1
|
Thudium CS, Rasmussen S, Karsdal MA, Bay-Jensen AC. Association between type III collagen degradation and local tissue damage of a single joint. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100527. [PMID: 39502930 PMCID: PMC11535998 DOI: 10.1016/j.ocarto.2024.100527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
Objective The development of disease-modifying drugs is limited by OA's heterogeneity and the challenge of defining clinical endpoints. Serological biomarkers are considered potential surrogate endpoints, but their contribution from single joints to systemic levels in OA patients is unclear. In this exploratory study we longitudinally assessed systemic biomarker levels' response to tissue damage and healing before and after surgery in patients undergoing knee or hip joint replacement revision for aseptic failure. Patients with chronic pain associated with a prior hip or knee arthroplasty, but not receiving revision surgery were included as control. Method The serological biomarker of MMP mediated type III collagen degradation C3M, associated with synovial tissue degradation, was measured at baseline before revision surgery, after revision surgery and at a 6-month follow-up in 48 patients with aseptic loosening of a knee or hip prosthesis and in 18 patients with chronic pain from a hip or knee prosthesis. Longitudinal changes in biomarkers were modeled using linear mixed models. Results No differences between the aseptic loosening and chronic pain groups were observed at baseline. Revision surgery in the aseptic loosening group led to a swift increase in C3M, which normalized within 2-3 months. No changes in biomarker level were observed in chronic pain patients over three months. Conclusion These findings suggest that tissue damage in a single joint significantly impacts systemic biomarker levels and underscores the relevance of systemic biomarkers in assessing local tissue remodeling.
Collapse
Affiliation(s)
| | - Sten Rasmussen
- Department of Clinical Medicine, Aalborg University, Denmark
| | | | | |
Collapse
|
2
|
Ruscitti P, Allanore Y, Baldini C, Barilaro G, Bartoloni Bocci E, Bearzi P, Bellis E, Berardicurti O, Biaggi A, Bombardieri M, Cantarini L, Cantatore FP, Caporali R, Caso F, Cervera R, Ciccia F, Cipriani P, Chatzis L, Colafrancesco S, Conti F, Corberi E, Costa L, Currado D, Cutolo M, D'Angelo S, Del Galdo F, Di Cola I, Di Donato S, Distler O, D'Onofrio B, Doria A, Fautrel B, Fasano S, Feist E, Fisher BA, Gabini M, Gandolfo S, Gatto M, Genovali I, Gerli R, Grembiale RD, Guggino G, Hoffmann-Vold AM, Iagnocco A, Iaquinta FS, Liakouli V, Manoussakis MN, Marino A, Mauro D, Montecucco C, Mosca M, Naty S, Navarini L, Occhialini D, Orefice V, Perosa F, Perricone C, Pilato A, Pitzalis C, Pontarini E, Prete M, Priori R, Rivellese F, Sarzi-Puttini P, Scarpa R, Sebastiani G, Selmi C, Shoenfeld Y, Triolo G, Trunfio F, Yan Q, Tzioufas AG, Giacomelli R. Tailoring the treatment of inflammatory rheumatic diseases by a better stratification and characterization of the clinical patient heterogeneity. Findings from a systematic literature review and experts' consensus. Autoimmun Rev 2024; 23:103581. [PMID: 39069240 DOI: 10.1016/j.autrev.2024.103581] [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: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Inflammatory rheumatic diseases are different pathologic conditions associated with a deregulated immune response, codified along a spectrum of disorders, with autoinflammatory and autoimmune diseases as two-end phenotypes of this continuum. Despite pathogenic differences, inflammatory rheumatic diseases are commonly managed with a limited number of immunosuppressive drugs, sometimes with partial evidence or transferring physicians' knowledge in different patients. In addition, several randomized clinical trials, enrolling these patients, did not meet the primary pre-established outcomes and these findings could be linked to the underlying molecular diversities along the spectrum of inflammatory rheumatic disorders. In fact, the resulting patient heterogeneity may be driven by differences in underlying molecular pathology also resulting in variable responses to immunosuppressive drugs. Thus, the identification of different clinical subsets may possibly overcome the major obstacles that limit the development more effective therapeutic strategies for these patients with inflammatory rheumatic diseases. This clinical heterogeneity could require a diverse therapeutic management to improve patient outcomes and increase the frequency of clinical remission. Therefore, the importance of better patient stratification and characterization is increasingly pointed out according to the precision medicine principles, also suggesting a new approach for disease treatment. In fact, based on a better proposed patient profiling, clinicians could more appropriately balance the therapeutic management. On these bases, we synthetized and discussed the available literature about the patient profiling in regard to therapy in the context of inflammatory rheumatic diseases, mainly focusing on randomized clinical trials. We provided an overview of the importance of a better stratification and characterization of the clinical heterogeneity of patients with inflammatory rheumatic diseases identifying this point as crucial in improving the management of these patients.
Collapse
Affiliation(s)
- Piero Ruscitti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Yannick Allanore
- Rheumatology Department, Cochin Hospital, APHP, INSERM U1016, Université Paris Cité, Paris, France
| | - Chiara Baldini
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giuseppe Barilaro
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Member of ERN-ReCONNET/RITA, Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Elena Bartoloni Bocci
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Pietro Bearzi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Elisa Bellis
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Onorina Berardicurti
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Alice Biaggi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Luca Cantarini
- Department of Medical Sciences, Surgery and Neurosciences, Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, University of Siena, Siena, Italy; Azienda Ospedaliero-Universitaria Senese [European Reference Network (ERN) for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) Center] Siena, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Roberto Caporali
- Department of Clinical Sciences and Community Health, University of Milan, Paediatric Rheumatology Unit, and Clinical Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Francesco Caso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Ricard Cervera
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases, Vasculitis and Autoinflammatory Diseases of the Catalan and Spanish Health Systems, Member of ERN-ReCONNET/RITA, Hospital Clínic, University of Barcelona, Barcelona, Catalonia, Spain
| | - Francesco Ciccia
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Loukas Chatzis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Serena Colafrancesco
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Fabrizio Conti
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Erika Corberi
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Luisa Costa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Damiano Currado
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Rheumatology, Department of Internal Medicine and Specialties, University of Genova Italy, IRCCS Polyclinic Hospital, Genova, Italy
| | - Salvatore D'Angelo
- Rheumatology Depatment of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Ilenia Di Cola
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Stefano Di Donato
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bernardo D'Onofrio
- Department of Internal Medicine and Therapeutics, Università di Pavia, Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Bruno Fautrel
- Sorbonne Université - Assistance Publique Hôpitaux de Paris, INSERM UMRS 1136, Hôpital de La Pitié Salpêtrière, Paris, France
| | - Serena Fasano
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Eugen Feist
- Department of Rheumatology, Helios Fachklinik, Sophie-von-Boetticher-Straße 1, 39245, Vogelsang-Gommern, Germany; Charité - Universitätsmedizin Berlin, Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Berlin, Germany
| | - Benjamin A Fisher
- Institute of Inflammation and Ageing, University Hospitals Birmingham, Birmingham, UK; Department of Rheumatology, National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Marco Gabini
- Rheumatology Unit, Santo Spirito Hospital, Pescara, Italy
| | - Saviana Gandolfo
- Unit of Rheumatology, San Giovanni Bosco Hospital, Naples, Italy
| | - Mariele Gatto
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Irene Genovali
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Roberto Gerli
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Rosa Daniela Grembiale
- Rheumatology Research Unit, Dipartimento di Scienze della Salute, Università degli studi "Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Giuliana Guggino
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Anna Maria Hoffmann-Vold
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Rheumatology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Dipartimento di Scienze Cliniche e Biologiche Università di Torino - AO Mauriziano di Torino, Turin, Italy
| | - Francesco Salvatore Iaquinta
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Vasiliki Liakouli
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Menelaos N Manoussakis
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Annalisa Marino
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Daniele Mauro
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Carlomaurizio Montecucco
- Department of Internal Medicine and Therapeutics, Università di Pavia, Division of Rheumatology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marta Mosca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Saverio Naty
