in the pathology of rheumatoid arthritis (RA). Autoantibodies are known to target citrullinated vimentin. The aim of this study was to investigate the relationship between the blood levels of MMP-degraded and citrullinated vimentin (VICM), as compared with the levels of MMP-degraded and non-citrullinated vimentin (VIM), and the standard anti-CCP biomarker in RA patients undergoing treatment. Thus, VIM, VICM and anti-CCP were quantified by ELISA in serum samples from baseline and week 8 of patients (n = 257) with RA, treated with either tocilizumab (8 mg/kg), methotrexate (7.5−15 mg/kg) or a placebo and compared with a reference cohort (n = 64). The three biomarkers were elevated in RA serum compared with the reference cohort: medians were 1.7 vs. 0.8 ng/mL (p < 0.05) for VIM; 7.5 vs. 0.7 ng/mL (p < 0.0001) for VICM; 57 vs. 4 RU/mL (p < 0.001) for anti-CCP. VICM was decreased in response to tocilizumab (2.9-fold, p < 0.0001) and to methotrexate (1.5-fold, p < 0.05) compared with the placebo, while anti-CCP was not. Serum VIM was also modulated by both drugs, although to a lesser degree. A high baseline level of VICM was predictive of a low disease activity response at week 8. In conclusion, VICM can differentiate between RA and healthy donors in a similar manner to anti-CCP; furthermore, VICM is also a pharmacodynamic marker.
Collapse
Affiliation(s)
- Patryk J. Drobinski
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, 2730 Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Neel I. Nissen
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
- Oncology, Nordic Bioscience, Herlev Hovedgade 207, 2730 Herlev, Denmark
| | - Dovile Sinkeviciute
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, 2730 Herlev, Denmark
| | | | - Morten A. Karsdal
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, 2730 Herlev, Denmark
- Oncology, Nordic Bioscience, Herlev Hovedgade 207, 2730 Herlev, Denmark
| | - Anne C. Bay-Jensen
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, 2730 Herlev, Denmark
- Correspondence:
| |
Collapse
|
12
|
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: 0.3] [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
|
13
|
Gao Y, Cai W, Zhou Y, Li Y, Cheng J, Wei F. Immunosenescence of T cells: a key player in rheumatoid arthritis. Inflamm Res 2022; 71:1449-1462. [DOI: 10.1007/s00011-022-01649-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/05/2022] Open
|
14
|
Narazaki M, Kishimoto T. Current status and prospects of IL-6–targeting therapy. Expert Rev Clin Pharmacol 2022; 15:575-592. [DOI: 10.1080/17512433.2022.2097905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Masashi Narazaki
- Department of Advanced Clinical and Translational Immunology, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Respiratory Medicine, Clinical Immunology, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tadamitsu Kishimoto
- Laboratory of Immune Regulation, World Premier International Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
15
|
Shiraishi K, Chiba K, Watanabe K, Oki N, Iwamoto N, Amano S, Yonekura A, Tomita M, Uetani M, Kawakami A, Osaki M. Analysis of bone erosions in rheumatoid arthritis using HR-pQCT: Development of a measurement algorithm and assessment of longitudinal changes. PLoS One 2022; 17:e0265833. [PMID: 35472146 PMCID: PMC9041818 DOI: 10.1371/journal.pone.0265833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/08/2022] [Indexed: 11/19/2022] Open
Abstract
Purpose
The purpose of this study was to establish an algorithm for measuring bone erosions at metacarpophalangeal (MCP) joints using high-resolution peripheral quantitative computed tomography (HR-pQCT), to investigate the precision of measurements, and to assess longitudinal changes in bone erosions among patients with rheumatoid arthritis (RA).
Methods
The 2nd and 3rd MCP joints were scanned at a voxel size of 60.7 μm using second-generation HR-pQCT. Bone erosions on MCP joints were identified using a semi-automated algorithm we developed, and each erosion parameter was measured. Measurement reproducibility was evaluated in 19 healthy subjects using intraclass correlation coefficients (ICCs) and root mean square percent coefficient of variance (RMS%CV). Finally, longitudinal changes in bone erosions over a period of 12 months were assessed in 26 patients with RA based on the calculated least significant change (LSC).
Results
Reproducibilities for measurement parameters regarding bone erosions with our algorithm were good (all ICCs ≥ 0.98; all RMS%CVs < 5%). No erosion parameters showed significant changes after 12 months of treatment in terms of median values in all erosions, while both progression and repair of erosions were observed individually (e.g., erosion volume: progression, 26% (+0.62 mm3); repair, 34% (-0.85 mm3); no change, 40%).
Conclusions
The measurement algorithm developed for bone erosions at MCP joints showed good reproducibility. Both progression and repair of bone erosions were observed in patients with RA even after 12 months of appropriate treatment. Our algorithm may be useful to investigate the etiology of RA and assess drug efficacy.
Collapse
Affiliation(s)
- Kazuteru Shiraishi
- 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
- * E-mail:
| | - Kounosuke Watanabe
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Nozomi Oki
- Department of Radiological Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naoki Iwamoto
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shoken Amano
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Akihiko Yonekura
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masato Tomita
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masataka Uetani
- Department of Radiological Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Atsushi Kawakami
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, 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
|
16
|
Soós B, Szentpétery Á, Raterman HG, Lems WF, Bhattoa HP, Szekanecz Z. Effects of targeted therapies on bone in rheumatic and musculoskeletal diseases. Nat Rev Rheumatol 2022; 18:249-257. [PMID: 35273387 DOI: 10.1038/s41584-022-00764-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2022] [Indexed: 12/17/2022]
Abstract
Generalized bone loss (osteoporosis) and fragility fractures can occur in rheumatic and musculoskeletal diseases including rheumatoid arthritis and spondyloarthritis (SpA; including ankylosing spondylitis and psoriatic arthritis). In addition, rheumatoid arthritis can involve localized, periarticular bone erosion and, in SpA, local (pathological) bone formation can occur. The RANK-RANKL-osteoprotegerin axis and the Wnt-β-catenin signalling pathway (along with its inhibitors sclerostin and Dickkopf 1) have been implicated in inflammatory bone loss and formation, respectively. Targeted therapies including biologic DMARDs and Janus kinase (JAK) inhibitors can stabilize bone turnover and inhibit radiographic joint damage, and potentially also prevent generalized bone loss. Targeted therapies interfere at various points in the mechanisms of local and generalized bone changes in systemic rheumatic diseases, and they effect biomarkers of bone resorption and formation, bone mass and risk of fragility fractures. Studies on the effects of targeted therapies on rates of fragility fracture are scarce. The efficacy of biologic DMARDs for arresting bone formation in axial SpA is debated. Improved understanding of the most relevant therapeutic targets and identification of important targeted therapies could lead to the preservation of bone in inflammatory rheumatic and musculoskeletal diseases.
