1
|
Slouma M, Bouzid S, Dhahri R, Rahmouni S, Litaiem N, Gharsallah I, Metoui L, Louzir B. Matrix Metalloproteinases; A Biomarker of Disease Activity and Prognosis in Spondyloarthritis: A Narrative Review. Curr Rev Clin Exp Pharmacol 2023; 18:31-38. [PMID: 35049445 DOI: 10.2174/2772432817666220113112809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 12/04/2021] [Accepted: 12/15/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Matrix metalloproteinases, as components of the proteolytic system, are deemed to be implicated in the pathogenesis and progression of several rheumatic diseases. Their role in spondyloarthritis has been investigated by several studies. OBJECTIVE This article aims to review and summarize the current knowledge related to metalloproteinases in patients with spondyloarthritis. METHODS To examine the association between matrix metalloproteinases and spondyloarthritis, we conducted a narrative review using a literature search in SCOPUS for English-language sources. The search included studies published from the database inception to December 2020. RESULTS A total number of 74 articles were included. It was found that levels of matrix metalloproteinases 3 were higher in radiographic axial spondyloarthritis patients and seemed to play a role in the progression of joint damage. The levels of matrix metalloproteinases 1, 2, and 9 were upregulated in psoriatic arthritis patients compared to psoriasis and could identify psoriasis patients who would develop rheumatic manifestations. The levels of matrix metalloproteinases correlated significantly with disease activity in ankylosing spondylitis and decreased upon treatment with Tumor Necrosis Factor inhibitors (TNFi). CONCLUSION Excessive matrix metalloproteinases activity is associated with articular destruction. Their levels can reflect disease activity, structural damage, and response to TNFi in patients with spondyloarthritis. Nevertheless, further studies are needed to confirm these results.
Collapse
Affiliation(s)
- Maroua Slouma
- Department of Rheumatology, Military Hospital, Tunis, Tunisia
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
| | - Sirine Bouzid
- Department of Rheumatology, Military Hospital, Tunis, Tunisia
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
| | - Rim Dhahri
- Department of Rheumatology, Military Hospital, Tunis, Tunisia
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
| | - Safa Rahmouni
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
- Department of Rheumatology, Rabta Hospital, Tunis, Tunisia
| | - Noureddine Litaiem
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
- Department of Dermatology, Charles Nicolle Hospital, Tunis, Tunisia
| | - Imen Gharsallah
- Department of Rheumatology, Military Hospital, Tunis, Tunisia
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
| | - Leila Metoui
- Department of Rheumatology, Military Hospital, Tunis, Tunisia
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
| | - Bassem Louzir
- Department of Rheumatology, Tunis El Manar University, Tunis, Tunisia
- Department of Internal Medicine, Military Hospital, Tunis, Tunisia
| |
Collapse
|
2
|
Wirth T, Balandraud N, Boyer L, Lafforgue P, Pham T. Biomarkers in psoriatic arthritis: A meta-analysis and systematic review. Front Immunol 2022; 13:1054539. [PMID: 36532039 PMCID: PMC9749424 DOI: 10.3389/fimmu.2022.1054539] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction Psoriatic arthritis (PsA) is a chronic inflammatory disease that frequently develops in patients with psoriasis (PsO) but can also occur spontaneously. As a result, PsA diagnosis and treatment is commonly delayed, or even missed outright due to the manifold of clinical presentations that patients often experience. This inevitably results in progressive articular damage to axial and peripheral joints and entheses. As such, patients with PsA frequently experience reduced expectancy and quality of life due to disability. More recently, research has aimed to improve PsA diagnosis and prognosis by identifying novel disease biomarkers. Methods Here, we conducted a systematic review of the published literature on candidate biomarkers for PsA diagnosis and prognosis in MEDLINE(Pubmed), EMBase and the Cochrane library with the goal to identify clinically applicable PsA biomarkers. Meta-analyses were performed when a diagnostic