- Department of Health Sciences, "Magna Græcia" University of Catanzaro, 88100 Catanzaro, Italy
| | - Luca Navarini
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Daniele Occhialini
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Valeria Orefice
- Rheumatology Unit, San Camillo-Forlanini Hospital, Rome, Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Carlo Perricone
- Section of Rheumatology, Department of Medicine and Surgery, University of Perugia, Italy
| | - Andrea Pilato
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Elena Pontarini
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Marcella Prete
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine (DIM), University of Bari Medical School, Italy
| | - Roberta Priori
- Department of Internal Medicine and Medical Specialties, Rheumatology Unit, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC & NHS Trust & National Institute for Health and Care Research (NIHR) Barts Biomedical Research Centre (BRC), London, UK
| | - Piercarlo Sarzi-Puttini
- Rheumatology Department, ASST Fatebenefratelli Luigi Sacco University Hospital, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Raffaele Scarpa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Yehuda Shoenfeld
- Zabludovwicz autoimmunity center, Sheba medical center, Tel Hashomer Israel, Reichman University, Herzeliya, Israel
| | - Giovanni Triolo
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Italy
| | - Francesca Trunfio
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Qingran Yan
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Athanasios G Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Roberto Giacomelli
- Rheumatology and Clinical Immunology, Department of Medicine, School of Medicine, University of Rome "Campus Bio-Medico", 00128 Rome, Italy; Clinical and Research Section of Rheumatology and Clinical Immunology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| |
Collapse
|
3
|
Madsen SF, Sinkeviciute D, Thudium CS, Karsdal MA, Bay-Jensen AC. The fibroid phenotype of biological naïve patients with rheumatoid arthritis are less likely to respond to anti-IL-6R treatment. Sci Rep 2024; 14:10751. [PMID: 38730088 PMCID: PMC11087519 DOI: 10.1038/s41598-024-61435-2] [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: 12/15/2023] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
Type III collagen gene expression is upregulated in the synovium of patients with rheumatoid arthritis (RA) presenting the fibroid phenotype. The soluble type III collagen formation biomarker, PRO-C3, is known to measure fibrogenesis in fibrotic diseases. In this exploratory study, we aimed to investigate the association between fibrogenesis (PRO-C3) and the disease- and treatment response in patients with RA. We measured PRO-C3 in subsets of two clinical trials assessing the effect of the anti-interleukin-6 (IL-6) receptor treatment tocilizumab (TCZ) as monotherapy or polytherapy with methotrexate. PRO-C3 levels had weak or very weak correlations with the clinical parameters (Spearman's). However, when the patients were divided into Disease Activity Score-28 groups characterized by the erythrocyte sedimentation rate (DAS28-ESR), there was a statistical difference between the PRO-C3 levels of the different groups (p < 0.05). To determine the response in relation to PRO-C3, a cut-off based on PRO-C3 levels and patients in remission (DAS28-ESR ≤ 2.6) was identified. This showed that a reduction in PRO-C3 after treatment initiation was associated with decreased DAS28-ESR and a higher response rate in patients with low PRO-C3 levels than in those with high PRO-C3 levels. This indicates that a fibrotic component affects the responsiveness of patients.
Collapse
Affiliation(s)
- Sofie Falkenløve Madsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 205, 2730, Herlev, Denmark.
| | - Dovile Sinkeviciute
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 205, 2730, Herlev, Denmark
| | - Christian S Thudium
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 205, 2730, Herlev, Denmark
| | - Morten A Karsdal
- ImmunoScience, Nordic Bioscience, Herlev Hovedgade 205, 2730, Herlev, Denmark
| | | |
Collapse
|
4
|
Yu SP, Deveza LA, Kraus VB, Karsdal M, Bay-Jensen AC, Collins JE, Guermazi A, Roemer FW, Ladel C, Bhagavath V, Hunter DJ. Association of biochemical markers with bone marrow lesion changes on imaging-data from the Foundation for the National Institutes of Health Osteoarthritis Biomarkers Consortium. Arthritis Res Ther 2024; 26:30. [PMID: 38238803 PMCID: PMC10795356 DOI: 10.1186/s13075-023-03253-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 12/27/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND To assess the prognostic value of short-term change in biochemical markers as it relates to bone marrow lesions (BMLs) on MRI in knee osteoarthritis (OA) over 24 months and, furthermore, to assess the relationship between biochemical markers involved with tissue turnover and inflammation and BMLs on MRI. METHODS Data from the Foundation for the National Institutes of Health OA Biomarkers Consortium within the Osteoarthritis Initiative (n = 600) was analyzed. BMLs were measured according to the MRI Osteoarthritis Knee Score (MOAKS) system (0-3), in 15 knee subregions. Serum and urinary biochemical markers assessed were as follows: serum C-terminal crosslinked telopeptide of type I collagen (CTX-I), serum crosslinked N-telopeptide of type I collagen (NTX-I), urinary CTX-Iα and CTX-Iβ, urinary NTX-I, urinary C-terminal cross-linked telopeptide of type II collagen (CTX-II), serum matrix metalloproteinase (MMP)-degraded type I, II, and III collagen (C1M, C2M, C3M), serum high sensitivity propeptide of type IIb collagen (hsPRO-C2), and matrix metalloproteinase-generated neoepitope of C-reactive protein (CRPM). The association between change in biochemical markers over 12 months and BMLs over 24 months was examined using regression models adjusted for covariates. The relationship between C1M, C2M, C3M, hsPRO-C2, and CRPM and BMLs at baseline and over 24 months was examined. RESULTS Increases in serum CTX-I and urinary CTX-Iβ over 12 months were associated with increased odds of changes in the number of subregions affected by any BML at 24 months. Increase in hsPRO-C2 was associated with decreased odds of worsening in the number of subregions affected by any BML over 24 months. C1M and C3M were associated with BMLs affected at baseline. CONCLUSIONS Short-term changes in serum CTX-I, hsPRO-C2, and urinary CTX-Iβ hold the potential to be prognostic of BML progression on MRI. The association of C1M and C3M with baseline BMLs on MRI warrants further investigation.
Collapse
Affiliation(s)
- Shirley P Yu
- Department of Rheumatology, Royal North Shore Hospital, Reserve Road, St Leonards, Sydney, NSW, 2065, Australia.
- Sydney Musculoskeletal Health, The Kolling Institute, School of Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
| | - Leticia A Deveza
- Department of Rheumatology, Royal North Shore Hospital, Reserve Road, St Leonards, Sydney, NSW, 2065, Australia
- Sydney Musculoskeletal Health, The Kolling Institute, School of Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Virginia B Kraus
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | | | | | - Jamie E Collins
- Orthopaedic and Arthritis Centre for Outcomes Research, Brigham and Women's Hospital, Boston, MA, USA
| | - Ali Guermazi
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Frank W Roemer
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
- Department of Radiology, Universitätsklinikum Erlangen & Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | | | - Venkatesha Bhagavath
- Sydney Musculoskeletal Health, The Kolling Institute, School of Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Northern Sydney Local Health District, Royal North Shore Hospital, St Leonards, Sydney, NSW, Australia
| | - David J Hunter
- Department of Rheumatology, Royal North Shore Hospital, Reserve Road, St Leonards, Sydney, NSW, 2065, Australia
- Sydney Musculoskeletal Health, The Kolling Institute, School of Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
5
|
Thudium CS, Frederiksen P, Karsdal MA, Bay-Jensen AC. Changes in type VI collagen degradation reflect clinical response to treatment in rheumatoid arthritis patients treated with tocilizumab. Arthritis Res Ther 2024; 26:3. [PMID: 38167226 PMCID: PMC10759322 DOI: 10.1186/s13075-023-03242-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/16/2023] [Indexed: 01/05/2024] Open
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by inflammation in multiple articular joints, causing pain, joint damage, and loss of joint function. Despite the successful development of disease-modifying therapies, the heterogeneity of RA means that a significant proportion of patients respond poorly to treatment. This highlights the need for personalized medicine and predictive biomarkers to optimize treatment efficacy, safety, and cost. This study aimed to explore the relationship between type VI collagen (Col VI) remodeling and clinical response to anti-IL-6 receptor treatment. METHODS Type VI collagen degradation was quantified using the C6M biomarker, a fragment of type VI collagen degraded by MMPs. Longitudinal differences in average biomarker levels between placebo and treatment groups were estimated using linear mixed models. The predictive capacity of the marker based on change from baseline to 4 weeks was analyzed using logistic regression. RESULTS Both 4 mg and 8 mg doses of Tocilizumab (TCZ) reduced serum C6M concentrations compared to the placebo. Furthermore, C6M levels were more reduced in patients responding to treatment compared to early non-responders. A lower early reduction in C6M was associated with reduced odds of ACR treatment response and lowered disease activity. CONCLUSION These findings suggest that quantifying type VI collagen turnover may aid in identifying patients less likely to respond to treatment, indicating a new path towards optimizing patient care. Further studies are needed to validate these findings and explore the underlying mechanisms driving the observed relationships.