Collapse
Affiliation(s)
- Boglárka Soós
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Szentpétery
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Department of Medical Sciences, Rheumatology, Uppsala University Hospital, Uppsala, Sweden
| | | | - Willem F Lems
- Amsterdam Rheumatology and Immunology Centre, Amsterdam, Netherlands
| | - Harjit P Bhattoa
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szekanecz
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
17
|
Maeda K, Yoshida K, Nishizawa T, Otani K, Yamashita Y, Okabe H, Hadano Y, Kayama T, Kurosaka D, Saito M. Inflammation and Bone Metabolism in Rheumatoid Arthritis: Molecular Mechanisms of Joint Destruction and Pharmacological Treatments. Int J Mol Sci 2022; 23:2871. [PMID: 35270012 PMCID: PMC8911191 DOI: 10.3390/ijms23052871] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/31/2022] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease characterized by a variety of symptoms and pathologies often presenting with polyarthritis. The primary symptom in the initial stage is joint swelling due to synovitis. With disease progression, cartilage and bone are affected to cause joint deformities. Advanced osteoarticular destruction and deformation can cause irreversible physical disabilities. Physical disabilities not only deteriorate patients' quality of life but also have substantial medical economic effects on society. Therefore, prevention of the progression of osteoarticular destruction and deformation is an important task. Recent studies have progressively improved our understanding of the molecular mechanism by which synovitis caused by immune disorders results in activation of osteoclasts; activated osteoclasts in turn cause bone destruction and para-articular osteoporosis. In this paper, we review the mechanisms of bone metabolism under physiological and RA conditions, and we describe the effects of therapeutic intervention against RA on bone.
Collapse
Affiliation(s)
- Kazuhiro Maeda
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Ken Yoshida
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Tetsuro Nishizawa
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Kazuhiro Otani
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Yu Yamashita
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Hinako Okabe
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Yuka Hadano
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Tomohiro Kayama
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Daitaro Kurosaka
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Mitsuru Saito
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| |
Collapse
|
18
|
CD34 +THY1 + synovial fibroblast subset in arthritic joints has high osteoblastic and chondrogenic potentials in vitro. Arthritis Res Ther 2022; 24:45. [PMID: 35168627 PMCID: PMC8845288 DOI: 10.1186/s13075-022-02736-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/02/2022] [Indexed: 12/12/2022] Open
Abstract
Objective Synovial fibroblasts (SFs) in rheumatoid arthritis (RA) and osteoarthritis (OA) play biphasic roles in joint destruction and regeneration of bone/cartilage as mesenchymal stem cells (MSCs). Although MSCs contribute to joint homeostasis, such function is impaired in arthritic joints. We have identified functionally distinct three SF subsets characterized by the expression of CD34 and THY1 as follows: CD34+THY1+, CD34−THY1−, and CD34−THY1+. The objective of this study was to clarify the differentiation potentials as MSCs in each SF subset since both molecules would be associated with the MSC function. Methods SF subsets were isolated from synovial tissues of 70 patients (RA: 18, OA: 52). Expressions of surface markers associated with MSCs (THY1, CD34, CD73, CD271, CD54, CD44, and CD29) were evaluated in fleshly isolated SF subsets by flow cytometry. The differentiation potentials of osteogenesis, chondrogenesis, and adipogenesis were evaluated with histological staining and a quantitative polymerase chain reaction of differentiation marker genes. Small interfering RNA was examined to deplete THY1 in SFs. Results The expression levels of THY1+, CD73+, and CD271+ were highest and those of CD54+ and CD29+ were lowest in CD34+THY1+ among three subsets. Comparing three subsets, the calcified area, alkaline phosphatase (ALP)-stained area, and cartilage matrix subset were the largest in the CD34+THY1+ subset. Consistently, the expressions of differentiation markers of the osteoblasts (RUNX2, ALPL, and OCN) or chondrocytes (ACAN) were the highest in the CD34+THY1+ subset, indicating that the CD34+THY1+ subset possessed the highest osteogenic and chondrogenic potential among three subsets, while the differentiation potentials to adipocytes were comparable among the subsets regarding lipid droplet formations and the expression of LPL and PPARγ. The knockdown of THY1 in bulk SFs resulted in impaired osteoblast differentiation indicating some functional aspects in this stem-cell marker. Conclusion The CD34+THY1+ SF subset has high osteogenic and chondrogenic potentials. The preferential enhancement of MSC functions in the CD34+THY1+ subset may provide a new treatment strategy for regenerating damaged bone/cartilage in arthritic joints. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02736-7.
Collapse
|
19
|
Jarlborg M, Gabay C. Systemic effects of IL-6 blockade in rheumatoid arthritis beyond the joints. Cytokine 2021; 149:155742. [PMID: 34688020 DOI: 10.1016/j.cyto.2021.155742] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/13/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022]
Abstract
Interleukin (IL)-6 is produced locally in response to an inflammatory stimulus, and is able to induce systemic manifestations at distance from the site of inflammation. Its unique signaling mechanism, including classical and trans-signaling pathways, leads to a major expansion in the number of cell types responding to IL-6. This pleiotropic cytokine is a key factor in the pathogenesis of rheumatoid arthritis (RA) and is involved in many extra-articular manifestations that accompany the disease. Thus, IL-6 blockade is associated with various biological effects beyond the joints. In this review, the systemic effects of IL-6 in RA comorbidities and the consequences of its blockade will be discussed, including anemia of chronic disease, cardiovascular risks, bone and muscle functions, and neuro-psychological manifestations.
Collapse
Affiliation(s)
- Matthias Jarlborg
- Division of Rheumatology, University Hospital of Geneva, and Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland; VIB-UGent Center for Inflammation Research and Ghent University, Ghent, Belgium
| | - Cem Gabay
- Division of Rheumatology, University Hospital of Geneva, and Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland.
| |
Collapse
|
20
|
Kondo N, Kuroda T, Kobayashi D. Cytokine Networks in the Pathogenesis of Rheumatoid Arthritis. Int J Mol Sci 2021; 22:ijms222010922. [PMID: 34681582 PMCID: PMC8539723 DOI: 10.3390/ijms222010922] [Citation(s) in RCA: 204] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic systemic inflammation causing progressive joint damage that can lead to lifelong disability. The pathogenesis of RA involves a complex network of various cytokines and cells that trigger synovial cell proliferation and cause damage to both cartilage and bone. Involvement of the cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-6 is central to the pathogenesis of RA, but recent research has revealed that other cytokines such as IL-7, IL-17, IL-21, IL-23, granulocyte macrophage colony-stimulating factor (GM-CSF), IL-1β, IL-18, IL-33, and IL-2 also play a role. Clarification of RA pathology has led to the development of therapeutic agents such as biological disease-modifying anti-rheumatic drugs (DMARDs) and Janus kinase (JAK) inhibitors, and further details of the immunological background to RA are emerging. This review covers existing knowledge regarding the roles of cytokines, related immune cells and the immune system in RA, manipulation of which may offer the potential for even safer and more effective treatments in the future.