bone and cartilage turnover biomarker was reported in 2 or moredifferent cohorts of PsA and control. Results We identified 1444 publications and 124 studies met eligibility criteria. We highlighted bone and cartilage turnover biomarkers, genetic markers, and autoantibodies used for diagnostic purposes of PsA, as well as acute phase reactant markers and bone and cartilage turnover biomarkers for activity or prognostic severity purposes. Serum cartilage oligometrix metalloproteinase levels were significantly increased in the PsA sera compared to Healthy Control (HC) with a standardized mean difference (SMD) of 2.305 (95%CI 0.795-3.816, p=0.003) and compared to osteoarthritis (OA) with a SMD of 0.783 (95%CI 0.015-1.551, p=0.046). The pooled serum MMP-3 levels were significantly higher in PsA patients than in PsO patients with a SMD of 0.419 (95%CI 0.119-0.719; p=0.006), but no significant difference was highlighted when PsA were compared to HC. While we did not identify any new genetic biomarkers that would be useful in the diagnosis of PsA, recent data with autoantibodies appear to be promising in diagnosis, but no replication studies have been published. Conclusion In summary, no specific diagnostic biomarkers for PsA were identified and further studies are needed to assess the performance of potential biomarkers that can distinguish PsA from OA and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Theo Wirth
- Rheumatology Department, Sainte Marguerite Hospital, Aix-Marseille University, APHM, Marseille, France,*Correspondence: Theo Wirth,
| | - Nathalie Balandraud
- Rheumatology Department, Sainte Marguerite Hospital, Aix-Marseille University, APHM, Marseille, France,Autoimmune Arthritis Laboratory, INSERM UMRs1097, Aix Marseille University, Marseille, France
| | - Laurent Boyer
- School of Medicine, EA 3279, CEReSS, Research Center on Health Services and Quality of Life, Aix Marseille University, Marseille, France
| | - Pierre Lafforgue
- Rheumatology Department, Sainte Marguerite Hospital, Aix-Marseille University, APHM, Marseille, France
| | - Thao Pham
- Rheumatology Department, Sainte Marguerite Hospital, Aix-Marseille University, APHM, Marseille, France
| |
Collapse
|
3
|
Gurke R, Bendes A, Bowes J, Koehm M, Twyman RM, Barton A, Elewaut D, Goodyear C, Hahnefeld L, Hillenbrand R, Hunter E, Ibberson M, Ioannidis V, Kugler S, Lories RJ, Resch E, Rüping S, Scholich K, Schwenk JM, Waddington JC, Whitfield P, Geisslinger G, FitzGerald O, Behrens F, Pennington SR. Omics and Multi-Omics Analysis for the Early Identification and Improved Outcome of Patients with Psoriatic Arthritis. Biomedicines 2022; 10:2387. [PMID: 36289648 PMCID: PMC9598654 DOI: 10.3390/biomedicines10102387] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
The definitive diagnosis and early treatment of many immune-mediated inflammatory diseases (IMIDs) is hindered by variable and overlapping clinical manifestations. Psoriatic arthritis (PsA), which develops in ~30% of people with psoriasis, is a key example. This mixed-pattern IMID is apparent in entheseal and synovial musculoskeletal structures, but a definitive diagnosis often can only be made by clinical experts or when an extensive progressive disease state is apparent. As with other IMIDs, the detection of multimodal molecular biomarkers offers some hope for the early diagnosis of PsA and the initiation of effective management and treatment strategies. However, specific biomarkers are not yet available for PsA. The assessment of new markers by genomic and epigenomic profiling, or the analysis of blood and synovial fluid/tissue samples using proteomics, metabolomics and lipidomics, provides hope that complex molecular biomarker profiles could be developed to diagnose PsA. Importantly, the integration of these markers with high-throughput histology, imaging and standardized clinical assessment data provides an important opportunity to develop molecular profiles that could improve the diagnosis of PsA, predict its occurrence in cohorts of individuals with psoriasis, differentiate PsA from other IMIDs, and improve therapeutic responses. In this review, we consider the technologies that are currently deployed in the EU IMI2 project HIPPOCRATES to define biomarker profiles specific for PsA and discuss the advantages of combining multi-omics data to improve the outcome of PsA patients.