Collapse
Affiliation(s)
| | | | - Morten A Karsdal
- Nordic Bioscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | | |
Collapse
|
6
|
Sheng XR, Gao X, Schiffman C, Jiang J, Ramalingam TR, Lin CJF, Khanna D, Neighbors M. Biomarkers of fibrosis, inflammation, and extracellular matrix in the phase 3 trial of tocilizumab in systemic sclerosis. Clin Immunol 2023; 254:109695. [PMID: 37479123 DOI: 10.1016/j.clim.2023.109695] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/16/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Drug development for systemic sclerosis (SSc) benefits from understanding the relationship between disease and circulating biomarkers to enable activities such as patient stratification and evaluation of therapeutic response. We measured biomarkers in serum from SSc patients from a phase 3 trial of tocilizumab (focuSSced) and compared baseline levels with healthy controls (HCs). Several baseline biomarkers appeared elevated in SSc patients compared to HCs, suggesting activation of epithelial damage, inflammation, fibrosis, and extracellular matrix (ECM) remodeling. Baseline correlations among both periostin/COMP and ECM biomarker subsets implicated their participation in fibroblast activation. Tocilizumab treatment modulated serum biomarkers of macrophage activation, inflammation, and ECM turnover, including collagen formation and degradation neoepitopes. Baseline CRP, periostin, and SP-D showed prognostic trends for worsening lung function, and IL-6, COMP, periostin, and Pro-C3 showed prognostic trends for worsening skin thickness. These prognostic results warrant confirmation in additional patient cohorts to verify their utility.
Collapse
Affiliation(s)
- X Rebecca Sheng
- Genentech Inc., South San Francisco, CA, United States of America.
| | - Xia Gao
- Genentech Inc., South San Francisco, CA, United States of America
| | | | - Jenny Jiang
- Genentech Inc., South San Francisco, CA, United States of America
| | | | - Celia J F Lin
- Genentech Inc., South San Francisco, CA, United States of America
| | - Dinesh Khanna
- University of Michigan Scleroderma Program, Division of Rheumatology/Department of Internal Medicine, Ann Arbor, MI, United States of America
| | | |
Collapse
|
7
|
Chen SF, Yeh FC, Chen CY, Chang HY. Tailored therapeutic decision of rheumatoid arthritis using proteomic strategies: how to start and when to stop? Clin Proteomics 2023; 20:22. [PMID: 37301840 DOI: 10.1186/s12014-023-09411-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Unpredictable treatment responses have been an obstacle for the successful management of rheumatoid arthritis. Although numerous serum proteins have been proposed, there is a lack of integrative survey to compare their relevance in predicting treatment outcomes in rheumatoid arthritis. Also, little is known about their applications in various treatment stages, such as dose modification, drug switching or withdrawal. Here we present an in-depth exploration of the potential usefulness of serum proteins in clinical decision-making and unveil the spectrum of immunopathology underlying responders to different drugs. Patients with robust autoimmunity and inflammation are more responsive to biological treatments and prone to relapse during treatment de-escalation. Moreover, the concentration changes of serum proteins at the beginning of the treatments possibly assist early recognition of treatment responders. With a better understanding of the relationship between the serum proteome and treatment responses, personalized medicine in rheumatoid arthritis will be more achievable in the near future.
Collapse
Affiliation(s)
- Shuo-Fu Chen
- Department of Heavy Particles & Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Fu-Chiang Yeh
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Yun Chen
- Department of Biomedical Sciences and Engineering, Institute of Biomedical Engineering and Nanomedicine, National Central University, Taoyuan, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Hui-Yin Chang
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, No. 300, Zhongda Rd., Zhongli District, Taoyuan, 320317, Taiwan.
| |
Collapse
|
8
|
Kraus VB, Karsdal MA. Clinical monitoring in osteoarthritis: Biomarkers. Osteoarthritis Cartilage 2022; 30:1159-1173. [PMID: 34536529 PMCID: PMC8924021 DOI: 10.1016/j.joca.2021.04.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/23/2021] [Accepted: 04/07/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The purpose of this overview of osteoarthritis (OA) biomarkers is to provide the non-specialist with a toolbox, based on experience acquired by biomarker researchers over many years, to understand biomarkers in general and their use in the OA field. METHODS We provide an update on this subject since the OARSI Primer on osteoarthritis (OA) nearly a decade ago. RESULTS Since the last update, the importance of molecular biomarkers has been increasingly recognized in the field, but no OA-related biomarkers have been adopted for routine use in clinical practice. The current lack of chondroprotective treatments for OA impairs the assessment, validation and qualification of the potential role of biomarkers as tools for monitoring disease status and patient responses to treatment of OA. Yet there is no lack of an evolving compendium of OA-related biomarkers, ever more fit-for-purpose, that could currently facilitate drug development for OA. We provide an abbreviated update and overview of specific soluble OA-related biomarkers for this new OARSI Primer on OA with OA-relevant examples encompassing the concepts of biomarker nomenclature, qualification, interpretation, measurement, reporting requirements, application to research, drug discovery and clinical care, and future needs for biomarker advancement. CONCLUSION Appropriate biomarkers should play a role at all stages of OA diagnosis, prognosis, drug development, and treatment. The future of OA biomarker research and development holds great promise as its foundation is increasingly robust.
Collapse
Affiliation(s)
- V B Kraus
- Duke Molecular Physiology Institute and Division of Rheumatology, Duke University School of Medicine, Durham, NC, USA.
| | - M A Karsdal
- Rheumatology, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark
| |
Collapse
|
9
|
van de Stadt LA, Kroon FPB, Thudium CF, Bay-Jensen AC, Kloppenburg M. Serum biomarkers in prednisolone-treated hand osteoarthritis patients. Rheumatology (Oxford) 2022; 62:1350-1355. [PMID: 35946535 PMCID: PMC9977113 DOI: 10.1093/rheumatology/keac442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To investigate whether biomarkers are modulated by prednisolone treatment in patients with hand OA and whether they can predict response to prednisolone. METHODS Biomarkers reflecting tissue turnover and inflammation [aggrecanase-derived neoepitope of arggecan (ARGS), MMP-derived neoepitope of type I collagen (C1M), MMP-derived neoepitope of type III collagen (C3M), marker of true type V collagen formation (PROC5), MMP-derived neoepitope of CRP (CRPM), citrullinated vimentin fragment (VICM), high-sensitivity (hsCRP)] were measured in sera from 78 patients with painful inflammatory hand OA, who were randomized between prednisolone or placebo treatment. Association of baseline biomarker levels with disease characteristics [visual analogue scale (VAS) pain, synovial thickening ultrasonography sum score and erosive OA] and OMERACT-Osteoarthritis Research Society International (OARSI) response after 6 weeks were analysed with linear or logistic regression and adjusted for age, BMI and sex. Change in biomarker levels after 6 weeks was assessed with linear regression adjusted for baseline biomarker levels, age, BMI and sex. RESULTS For all patients (mean age 64 years, 79% female), there were no associations between biomarker levels and VAS finger pain or synovial thickening score at baseline. Patients with erosive hand OA had higher levels of C1M and hsCRP [adjusted geometric mean ratio 1.24 (95% CI 1.03, 1.49) and 1.91 (1.19, 3.06), respectively]. Biomarker levels did not decrease over time. There was no association between baseline biomarkers levels and OARSI response, except for CRPM [geometric mean ratio of 0.88 (0.77, 1.00)]. CONCLUSION Erosive disease was associated with higher levels of C1M and hsCRP. Biomarker levels were not influenced by treatment with prednisolone. Current biomarkers were not associated with response to prednisolone in hand OA.