Collapse
Affiliation(s)
- Naoki Kondo
- Division of Orthopedic Surgery, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi-Dori, Chuo-ku, Niigata City 951-8510, Japan;
| | - Takeshi Kuroda
- Health Administration Center, Niigata University, 2-8050 Ikarashi, Nishi-ku, Niigata City 950-2181, Japan
- Correspondence: ; Tel.: +81-25-262-6244; Fax: +81-25-262-7517
| | - Daisuke Kobayashi
- Division of Clinical Nephrology and Rheumatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi-Dori, Chuo-ku, Niigata City 951-8510, Japan;
| |
Collapse
|
21
|
Platzer A, Alasti F, Smolen JS, Aletaha D, Radner H, Blüml S. Trajectory clusters of radiographic progression in patients with rheumatoid arthritis: associations with clinical variables. Ann Rheum Dis 2021; 81:175-183. [PMID: 34376384 DOI: 10.1136/annrheumdis-2021-220331] [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: 03/10/2021] [Accepted: 07/29/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Identification of trajectories of radiographic damage in rheumatoid arthritis (RA) by clustering patients according to the shape of their curve of Sharp-van der Heijde scores (SHSs) over time. Developing models to predict their progression cluster from baseline characteristics. METHODS Patient-level data over a 2-year period from five large randomised controlled trials on tumour necrosis factor inhibitors in RA were used. SHSs were clustered in a shape-respecting manner to identify distinct clusters of radiographic progression. Characteristics of patients within different progression clusters were compared at baseline and over time. Logistic regression models were developed to predict trajectory of radiographic progression using information at baseline. RESULTS In total, 1887 patients with 7738 X-rays were used for cluster analyses. We identified four distinct clusters with characteristic shapes of radiographic progression: one with a stable SHS over the whole 2-year period (C0/lowChange; 86%); one with relentless progression (C1/rise; 5.8%); one with decreasing SHS (C2/improvement; 6.9%); one going up and down (C3/bothWays; 1.4%) of the SHS. Robustness of clusters were confirmed using different clustering methods. Regression models identified disease duration, baseline C-reactive protein (CRP) and SHS and treatment status as predictors for cluster assignment. CONCLUSIONS We were able to identify and partly characterise four different clusters of radiographic progression over time in patients with RA, most remarkably one with relentless progression and another one with amelioration of joint damage over time, suggesting the existence of distinct patterns of joint damage accrual in RA.
Collapse
Affiliation(s)
- Alexander Platzer
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| | - Farideh Alasti
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| | - Josef S Smolen
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| | - Daniel Aletaha
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| | - Helga Radner
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| | - Stephan Blüml
- Department of Rheumatology, Medical University of Vienna, Wien, Vienna, Austria
| |
Collapse
|
22
|
Takeuchi T, Yoshida H, Tanaka S. Role of interleukin-6 in bone destruction and bone repair in rheumatoid arthritis. Autoimmun Rev 2021; 20:102884. [PMID: 34229044 DOI: 10.1016/j.autrev.2021.102884] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022]
Abstract
Rheumatoid arthritis (RA) is a common inflammatory form of arthritis leading to the progressive bone and joint destruction. Many factors are closely involved in the pathology of RA, in particular bone-related cells and inflammatory cytokines such as TNF-α and interleukin-6 (IL-6). Because RA patients with progressive bone destruction experience accelerated deterioration of their quality of life, inhibition of disease progression and joint destruction has become an important clinical goal. Recent studies have also found that drug intervention targeting proinflammatory cytokines such as IL-6 results in bone repair in addition to suppression of bone and joint destruction, and these results suggest the potential for new therapeutic goals. Regarding the relationship between IL-6 and bone destruction, essential roles of osteoclasts have been reported over many years; however, more recent studies have explored the relationship of IL-6 with osteoblasts and osteocytes. In this review, we highlight the perspectives of basic and clinical research, adding new findings on the relationships between IL-6 and bone-related cells associated with inflammation, and the possibility of bone repair by blocking IL-6.
Collapse
Affiliation(s)
- Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Hiroto Yoshida
- Chugai Pharmaceutical Co. Ltd., 200 Kajiwara, Kamakura City, Kanagawa, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
23
|
Figueiredo CP, Perez MO, Sales LP, Caparbo VDF, Pereira RMR. Evaluation of bone erosion in rheumatoid arthritis patients by high-resolution peripheral quantitative computed tomography scans: Comparison between two semi-automated programs in a three-dimensional setting. Int J Rheum Dis 2021; 24:948-953. [PMID: 34151526 DOI: 10.1111/1756-185x.14157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/07/2021] [Accepted: 05/20/2021] [Indexed: 01/01/2023]
Abstract
AIM The aim of this study was to compare OsiriX software with the previous published Medical Image Analysis Framework (MIAF) method to assess the volume of erosion in patients with rheumatoid arthritis (RA). METHODS Forty RA patients underwent high-resolution peripheral quantitative computed tomography scans of the second and third metacarpophalangeal joints, and thirty-four patients with any bone erosion were enrolled. Two techniques were applied to erosion evaluation: (a) semi-automated MIAF software, and (b) semi-automated segmentation by free open-source Digital Imaging and Communications in Medicine viewer, OsiriX software. MIAF has been published before, but this is the first time that OsiriX has been used in this way in rheumatology. Bland & Altman plots described agreement between methods. RESULTS Forty-eight erosions from 34 patients were analyzed. Mean age was 40.74 ± 5.32 years and mean disease duration was 10.68 ± 4.96 years. Both methods demonstrated a strong correlation regarding erosion volume (r = 0.96, P < 0.001). Median (interquartile range) of erosion volume was 12.14 (4.5-36.07) when MIAF was considered, and 11.80 (3.45-29.42) when the OsiriX tool was used (P = 0.139). MIAF and OsiriX showed good agreement when the Bland & Altman plot was performed. Evaluation by MIAF took 22.69 ± 6.71 minutes, whereas OsiriX took only 2.62 ± 1.09 minutes (P < 0.001). CONCLUSION The three-dimensional segmentation of bone erosions can be done by both MIAF and OsiriX software with good agreement. However, because OsiriX is a widespread tool and faster, its method seems to be more feasible for evaluating peripheral bone damage, especially bone erosions.