Collapse
Affiliation(s)
- Robert Gurke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Annika Bendes
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - John Bowes
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WU, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Michaela Koehm
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | | | - Anne Barton
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WU, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Dirk Elewaut
- VIB-UGent Center for Inflammation Research, Ghent University, 9052 Ghent, Belgium
| | - Carl Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8QQ, UK
| | - Lisa Hahnefeld
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | | | - Ewan Hunter
- Oxford BioDynamics Limited, Oxford OX4 2JZ, UK
| | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Vassilios Ioannidis
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Sabine Kugler
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer IAIS, Institute for Intelligent Analysis and Information Systems, Schloss Birlinghoven 1, 53757 Sankt Augustin, Germany
| | - Rik J. Lories
- Department of Development and Regeneration, KU Leuven, Skeletal Biology and Engineering Research Centre, P.O. Box 813 O&N, Herestraat 49, 3000 Leuven, Belgium
| | - Eduard Resch
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Stefan Rüping
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer IAIS, Institute for Intelligent Analysis and Information Systems, Schloss Birlinghoven 1, 53757 Sankt Augustin, Germany
| | - Klaus Scholich
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jochen M. Schwenk
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - James C. Waddington
- Atturos Ltd., c/o UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Phil Whitfield
- Glasgow Polyomics, College of Medical, Veterinary and Life Sciences, Garscube Campus, University of Glasgow, Glasgow G61 1QH, UK
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Oliver FitzGerald
- UCD Conway Institute, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | - Frank Behrens
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Stephen R. Pennington
- Atturos Ltd., c/o UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
- UCD Conway Institute, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| | | |
Collapse
|
4
|
Geneva-Popova M, Popova-Belova S, Popova V, Chompalov K, Batalov A. Assessment of serum and synovial fluid MMP-3 and MPO as biomarkers for psoriatic arthritis and their relation to disease activity indices. Rheumatol Int 2022; 42:1605-1615. [PMID: 35708757 DOI: 10.1007/s00296-022-05159-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
Research on biomarkers for the diagnosis and monitoring of psoriatic arthritis (PsA) is ongoing. The purpose of this study was to assess the potential of serum and synovial fluid matrix metalloproteinase-3 (MMP-3) and myeloperoxidase (MPO) as biomarkers for PsA and their relation to disease activity indices. This case-control study involved 156 psoriatic arthritis patients, 50 gonarthrosis patients, and 30 healthy controls. The target parameters were measured with the enzyme-linked immunosorbent assay (ELISA) kits. Serum MMP-3 and MPO levels were elevated in the PsA patients in comparison to the two control groups (p < 0.001) and distinguished PsA from GoA patients and healthy controls with 100% accuracy. Synovial MMP-3 discriminated PsA from GoA patients irrespective of the presence of crystals (AUC = 1.00). PsA patients with crystals in the synovial fluid had elevated synovial MPO (p < 0.001) and were distinguished from PsA patients without crystals with accuracy of 88.50% and from GoA patients with accuracy of 88.30%. Synovial fluid MPO was positively associated with the following indicators of disease activity: VAS (rs = 0.396); DAPSA (rs = 0.365); mCPDAI (rs = 0.323). Synovial MMP-3 showed a weaker positive association with DAPSA (rs = 0.202) and mCPDAI (rs = 0.223). Our results suggest that serum MMP-3 and MPO could serve as biomarkers for PsA. Synovial fluid MMP-3 showed a potential as a biomarker for PsA versus GoA. Synovial MPO could be utilized as a marker for the presence of crystals in PsA patients.
Collapse
Affiliation(s)
- Mariela Geneva-Popova
- Department of Propedeutic of Internal Diseases, Clinic of Rheumatology, Medical University of Plovdiv, University General Hospital "SvetiGeorgi", Plovdiv, Bulgaria.
| | - Stanislava Popova-Belova
- Department of Propedeutic of Internal Diseases, Clinic of Rheumatology, Medical University of Plovdiv, University General Hospital "SvetiGeorgi", Plovdiv, Bulgaria
| | - Velichka Popova
- Clinic of Rheumatology, Medical University of Plovdiv, University General Hospital "Kaspela", Plovdiv, Bulgaria
| | - Kostadin Chompalov
- Department of Rheumatology, Medical University, University General Hospital "St. Georgi", Plovdiv, Bulgaria
| | - Anastas Batalov
- Department of Propedeutic of Internal Diseases, Clinic of Rheumatology, Medical University of Plovdiv, University General Hospital "Kaspela", Plovdiv, Bulgaria
| |
Collapse
|
5
|
Elliott A, McGonagle D, Rooney M. Integrating imaging and biomarker assessment to better define psoriatic arthritis and predict response to biologic therapy. Rheumatology (Oxford) 2021; 60:vi38-vi52. [PMID: 34951926 PMCID: PMC8709569 DOI: 10.1093/rheumatology/keab504] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
The treatment options for PsA have substantially expanded over the last decade. Approximately 40% of patients will not respond to first-line anti-TNF-α therapies. There is limited data to help clinicians select the most appropriate biologic therapy for PsA patients, including guidance for decisions on biologic therapy switching. In this review we will examine the current understanding of predictors of response to treatment. Imaging technology has evolved to allow us to better study psoriatic disease and define disease activity, including synovitis and enthesitis. Enthesitis is implicated in the pathogenesis, diagnosis and prognosis of PsA. It appears to be a common thread among all of the various PsA clinical presentations. Enthesitis mainly manifests as tenderness, which is difficult to distinguish from FM, chronic pain and mechanically associated enthesopathy, and it might be relevant for understanding the apparent 40% failure of existing therapy. Excess adipose tissue makes if more difficult to detect joint swelling clinically, as many PsA patients have very high BMIs. Integrating imaging and clinical assessment with biomarker analysis could help to deliver stratified medicine in PsA and allow better treatment decision making. This could include which patients require ongoing biologic therapy, which class of biologic therapy that should be, and who alternatively requires management of non-inflammatory disease.