Collapse
Affiliation(s)
- Lotte A van de Stadt
- Correspondence to: Lotte A. van de Stadt, Department Rheumatology, Leiden University Medical Center, postal zone C1-R, Albinusdreef 2, 2333 ZA Leiden, The Netherlands. E-mail:
| | - Féline P B Kroon
- Department of Rheumatology, Leiden University Medical Center (LUMC), Leiden,Department of Rheumatology, Zuyderland Medical Center, Heerlen, The Netherlands
| | | | | | - Margreet Kloppenburg
- Department of Rheumatology, Leiden University Medical Center (LUMC), Leiden,Department of Clinical Epidemiology, LUMC, Leiden, The Netherlands
| |
Collapse
|
10
|
Tsuchiya H, Fujio K. Title Current Status of the Search for Biomarkers for Optimal Therapeutic Drug Selection for Patients with Rheumatoid Arthritis. Int J Mol Sci 2021; 22:ijms22179534. [PMID: 34502442 PMCID: PMC8431405 DOI: 10.3390/ijms22179534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/28/2021] [Accepted: 08/28/2021] [Indexed: 12/19/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by destructive synovitis. It is significantly associated with disability, impaired quality of life, and premature mortality. Recently, the development of biological agents (including tumor necrosis factor-α and interleukin-6 receptor inhibitors) and Janus kinase inhibitors have advanced the treatment of RA; however, it is still difficult to predict which drug will be effective for each patient. To break away from the current therapeutic approaches that could be described as a “lottery,” there is an urgent need to establish biomarkers that stratify patients in terms of expected therapeutic responsiveness. This review deals with recent progress from multi-faceted analyses of the synovial tissue in RA, which is now bringing new insights into diverse features at both the cellular and molecular levels and their potential links with particular clinical phenotypes.
Collapse
|
11
|
Bay-Jensen AC, Siebuhr AS, Damgaard D, Drobinski P, Thudium C, Mortensen J, Nielsen CH. Objective and noninvasive biochemical markers in rheumatoid arthritis: where are we and where are we going? Expert Rev Proteomics 2021; 18:159-175. [PMID: 33783300 DOI: 10.1080/14789450.2021.1908892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects approximately 1% of the adult population. RA is multi-factorial, and as such our understanding of the molecular pathways involved in the disease is currently limited. An increasing number of studies have suggested that several molecular phenotypes (i.e. endotypes) of RA exist, and that different endotypes respond differently to various treatments. Biochemical markers may be an attractive means for achieving precision medicine, as they are objective and easily obtainable. AREAS COVERED We searched recent publications on biochemical markers in RA as either diagnostic or prognostic markers, or as markers of disease activity. Here, we provide a narrative overview of different classes of markers, such as autoantibodies, citrulline products, markers of tissue turnover and cytokines, that have been tested in clinical cohorts or trials including RA patients. EXPERT OPINION Although many biochemical markers have been identified and tested, few are currently being used in clinical practice. As more treatment options are becoming available, the need for precision medicine tools that can aid physicians and patients in choosing the right treatment is growing.
Collapse
Affiliation(s)
- Anne C Bay-Jensen
- ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Anne Sofie Siebuhr
- ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Dres Damgaard
- Center for Rheumatolology and Spine Diseases, Institute for Inflammation Research, University of Copenhagen, Copenhagen Ø, Denmark
| | - Patryk Drobinski
- ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Christian Thudium
- ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Joachim Mortensen
- ImmunoScience, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Claus H Nielsen
- Center for Rheumatolology and Spine Diseases, Institute for Inflammation Research, University of Copenhagen, Copenhagen Ø, Denmark
| |
Collapse
|
12
|
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
|
13
|
The growing role of precision medicine for the treatment of autoimmune diseases; results of a systematic review of literature and Experts' Consensus. Autoimmun Rev 2020; 20:102738. [PMID: 33326854 DOI: 10.1016/j.autrev.2020.102738] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Autoimmune diseases (AIDs) share similar serological, clinical, and radiological findings, but, behind these common features, there are different pathogenic mechanisms, immune cells dysfunctions, and targeted organs. In this context, multiple lines of evidence suggest the application of precision medicine principles to AIDs to reduce the treatment failure. Precision medicine refers to the tailoring of therapeutic strategies to the individual characteristics of each patient, thus it could be a new approach for management of AIDS which considers individual variability in genes, environmental exposure, and lifestyle. Precision medicine would also assist physicians in choosing the right treatment, the best timing of administration, consequently trying to maximize drug efficacy, and, possibly, reducing adverse events. In this work, the growing body of evidence is summarized regarding the predictive factors for drug response in patients with AIDs, applying the precision medicine principles to provide high-quality evidence for therapeutic opportunities in improving the management of these patients.
Collapse
|
14
|
Carvajal Alegria G, Garrigues F, Bettacchioli E, Loeuille D, Saraux A, Cornec D, Devauchelle-Pensec V, Renaudineau Y. Tocilizumab controls bone turnover in early polymyalgia rheumatica. Joint Bone Spine 2020; 88:105117. [PMID: 33301930 DOI: 10.1016/j.jbspin.2020.105117] [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] [Received: 06/19/2020] [Accepted: 11/17/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVES This study explores changes in the bone homeostasis by testing the N-terminal collagen type I extension propeptide (PINP) marker for osteo-formation and the carboxy-terminal region of collagen type I (CTX-I) marker for osteo-resorption in patients taking tocilizumab for polymyalgia rheumatica (PMR). METHODS Twenty patients were included in the prospective open-label TENOR study (Clinicaltrials.gov NCT01713842) and received three monthly tocilizumab infusions, followed by corticosteroids starting at week (W) 12. PINP and CTX-I were tested at inclusion (W0), after tocilizumab but before steroid initiation (W12), at the end of the protocol (W24) and were compared to healthy controls. Information regarding disease activity, bone mineral density using scanographic bone attenuation correlation (SBAC), inflammatory parameters and interleukin (IL)-6 levels were collected during the follow-up of the patients. RESULTS PMR patients were characterised by a reduction in bone mineral density and a higher level of CTX-I relative to healthy controls matched in age and sex at baseline. PINP levels increased at W12 (P< 0.001, versus W0) following tocilizumab introduction and CTX-I levels decreased at W24 and after steroid initiation (P=0.001, versus W0). Such modifications explain the altered correlation observed between PINP and CTX-I at W0 (r=0.255 at W0 versus r=0.641 in healthy controls) and its correction after treatment (r=0.760 at W12 and r=0.767 at W24). Finally, greater changes in PINP were observed in patients whose circulating IL-6 levels decreased after tocilizumab therapy. CONCLUSIONS Control of bone turnover, in part through the inhibition of the IL-6 axis, is observed during tocilizumab and subsequent steroid treatment of PMR.
Collapse
Affiliation(s)
- Guillermo Carvajal Alegria
- Rheumatology department, CHRU Cavale Blanche, Brest, France; Lymphocytes B et autoimmunité, UMR1227, INSERM, Université de Bretagne Occidentale, Brest, France.
| | | | | | - Damien Loeuille
- Department of Rheumatology, University Hospital of Nancy, 54500 Vandoeuvre-lès-Nancy, France; INSERM, CIC-EC CIE6, Nancy, France University Hospital of Nancy, Epidemiology and Clinical Evaluation, 545 Vandoeuvre-lès-Nancy, France
| | - Alain Saraux
- Rheumatology department, CHRU Cavale Blanche, Brest, France
| | - Divi Cornec
- Rheumatology department, CHRU Cavale Blanche, Brest, France; Lymphocytes B et autoimmunité, UMR1227, INSERM, Université de Bretagne Occidentale, Brest, France
| | - Valérie Devauchelle-Pensec
- Rheumatology department, CHRU Cavale Blanche, Brest, France; Lymphocytes B et autoimmunité, UMR1227, INSERM, Université de Bretagne Occidentale, Brest, France
| | - Yves Renaudineau
- Laboratory of immunology and immunotherapy, UMR1227, CHRU Morvan, Brest, France
| |
Collapse
|
15
|
Bone phenotypes in rheumatology - there is more to bone than just bone. BMC Musculoskelet Disord 2020; 21:789. [PMID: 33248451 PMCID: PMC7700716 DOI: 10.1186/s12891-020-03804-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis, rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis, all have one clear common denominator; an altered turnover of bone. However, this may be more complex than a simple change in bone matrix and mineral turnover. While these diseases share a common tissue axis, their manifestations in the area of pathology are highly diverse, ranging from sclerosis to erosion of bone in different regions. The management of these diseases will benefit from a deeper understanding of the local versus systemic effects, the relation to the equilibrium of the bone balance (i.e., bone formation versus bone resorption), and the physiological and pathophysiological phenotypes of the cells involved (e.g., osteoblasts, osteoclasts, osteocytes and chondrocytes). For example, the process of endochondral bone formation in chondrocytes occurs exists during skeletal development and healthy conditions, but also in pathological conditions. This review focuses on the complex molecular and cellular taxonomy of bone in the context of rheumatological diseases that alter bone matrix composition and maintenance, giving rise to different bone turnover phenotypes, and how biomarkers (biochemical markers) can be applied to potentially describe specific bone phenotypic tissue profiles.