Collapse
Affiliation(s)
- Camille Pinto Figueiredo
- Bone Metabolism Laboratory, Rheumatology Division, Faculdade de Medicina FMUSP Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Mariana Ortega Perez
- Bone Metabolism Laboratory, Rheumatology Division, Faculdade de Medicina FMUSP Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Lucas Peixoto Sales
- Bone Metabolism Laboratory, Rheumatology Division, Faculdade de Medicina FMUSP Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Valeria de Falco Caparbo
- Bone Metabolism Laboratory, Rheumatology Division, Faculdade de Medicina FMUSP Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Rosa Maria Rodrigues Pereira
- Bone Metabolism Laboratory, Rheumatology Division, Faculdade de Medicina FMUSP Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
24
|
Yokota K, Sato K, Miyazaki T, Aizaki Y, Tanaka S, Sekikawa M, Kozu N, Kadono Y, Oda H, Mimura T. Characterization and Function of Tumor Necrosis Factor and Interleukin-6-Induced Osteoclasts in Rheumatoid Arthritis. Arthritis Rheumatol 2021; 73:1145-1154. [PMID: 33512089 PMCID: PMC8361923 DOI: 10.1002/art.41666] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 01/21/2021] [Indexed: 12/18/2022]
Abstract
Objective We have previously reported that stimulation of mouse bone marrow–derived macrophages with tumor necrosis factor (TNF) and interleukin‐6 (IL‐6) induces differentiation of osteoclast‐like cells. We undertook this study to clarify the characterization and function of human TNF and IL‐6–induced osteoclasts using peripheral blood collected from patients with rheumatoid arthritis (RA) and healthy donors. Methods Peripheral blood monocytes were cultured with a combination of TNF and IL‐6, TNF alone, IL‐6 alone, or with RANKL, and their bone resorption ability was evaluated. Expression levels of NFATc1, proinflammatory cytokines, and matrix metalloproteinase 3 were analyzed. The effects of NFAT inhibitor and JAK inhibitor were examined. Furthermore, the relationship between the number of TNF and IL‐6–induced osteoclasts or RANKL‐induced osteoclasts differentiated from peripheral blood mononuclear cells (PBMCs) in patients with RA and the modified total Sharp score (mTSS) or whole‐body bone mineral density (BMD) was examined. Results Peripheral blood monocytes stimulated with a TNF and IL‐6–induced osteoclasts were shown to demonstrate the ability to absorb bone matrix. Cell differentiation was not inhibited by the addition of osteoprotegerin. Stimulation with a combination of TNF and IL‐6 promoted NFATc1 expression, whereas the NFAT and JAK inhibitors prevented TNF and IL‐6–induced osteoclast formation. Expression levels of IL1β, TNF, IL12p40, and MMP3 were significantly increased in TNF and IL‐6–induced osteoclasts, but not in RANKL‐induced osteoclasts. The number of TNF and IL‐6–induced osteoclasts differentiated from PBMCs in patients with RA positively correlated with the mTSS, whereas RANKL‐induced osteoclast numbers negatively correlated with the whole‐body BMD of the same patients. Conclusion Our results demonstrate that TNF and IL‐6–induced osteoclasts may contribute to the pathology of inflammatory arthritis associated with joint destruction, such as RA.
Collapse
Affiliation(s)
- Kazuhiro Yokota
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama Medical University, Saitama, Japan
| | - Kojiro Sato
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | | | - Yoshimi Aizaki
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama Medical University, Saitama, Japan
| | - Shinya Tanaka
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
| | - Miyoko Sekikawa
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
| | | | - Yuho Kadono
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama Medical University, Saitama, Japan
| | - Hiromi Oda
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
| | - Toshihide Mimura
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, Saitama Medical University, Saitama, Japan
| |
Collapse
|
25
|
Potere N, Batticciotto A, Vecchié A, Porreca E, Cappelli A, Abbate A, Dentali F, Bonaventura A. The role of IL-6 and IL-6 blockade in COVID-19. Expert Rev Clin Immunol 2021; 17:601-618. [PMID: 33874829 DOI: 10.1080/1744666x.2021.1919086] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces a dysregulated hyperinflammatory response. AREAS COVERED Authors review evidence on IL-6 and IL-6 blockade in coronavirus disease 2019 (COVID-19) and discuss the pathophysiological and prognostic roles of this cytokine and the clinical impact of pharmacological blockade of IL-6 . The material includes original articles and reviews published from March 2020 to March 2021 and searched on PubMed, medRxiv, and bioRxiv. EXPERT OPINION IL-6 is one of the most prominent pro-inflammatory cytokines. Increased levels are recorded in COVID-19 patients, especially those with severe-to-critical disease. Evidence is accumulating on the relevance of IL-6 as a prognostic marker in COVID-19. Since IL-6 is a druggable target for several inflammatory diseases, pharmacological blockers of the IL-6 signaling pathway were repurposed to blunt the abnormal SARS-CoV-2-induced cytokine release. Data are limited to few randomized controlled trials that reported encouraging, though not conclusive, results, indicating the usefulness of IL-6 blockade early in the course of the disease in patients with hyperinflammation and no or limited organ damage. Further research is warranted to explore the role of IL-6 in different COVID-19 phenotypes and identify subgroups of patients who may mostly benefit from IL-6 pathway inhibition.
Collapse
Affiliation(s)
- Nicola Potere
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Alberto Batticciotto
- Rheumatology Unit, Internal Medicine Department, ASST Sette Laghi, Ospedale Di Circolo - Fondazione Macchi, Varese, Italy
| | | | - Ettore Porreca
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Antonella Cappelli
- Rheumatology Unit, Internal Medicine Department, ASST Sette Laghi, Ospedale Di Circolo - Fondazione Macchi, Varese, Italy
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Francesco Dentali
- Department of Medicine and Surgery, Insubria University, Varese, Italy
| | - Aldo Bonaventura
- Department of Internal Medicine, ASST Sette Laghi, Varese, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
26
|
Brunet SC, Tse JJ, Kuczynski MT, Engelke K, Boyd SK, Barnabe C, Manske SL. Heterogenous bone response to biologic DMARD therapies in rheumatoid arthritis patients and their relationship to functional indices. Scand J Rheumatol 2021; 50:417-426. [PMID: 33775211 DOI: 10.1080/03009742.2020.1869303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Objectives: Previous studies of high-resolution peripheral quantitative computed tomography (HR-pQCT) imaging of hand joints in patients with rheumatoid arthritis (RA) have suggested that erosion healing may occur. Our objective was to examine changes in erosion volume, joint space width (JSW), bone mineral density (BMD), and bone remodelling, and their association with clinical outcomes and measures of patient hand function.Method: We examined 48 patients who achieved a good response to a newly initiated biologic therapy. HR-pQCT images of the dominant hands' second and third metacarpophalangeal joints were obtained 3 and 12 months after therapy initiation. Bone erosion volume, JSW, BMD, and bone remodelling were quantified from HR-pQCT images, with improvement, no change (unchanged), or progression in these measures determined by least significant change. Disease activity and hand function measures were collected.Results: There were no significant group changes in HR-pQCT outcomes over the 9 month period. Twenty-two patients had total erosion volumes that remained unchanged, nine showed improvement, and two progressed. The majority of JSW and BMD measures remained unchanged. There was a significant association between the baseline Health Assessment Questionnaire score and the change in minimum JSW, but no other significant associations between HR-pQCT outcomes and function were observed.Conclusions: The vast majority of patients maintained unchanged JSW and BMD over the course of follow-up. Significant improvements in total erosion volume occurred in 27% of patients, suggesting that biologic therapies may lead to erosion healing in some patients, although this did not have an impact on self-reported and demonstrated hand function.