Collapse
Affiliation(s)
- Ashley Elliott
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton Hospital, Leeds, UK
| | - Madeleine Rooney
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
6
|
Yan D, Gudjonsson JE, Le S, Maverakis E, Plazyo O, Ritchlin C, Scher JU, Singh R, Ward NL, Bell S, Liao W. New Frontiers in Psoriatic Disease Research, Part I: Genetics, Environmental Triggers, Immunology, Pathophysiology, and Precision Medicine. J Invest Dermatol 2021; 141:2112-2122.e3. [PMID: 34303522 PMCID: PMC8384663 DOI: 10.1016/j.jid.2021.02.764] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
Abstract
Psoriasis is a chronic inflammatory condition characterized by systemic immune dysregulation. Over the past several years, advances in genetics, microbiology, immunology, and mouse models have revealed the complex interplay between the heritable and microenvironmental factors that drive the development of psoriatic inflammation. In the first of this two-part review series, the authors will discuss the newest insights into the pathogenesis of psoriatic disease and highlight how the evolution of these scientific fields has paved the way for a more personalized approach to psoriatic disease treatment.
Collapse
Affiliation(s)
- Di Yan
- Ronald O. Perelman Department of Dermatology, NYU Langone Health, New York, New York, USA
| | | | - Stephanie Le
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Olesya Plazyo
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Ritchlin
- Center for Musculoskeletal Research, Division of Allergy, Immunology and Rheumatology, University of Rochester School of Medicine & Dentistry, Rochester, New York, USA
| | - Jose U Scher
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, New York, USA
| | - Roopesh Singh
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Nicole L Ward
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Stacie Bell
- National Psoriasis Foundation, Portland, Oregon, USA
| | - Wilson Liao
- UCSF Department of Dermatology, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
7
|
Assessment of Selected Matrix Metalloproteinases (MMPs) and Correlation with Cytokines in Psoriatic Patients. Mediators Inflamm 2021; 2021:9913798. [PMID: 34305455 PMCID: PMC8263227 DOI: 10.1155/2021/9913798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/16/2021] [Indexed: 11/17/2022] Open
Abstract
Metalloproteinases (MMPs) and cytokines have a great impact on the pathogenesis of psoriasis. Cytokines, as key mediators of inflammation and autoimmune processes, play a crucial role in the regulation of MMP expression in different cell types. Parallel, MMPs have an influence on cytokine production. This interaction was not well recognized in psoriatic patients. Our study is aimed at assessing the selected serum MMP levels and their correlations with cytokine levels in the serum of psoriatic patients. We observed a significantly elevated level of pro-MMP-1 and MMP-9 in psoriatic patients' serum in comparison to the control group. We did not observe any statistically significant differences of MMP-3 and pro-MMP-10 between the psoriatic patients and the control group. We did not observe any statistically significant differences in all the studied MMP levels between the patients with and without psoriatic arthritis (PsA). MMP-3 level correlated positively with proinflammatory cytokines, i.e., IL-12p/70, IL-17A, and TNF-α as well as MMP-3 and pro MMP-1 correlated positively with IL-4 in the psoriatic patients. In the control group, a positive correlation between pro-MMP-1 and TNF-α was found. These results confirm MMPs and Th1 and Th17 cytokine interaction in the inflammatory regulation in psoriasis.