Collapse
|
16
|
Shatunova EA, Korolev MA, Omelchenko VO, Kurochkina YD, Davydova AS, Venyaminova AG, Vorobyeva MA. Aptamers for Proteins Associated with Rheumatic Diseases: Progress, Challenges, and Prospects of Diagnostic and Therapeutic Applications. Biomedicines 2020; 8:biomedicines8110527. [PMID: 33266394 PMCID: PMC7700471 DOI: 10.3390/biomedicines8110527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid aptamers capable of affine and specific binding to their molecular targets have now established themselves as a very promising alternative to monoclonal antibodies for diagnostic and therapeutic applications. Although the main focus in aptamers’ research and development for biomedicine is made on cardiovascular, infectious, and malignant diseases, the use of aptamers as therapeutic or diagnostic tools in the context of rheumatic diseases is no less important. In this review, we consider the main features of aptamers that make them valuable molecular tools for rheumatologists, and summarize the studies on the selection and application of aptamers for protein biomarkers associated with rheumatic diseases. We discuss the progress in the development of aptamer-based diagnostic assays and targeted therapeutics for rheumatic disorders, future prospects in the field, and issues that have yet to be addressed.
Collapse
Affiliation(s)
- Elizaveta A. Shatunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.S.); (A.S.D.); (A.G.V.)
| | - Maksim A. Korolev
- Research Institute of Clinical and Experimental Lymphology, Affiliated Branch of Federal Research Center of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, 630060 Novosibirsk, Russia; (M.A.K.); (V.O.O.); (Y.D.K.)
| | - Vitaly O. Omelchenko
- Research Institute of Clinical and Experimental Lymphology, Affiliated Branch of Federal Research Center of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, 630060 Novosibirsk, Russia; (M.A.K.); (V.O.O.); (Y.D.K.)
| | - Yuliya D. Kurochkina
- Research Institute of Clinical and Experimental Lymphology, Affiliated Branch of Federal Research Center of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, 630060 Novosibirsk, Russia; (M.A.K.); (V.O.O.); (Y.D.K.)
| | - Anna S. Davydova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.S.); (A.S.D.); (A.G.V.)
| | - Alya G. Venyaminova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.S.); (A.S.D.); (A.G.V.)
| | - Mariya A. Vorobyeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.S.); (A.S.D.); (A.G.V.)
- Correspondence:
| |
Collapse
|
17
|
Karsdal MA, Kraus VB, Shevell D, Bay-Jensen AC, Schattenberg J, Rambabu Surabattula R, Schuppan D. Profiling and targeting connective tissue remodeling in autoimmunity - A novel paradigm for diagnosing and treating chronic diseases. Autoimmun Rev 2020; 20:102706. [PMID: 33188918 DOI: 10.1016/j.autrev.2020.102706] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 08/16/2020] [Indexed: 12/14/2022]
Abstract
Connective tissue (ConT) remodeling is an essential process in tissue regeneration, where a balanced replacement of old tissue by new tissue occurs. This balance is disturbed in chronic diseases, often autoimmune diseases, usually resulting in the buld up of fibrosis and a gradual loss of organ function. During progression of liver, lung, skin, heart, joint, skeletal and kidney diseasesboth ConT formation and degradation are elevated, which is tightly linked to immune cell activation and a loss of specific cell types and extracellular matrix (ECM) structures that are required for normal organ function. Here, we address the balance of key general and organ specific components of the ECM during homeostasis and in disease, with a focus on collagens, which are emerging as both structural and signaling molecules harbouring neoepitopes and autoantigens that are released during ConT remodeling. Specific collagen molecular signatures of ConT remodeling are linked to disease activity and stage, and to prognosis across different organs. These signatures accompany and further drive disease progression, and often become detectable before clinical disease manifestation (illness). Recent advances allow to quantify and define the nature of ConT remodeling via blood-based assays that measure the levels of well-defined collagen fragments, reflecting different facets of ConT formation and degradation, and associated immunological processes. These novel serum assays are becoming important tools of precision medicine, to detect various chronic and autoimmune diseases before their clinical manifestation, and to non-invasively monitor the efficacy of a broad range of pharmacological interventions.
Collapse
Affiliation(s)
- Morten Asser Karsdal
- Nordic Bioscience, Biomarkers & Research A/S, Herlev, Metabolic Liver Research Program, Denmark
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Diane Shevell
- Clinical Biomarkers and Immunology, Bristol-Myers Squibb, Westfield, NJ, USA
| | | | | | - R Rambabu Surabattula
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Thudium CS, Bay-Jensen AC, Cahya S, Dow ER, Karsdal MA, Koch AE, Zhang W, Benschop RJ. The Janus kinase 1/2 inhibitor baricitinib reduces biomarkers of joint destruction in moderate to severe rheumatoid arthritis. Arthritis Res Ther 2020; 22:235. [PMID: 33046136 PMCID: PMC7552555 DOI: 10.1186/s13075-020-02340-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/02/2020] [Indexed: 12/26/2022] Open
Abstract
Background Tissue released blood-based biomarkers can provide insight into drug mode of action and response. To understand the changes in extracellular matrix turnover, we analyzed biomarkers associated with joint tissue turnover from a phase 3, randomized, placebo-controlled study of baricitinib in patients with active rheumatoid arthritis (RA). Methods Serum biomarkers associated with synovial inflammation (C1M, C3M, and C4M), cartilage degradation (C2M), bone resorption (CTX-I), and bone formation (osteocalcin) were analyzed at baseline, and weeks 4 and 12, from a subgroup of patients (n = 240) randomized to placebo or 2-mg or 4-mg baricitinib (RA-BUILD, NCT01721057). Mixed-model repeated measure was used to identify biomarkers altered by baricitinib. The relationship between changes in biomarkers and clinical measures was evaluated using correlation analysis. Results Treatment arms were well balanced for baseline biomarkers, demographics, and disease activity. At week 4, baricitinib 4-mg significantly reduced C1M from baseline by 21% compared to placebo (p < 0.01); suppression was sustained at week 12 (27%, p < 0.001). Baricitinib 4-mg reduced C3M and C4M at week 4 by 14% and 12% compared to placebo, respectively (p < 0.001); they remained reduced by 16% and 11% at week 12 (p < 0.001). In a pooled analysis including all treatment arms, patients with the largest reduction (upper 25% quartile) in C1M, C3M, and C4M by week 12 had significantly greater clinical improvement in the Simplified Disease Activity Index at week 12 compared to patients with the smallest reduction (lowest 25% quartile). Conclusion Baricitinib treatment resulted in reduced circulating biomarkers associated with joint tissue destruction as well as concomitant RA clinical improvement. Trial registration ClinicalTrials.gov NCT01721057; date of registration: November 1, 2012
Collapse
Affiliation(s)
- Christian S Thudium
- Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark.