Collapse
Affiliation(s)
- S C Brunet
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Biomedical Engineering Graduate Program, Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - J J Tse
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - M T Kuczynski
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Biomedical Engineering Graduate Program, Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - K Engelke
- Department of Medicine, FAU University Erlangen-Nürnberg and University Clinic Erlangen, Erlangen, Germany
| | - S K Boyd
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Biomedical Engineering Graduate Program, Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - C Barnabe
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - S L Manske
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Biomedical Engineering Graduate Program, Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
27
|
Miao C, Bai L, Yang Y, Huang J. Dysregulation of lncRNAs in Rheumatoid Arthritis: Biomarkers, Pathogenesis and Potential Therapeutic Targets. Front Pharmacol 2021; 12:652751. [PMID: 33776780 PMCID: PMC7994855 DOI: 10.3389/fphar.2021.652751] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease of unknown etiology, mainly manifested by persistent abnormal proliferation of fibroblast-like synoviocytes (FLSs), inflammation, synovial hyperplasia and cartilage erosion, accompanied by joint swelling and joint destruction. Abnormal expression or function of long noncoding RNAs (lncRNAs) are closely related to human diseases, including cancers, mental diseases, autoimmune diseases and others. The abnormal sequence and spatial structure of lncRNAs, the disorder expression and the abnormal interaction with the binding protein will lead to the change of gene expression in the way of epigenetic modification. Increasing evidence demonstrated that lncRNAs were involved in the activation of FLSs, which played a key role in the pathogenesis of RA. In this review, the research progress of lncRNAs in the pathogenesis of RA was systematically summarized, including the role of lncRNAs in the diagnosis of RA, the regulatory mechanism of lncRNAs in the pathogenesis of RA, and the intervention role of lncRNAs in the treatment of RA. Furthermore, the activated signal pathways, the role of DNA methylation and other mechanism have also been overview in this review.
Collapse
Affiliation(s)
- Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Anhui Provincial Key Laboratory of Chinese Medicine Compound, Anhui University of Chinese Medicine, Hefei, China.,Department of Pharmacy, School of Life and Health Sciences, Anhui University of Science and Technology, Fengyang, China
| | - Liangliang Bai
- Department of Biomedical Engineering, School of Biomedical Engineering, Anhui Medical University, Hefei, China
| | - Yaru Yang
- Department of Pharmacy, First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Jinling Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
28
|
IL-6R blockade by chikusetsusaponin IVa butyl ester inhibits Th17 cell differentiation and ameliorates collagen-induced arthritis. Cell Mol Immunol 2021; 18:1584-1586. [PMID: 33637959 DOI: 10.1038/s41423-021-00651-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 11/08/2022] Open
|
29
|
Chen JF, Hsu CY, Yu SF, Ko CH, Chiu WC, Lai HM, Chen YC, Su YJ, Cheng TT. The impact of long-term biologics/target therapy on bone mineral density in rheumatoid arthritis: a propensity score-matched analysis. Rheumatology (Oxford) 2021; 59:2471-2480. [PMID: 31984422 PMCID: PMC7449814 DOI: 10.1093/rheumatology/kez655] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/29/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES To investigate changes in BMD in RA patients receiving 3-year biological/targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARD) or conventional synthetic DMARD (csDMARD). METHODS Patients with RA were recruited from September 2014 until March 2019. Clinical characteristics, BMD and evidence of fragility fractures at enrolment were documented. Participants were treated according to the National Institute for Health and Care Excellence (NICE) guidelines over a 3-year observation period. Repeated BMD was measured at the end of the study period. Participants were grouped into those receiving b/tsDMARD or csDMARD and by propensity score matching (1:2). RESULTS A total of 388 participants completed the 3-year follow-up. After propensity score matching, 92 and 184 participants were allocated to the b/tsDMARD (Group I) and csDMARD (Group II), respectively. After 3 years, BMD remained stable at the femoral neck (FN), hip (total) (TH) and lumbar vertebra (L1-4) (P =0.09, 0.15, 0.87) in Group I. However, BMD decreased significantly in Group II (P=0.045, <0.001, 0.004) at corresponding sites. Participants receiving combined b/tsDMARD and anti-osteoporosis therapy experienced a greater BMD preserving effect than other subgroups. CONCLUSION Long-term b/tsDMARDs therapy had protective effects on bone loss for patients with RA. Patients receiving concomitant anti-osteoporosis therapy and b/tsDMARDs therapy experienced the greatest BMD preserving effect.
Collapse
Affiliation(s)
- Jia-Feng Chen
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan.,Department of Internal Medicine, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chung-Yuan Hsu
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan.,Department of Internal Medicine, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shan-Fu Yu
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan.,Department of Internal Medicine, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chi-Hua Ko
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan
| | - Wen-Chan Chiu
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan
| | - Han-Ming Lai
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan
| | - Ying-Chou Chen
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan.,Department of Internal Medicine, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Jih Su
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan.,Department of Internal Medicine, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tien-Tsai Cheng
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital Kaohsiung, Taiwan.,Department of Internal Medicine, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
30
|
Hirabayashi Y. Tocilizumab can efficiently prevent bone destruction in patients with recent-onset rheumatoid arthritis. Mod Rheumatol 2021; 31:966-971. [PMID: 33274666 DOI: 10.1080/14397595.2020.1859674] [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] [Indexed: 10/22/2022]
Abstract
OBJECTIVES To assess whether tocilizumab (TCZ) can prevent bone destruction in patients with recent-onset rheumatoid arthritis (RA). METHODS DAS28-ESR and van der Heijde-modified Sharp score (mTSS) were evaluated in 50 patients who received TCZ within 1 year from the onset of RA. TCZ was consecutively administered during the observation period within the first 2 years. In 15 patients, mTSS could be evaluated at 5 years. RESULTS The mean DAS28-ESR at baseline, 1, 2, and 5 years was 4.86, 1.29, 1.19, and 1.18, respectively. The change in mTSS (ΔmTSS) between baseline and 2 years was -0.33. The structural remission rates (ΔmTSS/year ≤0.5) were 91.8% and 92.7% during the first and second years, respectively. Only one increase in erosion score was observed in the first year in 2 patients and the erosion score of all patients did not increase in the second year. In 15 patients, the ΔmTSS over 5 years was 0.80, corresponding to 0.16 per year. The structural remission rate at 5 years (ΔmTSS ≤2.5) was 93.3%. The erosion score was 0 in all 15 patients at 5 years, indicating that bone destruction did not become apparent. CONCLUSION TCZ can efficiently prevent bone destruction in patients with recent-onset RA.