Collapse
|
8
|
Waszczykowski M, Bednarski I, Lesiak A, Waszczykowska E, Narbutt J, Fabiś J. The influence of tumour necrosis factor α inhibitors treatment - etanercept on serum concentration of biomarkers of inflammation and cartilage turnover in psoriatic arthritis patients. Postepy Dermatol Alergol 2020; 37:995-1000. [PMID: 33603621 PMCID: PMC7874864 DOI: 10.5114/ada.2020.96705] [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: 05/10/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Effective treatment in psoriatic arthritis (PsA) patients can protect them from severe musculoskeletal complications. For appropriate monitoring of anti-tumour necrosis factor α (anti-TNF-α) treatment in PsA, specific biomarkers are needed. AIM To investigate whether biological treatment with anti-TNF-α (etanercept 50 mg once a week subcutaneously) affects the activity of selected mediators of inflammation and destruction of articular cartilage: interleukin-6 (IL-6), interleukin-18 (IL-18), matrix metalloproteinases 1 and 3 (MMP-1, MMP-3), cartilage oligomeric matrix protein (COMP), human cartilage glycoprotein (YKL-40) in serum of patients with PsA. MATERIAL AND METHODS The study included 25 patients with PsA. The concentration of IL-6, IL-18, MMP-1, MMP-3, COMP and YKL-40 in serum was determined before, and 6 and 12 weeks after the beginning of anti-TNF-α treatment. Clinical severity of the disease according to the Body Surface Area, Psoriasis Area and Severity Index and Dermatology Life Quality Index as well as tender and swollen joint count (TJC, SJC) were also evaluated. RESULTS The study disclosed a statistically significant reduction in the serum concentration of IL-6, MMP-1 and YKL-40 in PsA patients after 6 and 12 weeks from the beginning of anti-TNF-α treatment (p = 0.00018 for IL-6; p = 0.01242 for MMP-1; p = 0.03263 for YKL-40). CONCLUSIONS IL-6, MMP-1 and YKL-40 may be useful for monitoring the effectiveness of anti-TNF-α treatment.
Collapse
Affiliation(s)
- Michał Waszczykowski
- Department of Arthroscopy, Minimally Invasive Surgery and Sports Traumatology, Medical University of Lodz, Lodz, Poland
| | - Igor Bednarski
- Dermatology, Paediatric Dermatology and Oncology Clinic, Medical University of Lodz, Lodz, Poland
| | - Aleksandra Lesiak
- Dermatology, Paediatric Dermatology and Oncology Clinic, Medical University of Lodz, Lodz, Poland
| | | | - Joanna Narbutt
- Dermatology, Paediatric Dermatology and Oncology Clinic, Medical University of Lodz, Lodz, Poland
| | - Jarosław Fabiś
- Department of Arthroscopy, Minimally Invasive Surgery and Sports Traumatology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
9
|
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
|
10
|
Garnero P, Landewé R, Chapurlat RD. The role of biochemical markers of joint tissue remodelling to predict progression and treatment efficacy in inflammatory rheumatic diseases. Rheumatology (Oxford) 2020; 59:1207-1217. [PMID: 32011708 DOI: 10.1093/rheumatology/kez647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/02/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Structural damage is a hallmark in RA, spondyloarthropy (SpA) and psoriatric arthritis (PsA). Its progression is difficult to predict and current radiological or inflammatory biological markers lack sensitivity. Biochemical markers of bone, cartilage and synovial tissues provide a dynamic indication of the anabolism and catabolism of joint tissues and can be easily measured by immunoassays. Novel biochemical markers including post-translational modifications of matrix proteins and enzyme-generated neoepitopes with increased tissue and/or biological pathway specificity have been developed. Their evaluation in clinical trials of novel biologic therapies and epidemiological studies indicated that their measurements could be useful to predict progression of structural damage and treatment efficacy, independently of current clinical, radiological and biological indices of disease activity. In this paper we briefly describe the latest developments in biochemical markers and critically analyse the clinical data assessing the utility of established and novel biochemical markers in RA, SpA and PsA.