| | - Anne C Bay-Jensen
- Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
| | | | - Ernst R Dow
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Morten A Karsdal
- Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
| | - Alisa E Koch
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
19
|
Blair JPM, Bay-Jensen AC, Tang MH, Frederiksen P, Bager C, Karsdal M, Brunak S. Identification of heterogenous treatment response trajectories to anti-IL6 receptor treatment in rheumatoid arthritis. Sci Rep 2020; 10:13975. [PMID: 32811969 PMCID: PMC7434906 DOI: 10.1038/s41598-020-70942-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/03/2020] [Indexed: 12/24/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease with fluctuating course of progression. Despite substantial improvement in treatments in recent years, treatment response is still not guaranteed. The aim of this study was to identify variation in Disease Activity Score 28 (DAS28) of RA patients in response to Tocilizumab, and to investigate both molecular and clinical factors influencing response. Clinical and biochemical data for 485 RA patients receiving Tocilizumab in combination with methotrexate were extracted from the LITHE phase III clinical study (NCT00106535), and post-hoc analysis conducted. Latent class mixed models were used to identify statistically distinct trajectories of DAS28 after the initiation of treatment. Biomarker measurements were then analysed cross-sectionally and temporally, to characterise patients by serological biomarkers and clinical factors. We identified three distinct trajectories of drug response: class 1 (n = 85, 17.5%), class 2 (n = 338, 69.7%) and class 3 (n = 62, 12.8%). All groups started with high DAS28 on average (DAS28 > 5.1). Class 1 showed the least reduction in DAS28, with significantly more patients seeking escape therapy (p < 0.001). Class 3 showed significantly higher rates of improvement in DAS28, with 58.1% achieving ACR response levels compared to 2.4% in class 1 (p < 0.0001). Biomarkers of inflammation, MMP-3, CRP, C1M, showed greater reduction in class 3 compared to the other classes. Identification of more homogenous patient sub-populations of drug response may allow for more targeted therapeutic treatment regimens and a better understanding of disease aetiology.
Collapse
Affiliation(s)
- J P M Blair
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark. .,ProScion, Herlev Hovedgade 205-207, 2730, Herlev, Denmark.
| | - A-C Bay-Jensen
- ImmunoScience, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - M H Tang
- ProScion, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - P Frederiksen
- ImmunoScience, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - C Bager
- ProScion, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - M Karsdal
- ImmunoScience, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - S Brunak
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
Blair JPM, Bager C, Platt A, Karsdal M, Bay-Jensen AC. Identification of pathological RA endotypes using blood-based biomarkers reflecting tissue metabolism. A retrospective and explorative analysis of two phase III RA studies. PLoS One 2019; 14:e0219980. [PMID: 31339920 PMCID: PMC6655687 DOI: 10.1371/journal.pone.0219980] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/05/2019] [Indexed: 12/31/2022] Open
Abstract
There is an increasing demand for accurate endotyping of patients according to their pathogenesis to allow more targeted treatment. We explore a combination of blood-based joint tissue metabolites (neoepitopes) to enable patient clustering through distinct disease profiles. We analysed data from two RA studies (LITHE (N = 574, follow-up 24 and 52 weeks), OSKIRA-1 (N = 131, follow-up 24 weeks)). Two osteoarthritis (OA) studies (SMC01 (N = 447), SMC02 (N = 81)) were included as non-RA comparators. Specific tissue-derived neoepitopes measured at baseline, included: C2M (cartilage degradation); CTX-I and PINP (bone turnover); C1M and C3M (interstitial matrix degradation); CRPM (CRP metabolite) and VICM (macrophage activity). Clustering was performed to identify putative endotypes. We identified five clusters (A-E). Clusters A and B were characterized by generally higher levels of biomarkers than other clusters, except VICM which was significantly higher in cluster B than in cluster A (p<0.001). Biomarker levels in Cluster C were all close to the median, whilst Cluster D was characterised by low levels of all biomarkers. Cluster E also had low levels of most biomarkers, but with significantly higher levels of CTX-I compared to cluster D. There was a significant difference in ΔSHP score observed at 52 weeks (p<0.05). We describe putative RA endotypes based on biomarkers reflecting joint tissue metabolism. These endotypes differ in their underlining pathogenesis, and may in the future have utility for patient treatment selection.
Collapse
Affiliation(s)
- J. P. M. Blair
- ProScion, Herlev, Denmark
- University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
- * E-mail:
| | | | - A. Platt
- Target & Translational Science, Respiratory, Inflammation and Autoimmunity (RIA), IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - M. Karsdal
- Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - A. -C. Bay-Jensen
- Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| |
Collapse
|
21
|
Karsdal MA, Verburg KM, West CR, Bay-Jensen AC, Keller DS, Arends RHGP. Serological biomarker profiles of rapidly progressive osteoarthritis in tanezumab-treated patients. Osteoarthritis Cartilage 2019; 27:484-492. [PMID: 30576794 DOI: 10.1016/j.joca.2018.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/29/2018] [Accepted: 12/03/2018] [Indexed: 02/02/2023]
Abstract
UNLABELLED There is a need for efficacious and safe pain treatments for OA (osteoarthritis). The nerve growth factor (NGF) antibody tanezumab is associated with high efficacy, but when combined with chronic NSAID treatment shows an increased risk of rapidly progressive osteoarthritis (RPOA) in a small group of patients. AIM The aim of this study was to phenotype with biochemical biomarkers of bone, cartilage, soft tissue, synovial metabolism OA patients who are at risk of developing RPOA type-2, for both limited and chronic NSAIDs users. MATERIAL AND METHODS The dataset consisted of OA patients participating in tanezumab trials who used NSAIDs <90 days (limited NSAID users) or chronic users (NSAIDs ≥90 days) over an average 10 month period. Biomarker data were available for 47 cases (RPOA type-2) and 92 controls. Non-linear and linear multivariable predictive models were developed. RESULTS By use of two biomarkers at baseline the receiver operating characteristic (ROC) curve area for RPOA type-2 in limited NSAID users was 71%, [CI] (60-83%). OA subjects with this biomarker phenotype had 8-fold higher confidence interval [CI][(2-33)] relative risk of developing RPOA type-2 as compared to OA patients without this phenotype. The AUC of the model in chronic NSAIDs users based on 5 biomarkers was 78%, [CI](69-88%), with 4-fold [CI (2-6)] relative risk of developing RPOA type-2. CONCLUSION In this hypothesis generating and exploratory study, we identified combinations of biomarkers associated with OA patients who develop RPOA type-2, which may be related to the pathology of the RPOA type-2 joint.
Collapse
Affiliation(s)
- M A Karsdal
- Nordic Bioscience, Herlev Hovedgade, DK-2730 Herlev, Denmark.
| | - K M Verburg
- Pfizer Inc., 445 Eastern Point Road, Groton, CT 06340, USA.
| | - C R West
- Pfizer Inc., 445 Eastern Point Road, Groton, CT 06340, USA.
| | - A C Bay-Jensen
- Nordic Bioscience, Herlev Hovedgade, DK-2730 Herlev, Denmark.
| | - D S Keller
- Pfizer Inc., 445 Eastern Point Road, Groton, CT 06340, USA.
| | - R H G P Arends
- Pfizer Inc., 445 Eastern Point Road, Groton, CT 06340, USA.
| |
Collapse
|
22
|
Löfvall H, Katri A, Dąbrowska A, Karsdal MA, Luo Y, He Y, Manon-Jensen T, Dziegiel MH, Bay-Jensen AC, Thudium CS, Henriksen K. GPDPLQ 1237-A Type II Collagen Neo-Epitope Biomarker of Osteoclast- and Inflammation-Derived Cartilage Degradation in vitro. Sci Rep 2019; 9:3050. [PMID: 30816326 PMCID: PMC6395810 DOI: 10.1038/s41598-019-39803-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 01/31/2019] [Indexed: 01/21/2023] Open
Abstract
C-telopeptide of type II collagen (CTX-II) has been shown to be a highly relevant biomarker of cartilage degradation in human rheumatic diseases, if measured in synovial fluid or urine. However, serum or plasma CTX-II have not been demonstrated to have any clinical utility to date. Here, we describe the GPDPLQ1237 ELISA which targets the EKGPDPLQ↓ neo-epitope, an elongated version of the CTX-II neo-epitope (EKGPDP↓), speculated to be a blood-precursor of CTX-II generated by the cysteine protease cathepsin K. Human osteoclast cartilage resorption cultures as well as oncostatin M and tumour necrosis factor α-stimulated bovine cartilage explant cultures were used to validate GPDPLQ1237 biologically by treating the cultures with the cysteine protease inhibitor E-64 and/or the matrix metalloproteinase (MMP) inhibitor GM6001 to assess the potential contributions of these two protease classes to GPDPLQ1237 release. Cartilage resorption-derived GPDPLQ1237 release was inhibited by E-64 (72.1% inhibition), GM6001 (75.5%), and E-64/GM6001 (91.5%), whereas CTX-II release was inhibited by GM6001 (87.0%) but not by E-64 (5.5%). Cartilage explant GPDPLQ1237 and CTX-II release were both fully inhibited by GM6001 but were not inhibited by E-64. No clinically relevant GPDPLQ1237 reactivity was identified in human serum, plasma, or urine from healthy donors or arthritis patients. In conclusion, the GPDPLQ1237 biomarker is released during osteoclast-derived cysteine protease- and MMP-mediated cartilage degradation in vitro, whereas CTX-II release is mediated by MMPs and not by cysteine proteases, as well as from MMP-mediated cartilage degradation under a pro-inflammatory stimulus. These findings suggest that GPDPLQ1237 may be relevant in diseases with pathological osteoclast activity and cartilage degradation. Further studies are required to validate the neo-epitope in human samples.