Collapse
|
31
|
Drobinski PJ, Bay-Jensen AC, Karsdal MA, Sardar S, Siebuhr AS. Connective tissue remodelling is differently modulated by tocilizumab versus methotrexate monotherapy in patients with early rheumatoid arthritis: the AMBITION study. Arthritis Res Ther 2021; 23:13. [PMID: 33413588 PMCID: PMC7789531 DOI: 10.1186/s13075-020-02378-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/24/2020] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Associations between rheumatoid arthritis (RA) and effect of treatment at the tissue levels are poorly understood. We investigated the scope of released extracellular matrix (ECM) metabolites as a consequence of tissue remodelling in patients treated with methotrexate (MTX) and tocilizumab (TCZ) compared to placebo. METHODS Tissue metabolites from 387 RA patients treated with either TCZ (8 mg/kg) or MTX monotherapy (7.5-20 mg/kg) were measured at baseline and 8 weeks sera by validated ELISA assays. The levels of collagen biomarkers (C1M, C2M, C3M and C4M) together with C-reactive protein (CRP) and CRP metabolite (CRPM) were investigated. Baseline levels of biomarkers have been compared with 72 age- and gender-matched healthy controls. Comparison between treatment and response groups were done by ANCOVA, Spearman's correlation and logistic regression adjusted for age, gender, BMI and disease duration. RESULTS C1M and C3M were significantly (P < 0.05) inhibited by TCZ and C3M by MTX (P < 0.01) compared to placebo. C1M and C3M inhibition with TCZ was respectively 23% and 16% greater than that of MTX (P < 0.01 and P < 0.0001). C4M was inhibited by TCZ and MTX, but the effect of TCZ was 22% greater than MTX (P < 0.0001). TCZ and MTX had minimal effect on C2M levels. MTX had no effect on CRP and CRPM, whereas TCZ reduced their levels to 69% and 27% from baseline. Investigated biomarkers revealed a significant (P < 0.05) difference in biomarker profiles of MTX ACR50 treatment responders and non-responders. Change to week 8 in levels of C3M, C4M, CRP and CRPM in MTX patients correlated significantly (rho = 0.41 to 0.18, P < 0.0001 to 0.039) with change in disease activity (DAS28) at weeks 8, 16 and 24, whereas only CRP in TCZ patients (rho = 0.32 to 0.21, P < 0.0001 to 0.01). CONCLUSION Patients receiving TCZ treatment for 8 weeks had higher suppression of tissue remodelling and inflammatory biomarkers over patients treated with MTX. Measured biomarkers enabled for a discrimination of biomarker profiles of ACR50 treatment responding patients and identification of those who benefit at the early time point. Week 8 change in levels of C3M, C4M, CRP and CRPM significantly predicted clinical response to treatment and correlated with DAS28 at all time points. TRIAL REGISTRATION ClinicalTrials.gov, NCT00109408 . Date of registration: July 2005. Name of the registry: A Study to Assess the Safety and Efficacy of Tocilizumab in Patients with Active Rheumatoid Arthritis.
Collapse
Affiliation(s)
- Patryk J. Drobinski
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Anne C. Bay-Jensen
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Morten A. Karsdal
- Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Samra Sardar
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| | - Anne S. Siebuhr
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 207, DK-2730 Herlev, Denmark
| |
Collapse
|
32
|
Gandikota G, Fakuda T, Finzel S. Computed tomography in rheumatology - From DECT to high-resolution peripheral quantitative CT. Best Pract Res Clin Rheumatol 2020; 34:101641. [PMID: 33281053 DOI: 10.1016/j.berh.2020.101641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this chapter, we discuss current updates and applications of Dual Energy Computed Tomography (DECT), iodine-DECT mapping, and high-resolution peripheral quantitative CT (HR-pQCT) in rheumatology. DECT provides a noninvasive diagnosis of gout and can help to differentiate gout from CPPD. Accuracy of DECT varies in various stages of gout. DECT needs specialized hardware, software, and skilled post-processing and interpretation. Sensitivity reduces significantly with deeper tissues such as hip and shoulder. Iodine map enables to delineate inflammatory lesions such as capsulitis and tenosynovitis by improving iodine contrast. Iodine quantification with an iodine map is a promising objective method to evaluate therapeutic effect of inflammatory arthritis. HR-pQCT allows for highly sensitive and specific measures of bone erosions and osteophytes in inflammatory joint diseases, documenting change over time, e.g. in cohorts undergoing immunosuppressive treatments. However, assessing the images requires trained readers, and (semi)-automated scripts to detect bone damage are still undergoing validation and further development.
Collapse
Affiliation(s)
- Girish Gandikota
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA.
| | - Takeshi Fakuda
- Department of Radiology, The Jikei University School of Medicine, Japan
| | - Stephanie Finzel
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
33
|
Cheng Q, Wu H, Du Y. The roles of small-molecule inflammatory mediators in rheumatoid arthritis. Scand J Immunol 2020; 93:e12982. [PMID: 33025632 DOI: 10.1111/sji.12982] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial inflammation and joint destruction. Although great progress has been made in the treatment of RA with antagonists of pro-inflammatory cytokines such as TNF-α, IL-6 and IL-1, the disease remains refractory in some patients. Previous studies have found that small-molecule inflammatory mediators, such as prostaglandins, leukotrienes, reactive oxygen species, nitric oxide, lipoxins and platelet-activating factor, play a significant role in the development of RA. Such compounds help to induce, maintain or reduce inflammation and could therefore be potential therapeutic targets. In this review, we describe the roles of various classes of small-molecule inflammatory mediators in RA and discuss the effects of some drugs that modulate their activity. Many drugs targeting these mediators have demonstrated good efficacy in mouse models of RA but not in patients. However, it is clear that many small-molecule inflammatory mediators play key roles in the pathogenesis of RA, and a better understanding of the underlying molecular pathways may assist in the development of targeted therapies that are efficacious in RA patients.