Collapse
Affiliation(s)
- Patrick Garnero
- INSERM Research Unit 1033-Lyos, Hôpital E. Herriot, Lyon, France
| | - Robert Landewé
- Department of Clinical Immunology and Rheumatology, Academic Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
11
|
Lorenzin M, Ometto F, Ortolan A, Felicetti M, Favero M, Doria A, Ramonda R. An update on serum biomarkers to assess axial spondyloarthritis and to guide treatment decision. Ther Adv Musculoskelet Dis 2020; 12:1759720X20934277. [PMID: 32636944 PMCID: PMC7315656 DOI: 10.1177/1759720x20934277] [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: 03/12/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022] Open
Abstract
Axial spondyloarthritis (axSpA) is a group of debilitating, chronic, rheumatic conditions characterized by inflammation and new bone formation, mainly involving the spine and the sacroiliac joints. The lack of biomarkers in axSpA is well known. Despite significant treatment advances in recent years thanks to the introduction of drugs with a new mode of action, such as new biologic and targeted synthetic disease-modifying antirheumatic drugs, no relevant improvement in the identification of disease biomarkers has been achieved. Common parameters, such as erythrocyte sedimentation rate and C-reactive protein, which are routinely used to measure systemic inflammation, are the sole markers available to date and are not adequate to assess disease activity in all patients. The aim of this study is to review the most promising serum biomarkers that may help treatment decision in axSpA via a proper assessment of disease activity and identification of negative prognostic factors.
Collapse
Affiliation(s)
- Mariagrazia Lorenzin
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Francesca Ometto
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Augusta Ortolan
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Mara Felicetti
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Marta Favero
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Padova, Italy
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine -DIMED, University of Padova, Via Giustiniani 2, Padova, 35128, Italy
| |
Collapse
|
12
|
Castillo R, Scher JU. Not your average joint: Towards precision medicine in psoriatic arthritis. Clin Immunol 2020; 217:108470. [PMID: 32473975 DOI: 10.1016/j.clim.2020.108470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/31/2022]
Abstract
Precision medicine, propelled by advances in multi-omics methods and analytics, aims to revolutionize patient care by using clinically-actionable molecular markers to guide diagnostic and therapeutic decisions. We describe the applications of precision medicine in risk stratification, drug selection, and treatment response prediction in psoriatic arthritis, for which targeted, personalized approaches are steadily emerging.
Collapse
Affiliation(s)
- Rochelle Castillo
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, NY, United States of America
| | - Jose U Scher
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, NY, United States of America; Psoriatic Arthritis Center, New York University School of Medicine, New York, NY, United States of America.
| |
Collapse
|
13
|
Lorenzin M, Ortolan A, Felicetti M, Favero M, Vio S, Zaninotto M, Polito P, Cosma C, Scapin V, Lacognata C, Ramonda R. Serological Biomarkers in Early Axial Spondyloarthritis During 24-Months Follow Up (Italian Arm of Space Study). Front Med (Lausanne) 2019; 6:177. [PMID: 31440510 PMCID: PMC6692922 DOI: 10.3389/fmed.2019.00177] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/19/2019] [Indexed: 12/17/2022] Open
Abstract
Objectives: The study aimed to evaluate biomarkers facilitating early axial-spondyloarthritis (axSpA) diagnosis and disease activity and imaging indices correlated. Materials and Methods: Seventy-five patients with low back pain (LBP) (≥3 months, ≤2 years, onset ≤45 years) participating in the Italian arm of the SpondyloArthritis-Caught-Early (SPACE) study underwent a physical examination, questionnaires, laboratory tests, spine, and sacroiliac joints (SIJ) X-rays and magnetic resonance imaging (MRI) at baseline and during a 24-months follow-up. Two expert rheumatologists formulated axSpA diagnosis and assessed fulfillment of Assessment of SpondyloArthritis International Society (ASAS) criteria. Disease activity and physical functioning were assessed using imaging, clinical, and serological indices. Spine and SIJ MRI and X-rays were scored independently by 2 readers following the Spondyloarthritis Research Consortium of Canada (SPARCC), mSASSS, and mNY-criteria. Patients were classified in accordance to ASAS criteria as: 21 patients classified according to axSpA imaging arm; 29 patients classified according to axSpA clinical ± imaging arm; 25 patients not fulfilling ASAS criteria. Results: At baseline biomarker levels were not significantly increased in any of the patient groups. Instead, a significant decrease of all functional and disease activity indices from baseline to 24 months was observed in all the three groups. In the same period, there were no significant variation in the serological markers values within each group. The correlations between IL-17 and IL-23 and clinical and functional indices were not significant. On the other hand, significant correlations were found between IL-22 and Bath Ankylosing Spondylitis Functional Index (BASFI), Bath Ankylosing Spondylitis Patient Global Score (BASG1), Health Assessment Questionnaire (HAQ), Visual Analog Scale (VAS pain); MMP3 and mSASSS; MMP3 and hsCRP. Conclusions: Although not significantly higher in any of the cohorts, IL-22, MMP3, and hsCRP values correlated with some disease activity indices and with mSASSS. Further studies are warranted to confirm these preliminary findings.