Collapse
Affiliation(s)
- Henrik Löfvall
- Nordic Bioscience, Herlev, Denmark.,Division of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund, Sweden
| | - Anna Katri
- Nordic Bioscience, Herlev, Denmark.,Department of Drug Design and Pharmacology, Copenhagen University, Copenhagen, Denmark
| | | | | | | | - Yi He
- Nordic Bioscience, Herlev, Denmark
| | | | - Morten H Dziegiel
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | | |
Collapse
|
23
|
Katri A, Dąbrowska A, Löfvall H, Ding M, Karsdal MA, Andreassen KV, Thudium CS, Henriksen K. Combining naproxen and a dual amylin and calcitonin receptor agonist improves pain and structural outcomes in the collagen-induced arthritis rat model. Arthritis Res Ther 2019; 21:68. [PMID: 30795801 PMCID: PMC6387482 DOI: 10.1186/s13075-019-1819-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 01/11/2019] [Indexed: 12/16/2022] Open
Abstract
Background Pain is a debilitating symptom of rheumatoid arthritis (RA), caused by joint inflammation and cartilage and bone destruction. Nonsteroidal anti-inflammatory drugs (NSAIDs) are used to treat pain and inflammation in RA, but are not disease-modifying and do not prevent joint destruction when administered alone. KBPs (Key Bioscience peptides) are synthetic peptides based on salmon calcitonin and are expected to inhibit bone resorption and to be chondroprotective. In this study, we investigated if combining a standard of care NSAID (naproxen) with a KBP resulted in improvement in pain scores, as well as disease activity and structural damage in a rat model of RA. Methods Collagen-induced arthritis (CIA) was induced in 40 female Lewis rats by immunization with porcine type II collagen; 10 rats were given sham injections. CIA rats were treated with KBP and/or naproxen. Health scores and joint scores were evaluated daily. Mechanical and cold allodynia tests and burrowing tests were used to assess pain-like behaviors. Blood samples were collected for biomarker testing, and paws were collected for histology and microcomputed tomography. Results Naproxen monotherapy increased the time until humane endpoints was reached, and improved health score, pain assessments, and trabecular thickness, while KBP monotherapy did not result in improvements. Combination therapy had improved efficacy over naproxen monotherapy; combination therapy resulted in improved health scores, and importantly reduced mechanical and cold allodynia assessment. Furthermore, protection of articular cartilage structure and preservation of bone structure and bone volume were also observed. Conclusions This study demonstrates that combining KBP and naproxen may be a relevant therapeutic strategy for RA, resulting in improvements to the overall health, pain, inflammation, and joint structure. Electronic supplementary material The online version of this article (10.1186/s13075-019-1819-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Katri
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Biomarkers and Research, Nordic Bioscience, Hovedgade 205-207, 2730, Herlev, Denmark
| | - Aneta Dąbrowska
- Biomarkers and Research, Nordic Bioscience, Hovedgade 205-207, 2730, Herlev, Denmark
| | - Henrik Löfvall
- Biomarkers and Research, Nordic Bioscience, Hovedgade 205-207, 2730, Herlev, Denmark.,Division of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund, Sweden
| | - Ming Ding
- Department of Orthopaedics and Traumatology, Institute of Clinical Research, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Morten A Karsdal
- Biomarkers and Research, Nordic Bioscience, Hovedgade 205-207, 2730, Herlev, Denmark
| | - Kim V Andreassen
- Biomarkers and Research, Nordic Bioscience, Hovedgade 205-207, 2730, Herlev, Denmark
| | - Christian S Thudium
- Biomarkers and Research, Nordic Bioscience, Hovedgade 205-207, 2730, Herlev, Denmark
| | - Kim Henriksen
- Biomarkers and Research, Nordic Bioscience, Hovedgade 205-207, 2730, Herlev, Denmark.
| |
Collapse
|
24
|
Bay-Jensen AC, Platt A, Jenkins MA, Weinblatt ME, Byrjalsen I, Musa K, Genovese MC, Karsdal MA. Tissue metabolite of type I collagen, C1M, and CRP predicts structural progression of rheumatoid arthritis. BMC Rheumatol 2019; 3:3. [PMID: 30886991 PMCID: PMC6390574 DOI: 10.1186/s41927-019-0052-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/03/2019] [Indexed: 01/30/2023] Open
Abstract
Background Biomarkers of rheumatoid arthritis (RA) disease activity typically measure inflammation or autoimmunity (e.g. CRP, RF). C1M and C3M, metabolites of type I and III collagen, are markers reflecting tissue metabolism. These markers have been documented to provide additional prognostic and predictive value compared to commonly used biomarkers. We investigated the relationship of high serum levels of C1M or C3M to radiographic progression, and benchmarked them to CRP and RF. Methods Placebo treated patients of the OSK1, 2 and 3 studies (Phase III clinical trials testing efficacy of fostamatinib) with baseline serum biomarkers C1M, C3M, CRP and RF were included (nBL = 474). Van der Heijde mTSS was calculated at baseline and 24-week (n24 = 261). Progression was defined as moderate or rapid by ΔmTSS ≥0.5 or ≥ 5 units/year. Patients were divided into subgroups; low (L), high (H) or very high (V) C1M, C3M and CRP, or RF negative, positive and high positive. Difference in clinical parameters were analyzed by Mann-Whitney or χ2tests, and modelling for prediction of progression by logistic regression including covariates (age, gender, BMI, and clinical assessment scores). Results Levels of C1M, C3M, CRP and RF were significantly (p < 0.05) associated with measures of disease activity and mTSS at baseline. For prognostic measures, there were 2.5 and 4-fold as many rapid progressors in the C1MH and CRPH (p < 0.05), and in the C1MV and CRPV groups (p < 0.001) compared C1ML and CRPL, respectively. C1M and CRP performed similarly in the predictive analysis, where high levels predicted moderate and rapid progression with odds ratio of 2.1 to 3.8 and 3.7 to 13.1 after adjustment for covariates. C3M and RF did not provide prognostic value alone. Discussion Serum C1M and CRP showed prognostic value and may be tools for enrichment of clinical trials with structural progressor. The two markers reflect two different aspect of disease pathogenesis (tissue turnover vs. inflammation), thus may provide individual and supplementary information. Electronic supplementary material The online version of this article (10.1186/s41927-019-0052-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne C Bay-Jensen
- 1Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, 2730 Herlev, Denmark
| | - Adam Platt
- 2Precision Medicine and Genomics, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | - Michael E Weinblatt
- 4Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA USA
| | - Inger Byrjalsen
- 1Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, 2730 Herlev, Denmark
| | - Kishwar Musa
- 1Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, 2730 Herlev, Denmark
| | - Mark C Genovese
- 5Division of Immunology and Rheumatology, Stanford University, Palo Alto, California, USA
| | - Morten A Karsdal
- 1Rheumatology, Nordic Bioscience, Biomarkers and Research, Herlev Hovedgade 205-207, 2730 Herlev, Denmark
| |
Collapse
|
25
|
Mitchell A, Wright G, Sampson SN, Martin M, Cummings K, Gaddy D, Watts AE. Clodronate improves lameness in horses without changing bone turnover markers. Equine Vet J 2018; 51:356-363. [DOI: 10.1111/evj.13011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 08/20/2018] [Indexed: 12/18/2022]
Affiliation(s)
- A. Mitchell
- Department of Large Animal Clinical Sciences Cornell University Ithaca New York USA
| | - G. Wright
- Department of Veterinary Pathobiology Cornell University Ithaca New York USA
| | - S. N. Sampson
- Department of Large Animal Clinical Sciences Cornell University Ithaca New York USA
| | - M. Martin
- Department of Large Animal Clinical Sciences Cornell University Ithaca New York USA
| | - K. Cummings
- Department of Population Medicine and Diagnostic Sciences Cornell University Ithaca New York USA
| | - D. Gaddy
- Department of Veterinary Integrative Biosciences Texas A&M University College Station Texas USA
| | - A. E. Watts
- Department of Large Animal Clinical Sciences Cornell University Ithaca New York USA
| |
Collapse
|
26
|