Collapse
Affiliation(s)
- Qi Cheng
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Department of Clinic Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huaxiang Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Du
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
34
|
Shin A, Park EH, Dong YH, Ha YJ, Lee YJ, Lee EB, Song YW, Kang EH. Comparative risk of osteoporotic fracture among patients with rheumatoid arthritis receiving TNF inhibitors versus other biologics: a cohort study. Osteoporos Int 2020; 31:2131-2139. [PMID: 32514765 DOI: 10.1007/s00198-020-05488-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022]
Abstract
UNLABELLED In this population-based cohort study on comparative osteoporotic fracture risks between different biologic disease-modifying drugs among patients with rheumatoid arthritis (RA), we did not find a significant difference in the risk of osteoporotic fractures between RA patients receiving TNF inhibitors versus abatacept or tocilizumab. INTRODUCTION We aimed to investigate the comparative risk of osteoporotic fractures between rheumatoid arthritis (RA) patients who initiated TNF inhibitors (TNFis) versus abatacept or tocilizumab. METHODS Using the Korea National Health Insurance Service datasets from 2002 to 2016, RA patients who initiated TNFis, abatacept, or tocilizumab were identified. The primary outcome was a composite end point of non-vertebral fractures and hospitalized vertebral fractures; secondary outcomes were two components of the primary outcome and fractures occurring at the humerus/forearm. Propensity score (PS) matching with a variable ratio up to 10 TNFi initiators per 1 comparator drug initiator was used to adjust for > 50 baseline confounders. We estimated hazard ratios (HRs) and 95% confidence interval (CI) of fractures comparing TNFi initiators to abatacept and to tocilizumab by Cox proportional hazard models stratified by a matching ratio. RESULTS After PS-matching, 2307 TNFi initiators PS-matched on 588 abatacept initiators, and 2462 TNFi initiators on 640 tocilizumab initiators were included. A total of 77 fractures occurred during a mean follow-up of 454 days among TNFi and abatacept initiators and 83 fractures during 461 days among TNFi and tocilizumab initiators. The PS-matched HR (95% CI) was 0.91 (0.48-1.71) comparing TNFi versus abatacept initiators, and 1.00 (0.55-1.83) comparing TNFi versus tocilizumab initiators. Analysis on vertebral and non-vertebral fractures showed similar results. CONCLUSIONS In this nationally representative cohort, we did not find a significant difference in the risk of fractures between TNFi initiators versus abatacept or tocilizumab among RA patients.
Collapse
Affiliation(s)
- A Shin
- Division of Rheumatology Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - E H Park
- Division of Rheumatology Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Y-H Dong
- Faculty of Pharmacy School of Pharmaceutical Science, National Yang-Ming University, Taipei, Taiwan
- Institute of Public Health, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Y-J Ha
- Division of Rheumatology Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Y J Lee
- Division of Rheumatology Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - E B Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Y W Song
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Medicine, Seoul National University, Seoul, South Korea
| | - E H Kang
- Division of Rheumatology Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.
| |
Collapse
|
35
|
Tocilizumab Was Effective in Repairing the Large Geode in a Patient with Rheumatoid Arthritis. Case Rep Rheumatol 2020; 2020:8899391. [PMID: 32908770 PMCID: PMC7468654 DOI: 10.1155/2020/8899391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 11/17/2022] Open
Abstract
Rheumatoid arthritis is characterized by multiple chronic arthritis subsequently inducing joint destruction. Although subchondral geode is a well-known feature of high-disease activity, a large geode is rare. Moreover, the treatment effect of biologic agents in the repair of large geode has not been reported. The present report shows the significant effect of interleukin-6 receptor blocker, tocilizumab, in repairing the large geode in the left humeral lateral epicondyle. This case implies that tocilizumab might be an effective treatment in patients with rheumatoid arthritis even with large geode.
Collapse
|
36
|
Figueiredo CP, Perez MO, Sales LP, Schett G, Pereira RMR. HR-pQCT in vivo imaging of periarticular bone changes in chronic inflammatory diseases: Data from acquisition to impact on treatment indications. Mod Rheumatol 2020; 31:294-302. [PMID: 32735144 DOI: 10.1080/14397595.2020.1804669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Imaging is essential for the assessment of bone and inflammatory joint diseases. There are several imaging techniques available that differ regarding resolution, radiation exposure, time expending, precision, cost, availability or ability to predict disease progression. High-resolution peripheral quantitative computed tomography (HR-pQCT) that was introduced in 2004 allows the in vivo evaluation of peripheral bone microarchitecture and demonstrated high precision in assessing bone changes in inflammatory musculoskeletal diseases. This review summarizes the use of HR-pQCT for the evaluation of the hand skeleton in inflammatory joint diseases. We conducted a review of the literature regarding the protocols that involve hand joints assessment and evaluation of bone changes as erosions and osteophytes in chronic inflammatory diseases. Apart from measuring bone density and structure of the radius and the tibia, HR-pQCT has contributed to assessment of bone erosions and osteophytes, considered the hallmark of diseases as rheumatoid arthritis and psoriatic arthritis, respectively. In this way, there are some conventions recently established by rheumatic study groups that we just summarized here in order to standardize HR-pQCT measurements.
Collapse
Affiliation(s)
- Camille P Figueiredo
- Bone Metabolism Laboratory, Rheumatology Division, Faculdade de Medicina, FMUSP da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Mariana O Perez
- Bone Metabolism Laboratory, Rheumatology Division, Faculdade de Medicina, FMUSP da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Lucas Peixoto Sales
- Bone Metabolism Laboratory, Rheumatology Division, Faculdade de Medicina, FMUSP da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, University of Erlangen-Nuremberg and Universitatsklinikum, Erlangen, Germany
| | - Rosa M R Pereira
- Bone Metabolism Laboratory, Rheumatology Division, Faculdade de Medicina, FMUSP da Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
37
|
Klose-Jensen R, Tse JJ, Keller KK, Barnabe C, Burghardt AJ, Finzel S, Tam LS, Hauge EM, Stok KS, Manske SL. High-Resolution Peripheral Quantitative Computed Tomography for Bone Evaluation in Inflammatory Rheumatic Disease. Front Med (Lausanne) 2020; 7:337. [PMID: 32766262 PMCID: PMC7381125 DOI: 10.3389/fmed.2020.00337] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/05/2020] [Indexed: 12/25/2022] Open
Abstract
High resolution peripheral quantitative computed tomography (HR-pQCT) is a 3-dimensional imaging modality with superior sensitivity for bone changes and abnormalities. Recent advances have led to increased use of HR-pQCT in inflammatory arthritis to report quantitative volumetric measures of bone density, microstructure, local anabolic (e.g., osteophytes, enthesiophytes) and catabolic (e.g., erosions) bone changes and joint space width. These features may be useful for monitoring disease progression, response to therapy, and are responsive to differentiating between those with inflammatory arthritis conditions and healthy controls. We reviewed 69 publications utilizing HR-pQCT imaging of the metacarpophalangeal (MCP) and/or wrist joints to investigate arthritis conditions. Erosions are a marker of early inflammatory arthritis progression, and recent work has focused on improvement and application of techniques to sensitively identify erosions, as well as quantifying erosion volume changes longitudinally using manual, semi-automated and automated methods. As a research tool, HR-pQCT may be used to detect treatment effects through changes in erosion volume in as little as 3 months. Studies with 1-year follow-up have demonstrated progression or repair of erosions depending on the treatment strategy applied. HR-pQCT presents several advantages. Combined with advances in image processing and image registration, individual changes can be monitored with high sensitivity and reliability. Thus, a major strength of HR-pQCT is its applicability in instances where subtle changes are anticipated, such as early erosive progression in the presence of subclinical inflammation. HR-pQCT imaging results could ultimately impact decision making to uptake aggressive treatment strategies and prevent progression of joint damage. There are several potential areas where HR-pQCT evaluation of inflammatory arthritis still requires development. As a highly sensitive imaging technique, one of the major challenges has been motion artifacts; motion compensation algorithms should be implemented for HR-pQCT. New research developments will improve the current disadvantages including, wider availability of scanners, the field of view, as well as the versatility for measuring tissues other than only bone. The challenge remains to disseminate these analysis approaches for broader clinical use and in research.