Collapse
Affiliation(s)
- Mariagrazia Lorenzin
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Augusta Ortolan
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Mara Felicetti
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Marta Favero
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Stefania Vio
- Radiology Unit, University of Padova, Padova, Italy
| | | | - Pamela Polito
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Chiara Cosma
- Medicine of Laboratory, University of Padova, Padova, Italy
| | - Vanna Scapin
- Radiology Unit, University of Padova, Padova, Italy
| | | | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| |
Collapse
|
14
|
Morita A, Okuyama R, Katoh N, Tateishi C, Masuda K, Komori T, Ogawa E, Makino T, Nishida E, Nishimoto S, Muramoto K, Tsuruta D, Ihn H. Efficacy and safety of adalimumab in Japanese patients with psoriatic arthritis and inadequate response to non-steroidal anti-inflammatory drugs (NSAIDs): A prospective, observational study. Mod Rheumatol 2019; 30:155-165. [DOI: 10.1080/14397595.2019.1589739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryuhei Okuyama
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Norito Katoh
- Department of Dermatology, Kyoto Prefectural University of Medicine Graduate School of Medical Sciences, Kyoto, Japan
| | - Chiharu Tateishi
- Department of Dermatopathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koji Masuda
- Department of Dermatology, Kyoto Prefectural University of Medicine Graduate School of Medical Sciences, Kyoto, Japan
| | - Toshifumi Komori
- Department of Dermatology, Northern Medical Center, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eisaku Ogawa
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takamitsu Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Emi Nishida
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | | | - Daisuke Tsuruta
- Department of Dermatopathology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
15
|
Furst DE, Belasco J, Louie JS. Genetic and inflammatory factors associated with psoriatic arthritis: Relevance to diagnosis and management. Clin Immunol 2019; 202:59-75. [DOI: 10.1016/j.clim.2019.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/21/2019] [Accepted: 02/04/2019] [Indexed: 12/22/2022]
|
16
|
Mahendran SM, Chandran V. Exploring the Psoriatic Arthritis Proteome in Search of Novel Biomarkers. Proteomes 2018; 6:proteomes6010005. [PMID: 29364831 PMCID: PMC5874764 DOI: 10.3390/proteomes6010005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/16/2018] [Accepted: 01/21/2018] [Indexed: 12/28/2022] Open
Abstract
Psoriatic arthritis (PsA) is an inflammatory arthritis which develops in up to one-third of patients suffering from the cutaneous disorder, psoriasis. The complex and heterogeneous nature of PsA renders it difficult to diagnose, leading to poor outcomes and, therefore, warrants an examination into soluble biomarkers, which may facilitate early detection of the disease. Protein biomarkers are a dynamic resource of pathophysiological information able to provide an immediate reflection of pathological changes caused by disease. Investigations of the serum and synovial fluid of PsA patients has provided new insights into the molecular basis of this disease and led to the identification of sensitive diagnostic and prognostic biomarkers. The collection of novel PsA biomarkers identified through proteomic studies has been reviewed below.
Collapse
Affiliation(s)
- Shalini M Mahendran
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada.
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada.
| | - Vinod Chandran
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada.
- Centre for Prognosis Studies in Rheumatic Diseases, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON M5T 1M8, Canada.
- Division of Rheumatology, Department of Medicine, University of Toronto, Toronto, ON M5T 1A1, Canada.
- Institute of Medical Science, University of Toronto, Toronto, ON M5T 1A1, Canada.