Kjelgaard-Petersen CF, Platt A, Braddock M, Jenkins MA, Musa K, Graham E, Gantzel T, Slynn G, Weinblatt ME, Karsdal MA, Thudium CS, Bay-Jensen AC. Translational Biomarkers and Ex Vivo Models of Joint Tissues as a Tool for Drug Development in Rheumatoid Arthritis. Arthritis Rheumatol 2018; 70:1419-1428. [PMID: 29669391 PMCID: PMC6174937 DOI: 10.1002/art.40527] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 04/10/2018] [Indexed: 12/15/2022]
Abstract
Objective Rheumatoid arthritis (RA) is a chronic and degenerative autoimmune joint disease that leads to disability, reduced quality of life, and increased mortality. Although several synthetic and biologic disease‐modifying antirheumatic drugs are available, there is still a medical need for novel drugs that control disease progression. As only 10% of experimental drug candidates for treatment of RA that enter phase I trials are eventually registered by the Food and Drug Administration, there is an immediate need for translational tools to facilitate early decision‐making in drug development. In this study, we aimed to determine if the inability of fostamatinib (a small molecule inhibitor of Syk) to demonstrate sufficient efficacy in phase III of a previous clinical study could have been predicted earlier in the development process. Methods Biomarkers of bone, cartilage, and interstitial matrix turnover (C‐telopeptide of type I collagen [CTX‐I], matrix metalloproteinase–derived types I, II, and III collagen neoepitopes [C1M, C2M, and C3M]) were measured in 450 serum samples from the Oral Syk Inhibition in Rheumatoid Arthritis 1 study (OSKIRA‐1, a phase III clinical study of the efficacy of fostamatinib in RA) at baseline and follow‐up. Additionally, the same biomarkers were subsequently measured in conditioned media from osteoclast, cartilage, and synovial membrane cultured with the active metabolite of fostamatinib, R406, to assess the level of suppression induced by the drug. Results In OSKIRA‐1 serum samples and osteoclast and cartilage cultures, fostamatinib suppressed the levels of CTX‐I and C2M. In OSKIRA‐1 serum samples and synovial membrane cultures, fostamatinib did not mediate any clinical or preclinical effect on either C1M or C3M, which have previously been associated with disease response and efficacy. Conclusion These data demonstrate that translational biomarkers are a potential tool for early assessment and decision‐making in drug development for RA treatment.
Collapse
|
27
|
Löfvall H, Newbould H, Karsdal MA, Dziegiel MH, Richter J, Henriksen K, Thudium CS. Osteoclasts degrade bone and cartilage knee joint compartments through different resorption processes. Arthritis Res Ther 2018; 20:67. [PMID: 29636095 PMCID: PMC5894194 DOI: 10.1186/s13075-018-1564-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/12/2018] [Indexed: 01/13/2023] Open
Abstract
Background Osteoclasts have been strongly implicated in osteoarthritic cartilage degradation, at least indirectly via bone resorption, and have been shown to degrade cartilage in vitro. The osteoclast resorption processes required to degrade subchondral bone and cartilage—the remodeling of which is important in the osteoarthritic disease process—have not been previously described, although cathepsin K has been indicated to participate. In this study we profile osteoclast-mediated degradation of bovine knee joint compartments in a novel in vitro model using biomarkers of extracellular matrix (ECM) degradation to assess the potential of osteoclast-derived resorption processes to degrade different knee joint compartments. Methods Mature human osteoclasts were cultured on ECMs isolated from bovine knees—articular cartilage, cortical bone, and osteochondral junction ECM (a subchondral bone-calcified cartilage mixture)—in the presence of inhibitors: the cystein protease inhibitor E-64, the matrix metalloproteinase (MMP) inhibitor GM6001, or the vacuolar-type H+-ATPase (V-ATPase) inhibitor diphyllin. Biomarkers of bone (calcium and C-terminal type I collagen (CTX-I)) and cartilage (C2M) degradation were measured in the culture supernatants. Cultures without osteoclasts were used as background samples. Background-subtracted biomarker levels were normalized to the vehicle condition and were analyzed using analysis of variance with Tukey or Dunnett’s T3 post hoc test, as applicable. Results Osteochondral CTX-I release was inhibited by E-64 (19% of vehicle, p = 0.0008), GM6001 (51% of vehicle, p = 0.013), and E-64/GM6001 combined (4% of vehicle, p = 0.0007)—similarly to bone CTX-I release. Diphyllin also inhibited osteochondral CTX-I release (48% of vehicle, p = 0.014), albeit less than on bone (4% of vehicle, p < 0.0001). Osteochondral C2M release was only inhibited by E-64 (49% of vehicle, p = 0.07) and GM6001 (14% of vehicle, p = 0.006), with complete abrogation when combined (0% of vehicle, p = 0.004). Cartilage C2M release was non-significantly inhibited by E-64 (69% of vehicle, p = 0.98) and was completely abrogated by GM6001 (0% of vehicle, p = 0.16). Conclusions Our study supports that osteoclasts can resorb non-calcified and calcified cartilage independently of acidification. We demonstrated both MMP-mediated and cysteine protease-mediated resorption of calcified cartilage. Osteoclast functionality was highly dependent on the resorbed substrate, as different ECMs required different osteoclast processes for degradation. Our novel culture system has potential to facilitate drug and biomarker development aimed at rheumatic diseases, e.g. osteoarthritis, where pathological osteoclast processes in specific joint compartments may contribute to the disease process. Electronic supplementary material The online version of this article (10.1186/s13075-018-1564-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Henrik Löfvall
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark.,Division of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund, Sweden
| | - Hannah Newbould
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Morten A Karsdal
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Morten H Dziegiel
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Johan Richter
- Division of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund, Sweden
| | - Kim Henriksen
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | | |
Collapse
|
28
|
Effects of dietary weight loss with and without exercise on interstitial matrix turnover and tissue inflammation biomarkers in adults with knee osteoarthritis: the Intensive Diet and Exercise for Arthritis trial (IDEA). Osteoarthritis Cartilage 2017; 25:1822-1828. [PMID: 28756278 PMCID: PMC5650925 DOI: 10.1016/j.joca.2017.07.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/06/2017] [Accepted: 07/19/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To examine the effects of dietary weight loss, with and without exercise, on selected soluble biomarkers in overweight and obese older adults with symptomatic knee osteoarthritis (OA). DESIGN Blood samples were analyzed from 429 participants in the Intensive Diet and Exercise for Arthritis (IDEA) trial randomized to either an 18 month exercise control group (E), weight loss diet (D), or D + E. C1M, C2M, C3M and CRPM biomarkers and interleukin-6 (IL-6) were quantitated using ELISAs. Radiographic progression was defined as a decrease in joint space width of ≥0.7 mm. Statistical modeling of group means and associations used mixed models adjusted for visit, baseline body mass index (BMI), gender, and baseline values of the outcome. RESULTS Compared to the E control group, C1M was significantly lower in the D and D + E groups at both 6 and 18 months while C3M was significantly lower in D and D + E at 6 months and in D + E at 18 months. C2M did not change in any group. Using data from all groups, change in C1M (P < 0.0001), C3M (P < 0.0001), as well as CRPM (P = 0.0004) from baseline to 18 months was positively associated with change in weight. No marker was associated with change in Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain or radiographic progression. C3M (P = 0.008) and CRPM (P = 0.028) were positively associated with change in WOMAC function. Change in IL-6 was positively associated with change in C1M, C3M, and CRPM. CONCLUSION Overweight and obese adults with knee OA who lost weight from diet and diet plus exercise reduced serum markers of interstitial matrix turnover and inflammation but not type II collagen degradation.
Collapse
|