Collapse
Affiliation(s)
- Rasmus Klose-Jensen
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Justin J Tse
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Cheryl Barnabe
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Andrew J Burghardt
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Stephanie Finzel
- Department of Rheumatology and Clinical Immunology, Medical Centre - University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Ellen-Margrethe Hauge
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Kathryn S Stok
- Department of Biomedical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Sarah L Manske
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
38
|
Abstract
In 1973, IL-6 was identified as a soluble factor that is secreted by T cells and is important for antibody production by B cells. Since its discovery more than 40 years ago, the IL-6 pathway has emerged as a pivotal pathway involved in immune regulation in health and dysregulation in many diseases. Targeting of the IL-6 pathway has led to innovative therapeutic approaches for various rheumatic diseases, such as rheumatoid arthritis, juvenile idiopathic arthritis, adult-onset Still’s disease, giant cell arteritis and Takayasu arteritis, as well as other conditions such as Castleman disease and cytokine release syndrome. Targeting this pathway has also identified avenues for potential expansion into several other indications, such as uveitis, neuromyelitis optica and, most recently, COVID-19 pneumonia. To mark the tenth anniversary of anti-IL-6 receptor therapy worldwide, we discuss the history of research into IL-6 biology and the development of therapies that target IL-6 signalling, including the successes and challenges and with an emphasis on rheumatic diseases. In this Perspective article, the authors recount the earliest stages of translational research into IL-6 biology and the subsequent development of therapeutic IL-6 pathway inhibitors for the treatment of autoimmune rheumatic diseases and potentially numerous other indications.
Collapse
|
39
|
Ohsugi Y. The immunobiology of humanized Anti-IL6 receptor antibody: From basic research to breakthrough medicine. J Transl Autoimmun 2019; 3:100030. [PMID: 32743515 PMCID: PMC7388389 DOI: 10.1016/j.jtauto.2019.100030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 11/22/2019] [Accepted: 12/13/2019] [Indexed: 01/07/2023] Open
Abstract
The clinical use of monoclonal antibodies is well established in human medicine and has been amongst the most important contributions of basic science to clinical disease. One such antibody, the humanized anti-human IL-6 receptor antibody, is used to treat a variety of autoimmune diseases, particularly rheumatoid arthritis. It is extremely difficult and a laborious process to go from a concept at the research bench, to government approval. Such approval implies not only efficacy but, more importantly, an appropriate safety profile. In this review, the history of anti-human IL-6 receptor antibody is discussed in depth beginning with the author's experience during a sabbatical visit at the University of California at Davis in 1978. At that time, it was discovered that B cell activation was at least one critical factor in the development of autoimmunity. Approximately six years later, the cDNA encoding for IL-6 was cloned as BSF-2 (B cell stimulatory factor 2) to differentiate B cells to produce antibody. Soon after, it was suggested that this cytokine plays an important role in the development of autoimmune diseases. Based on this evidence, the journey began to search for an IL-6 inhibitor. Although there were numerous obstacles in finding lead compounds, ultimately, basic science developed the methodology for high throughput readouts that would inhibit the biologic function of IL-6. It was finally concluded that a mouse monoclonal antibody against IL-6 receptor would be optimal. In 1991, this antibody was humanized by using CDR-grafting technology in collaboration with the MRC (Medical Research Council). The drug was named tocilizumab and launched as an innovative anti-rheumatic drug in 2008 in Japan. Subsequently, the drug has been used throughout the world and has achieved enormous success in helping patients who suffer from inflammatory arthropathies. The lessons learned in the development of this antibody have application to the study of biologics and their application to other human diseases.
Collapse
Affiliation(s)
- Yoshiyuki Ohsugi
- Ohsugi BioPharma Consulting Co., Ltd., 5th Fl. Denbo Bldg., 1-39-11 Asakusa, Taito-ku, Tokyo, 111-0032, Japan
| |
Collapse
|
40
|
Gualtierotti R, Ingegnoli F, Boscolo M, Griffini S, Grovetti E, Cugno M. Tocilizumab Effects on Coagulation Factor XIII in Patients with Rheumatoid Arthritis. Adv Ther 2019; 36:3494-3502. [PMID: 31654331 PMCID: PMC6860466 DOI: 10.1007/s12325-019-01118-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Indexed: 12/19/2022]
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic systemic auto-immune disease associated with a prothrombotic state. Tocilizumab, an interleukin-6 receptor inhibitor, is highly effective in controlling disease activity and thrombotic risk. Factor XIII (FXIII), involved in thrombotic complications, has been reported to be reduced in RA patients during maintenance treatment with tocilizumab, but no data are available before and after the drug administration. Thus, we investigated the effects of tocilizumab on FXIII, thrombin generation and inflammation in patients with RA naïve for the drug. Methods We studied 15 consecutive adult patients with RA at baseline and 4 weeks after the onset of parenteral administration of tocilizumab, measuring disease activity and plasma levels of C-reactive protein (CRP), FXIII, and prothrombin fragments F1+2 by immunoenzymatic methods. Fifteen healthy subjects, sex-and age-matched with patients, served as normal controls for laboratory measurements. Results At baseline, patients with established RA had a median DAS28 of 4.8 (3.2–8.3) and, compared to healthy controls, had higher plasma levels of CRP (p < 0.0001), FXIII (p = 0.017) and F1+2 (p < 0.0001). Four weeks after starting treatment with tocilizumab, based on the EULAR response criteria, eight patients were classifiable as responders and seven as non-responders. In responders, we observed a statistically significant reduction not only of the values of DAS28 and CRP (p = 0.012 for both), ut also of plasma levels of FXIII (p = 0.05) and F1+2 (p = 0.025). In non-responders, all the studied parameters were unchanged. Conclusion The decrease of FXIII and F1+2 levels after tocilizumab treatment observed only in those patients who responded to the drug indicates that the effect of tocilizumab on the prothrombotic state is linked to the control of inflammation and disease activity and not to a direct effect of the drug, thus contributing to the reduction of the cardiovascular risk.
Collapse
|