| |
Collapse
|
17
|
Bakirci Ureyen S, Ivory C, Kalyoncu U, Karsh J, Aydin SZ. What does evidence-based medicine tell us about treatments for different subtypes of psoriatic arthritis? A systematic literature review on randomized controlled trials. Rheumatol Adv Pract 2018; 2:rkx019. [PMID: 31431950 PMCID: PMC6649907 DOI: 10.1093/rap/rkx019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/04/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE PsA is a heterogeneous disease with various subtypes of joint manifestations, which can affect the homogeneity of randomized controlled trials (RCTs). The aim of this systematic literature review was to evaluate the inclusion criteria, demographics and outcomes of RCTs to see whether the whole spectrum of PsA was represented. METHODS Medline, EMBASE and Cochrane databases were screened for RCTs on the efficacy of any treatment for PsA up to 4 October 2016 to investigate the inclusion criteria, demographics, outcomes and efficacy. RESULTS Two thousand and sixty-eight abstracts were identified at screening; 76 articles and 52 conference proceedings were included in the final analysis. The main inclusion criteria always included the number of active joints and never axial symptoms, enthesitis nor dactylitis. Only 10 studies provided information about subtypes, of which symmetrical polyarthritis was the main subtype. Mean (s.d.) tender and swollen joints were between 7.8 and 35.8 (1.8-22.1) and between 5.2 and 25.2 (1.5-16.2), respectively. All studies had responses in joint counts as their primary outcome. Responses in enthesitis and dactylitis were usually secondary or tertiary outcomes. Response in BASDAI was among the outcomes in four studies. The comparison of efficacy in polyarticular vs oligoarticular disease was given in three studies, whereas no information was available for DIP joint disease or arthritis mutilans. CONCLUSION There is evidence in the literature to guide clinicians on how to treat PsA patients with polyarticular disease, but there is a gap in knowledge about the other subtypes. PROTOCOL REGISTRATION The study protocol is registered at PROSPERO (CRD42017053907).
Collapse
Affiliation(s)
| | - Catherine Ivory
- University of Ottawa, Faculty of Medicine, Rheumatology, Ottawa, ON, Canada
| | - Umut Kalyoncu
- Faculty of Medicine, Rheumatology, Hacettepe University, Ankara, Turkey
| | - Jacob Karsh
- University of Ottawa, Faculty of Medicine, Rheumatology, Ottawa, ON, Canada
| | - Sibel Zehra Aydin
- University of Ottawa, Faculty of Medicine, Rheumatology, Ottawa, ON, Canada
- Faculty of Medicine, Ottawa Hospital Research Institute, Rheumatology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
18
|
Clinical Features of Psoriatic Arthritis: a Comprehensive Review of Unmet Clinical Needs. Clin Rev Allergy Immunol 2017; 55:271-294. [DOI: 10.1007/s12016-017-8630-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
19
|
Generali E, Scirè CA, Favalli EG, Selmi C. Biomarkers in psoriatic arthritis: a systematic literature review. Expert Rev Clin Immunol 2016; 12:651-60. [DOI: 10.1586/1744666x.2016.1147954] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
20
|
Collins ES, Butt AQ, Gibson DS, Dunn MJ, Fearon U, van Kuijk AW, Gerlag DM, Pontifex E, Veale DJ, Tak PP, FitzGerald O, Pennington SR. A clinically based protein discovery strategy to identify potential biomarkers of response to anti-TNF-α treatment of psoriatic arthritis. Proteomics Clin Appl 2015; 10:645-62. [PMID: 26108918 DOI: 10.1002/prca.201500051] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/05/2015] [Accepted: 06/22/2015] [Indexed: 12/19/2022]
Abstract
PURPOSE Psoriatic arthritis (PsA) can be treated using biologic therapies targeting biomolecules such as tumor necrosis factor alpha, interleukins (IL)-17 and IL-23. Although 70% PsA patients respond well to therapy, 30% patients show no or limited clinical improvement. Biomarkers that predict response to therapy would help to avoid unnecessary use of expensive biologics in nonresponding patients and enable alternative treatments to be explored. EXPERIMENTAL DESIGN Patient synovial tissue samples from two clinical studies were analysed using difference in-gel electrophoresis-based proteomics to identify protein expression differences in response to anti-TNF-α treatment. Subsequent multiplexed MRM measurements were used to verify potential biomarkers. RESULTS A total of 119 proteins were differentially expressed (p<0.05) in response to anti-TNF-α treatment and 25 proteins were differentially expressed (p<0.05) between "good responders" and "poor responders". From these differentially expressed proteins, MRM assays were developed for four proteins to explore their potential as treatment predictive biomarkers. CONCLUSION AND CLINICAL RELEVANCE Gel-based proteomics strategy has demonstrated differential protein expression in synovial tissue of PsA patients, in response to anti-TNF-α treatment. Development of multiplex MRM assays to these differentially expressed proteins has the potential to predict response to therapy and allow alternative, more effective treatments to be explored sooner.
Collapse
Affiliation(s)
- Emily S Collins
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland.,Department of Rheumatology, St Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Aisha Q Butt
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - David S Gibson
- Northern Ireland Centre for Stratified Medicine, University of Ulster, C-TRIC, Londonderry, UK
| | - Michael J Dunn
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Ursula Fearon
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland.,Department of Rheumatology, St Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Arno W van Kuijk
- Department of Clinical Immunology and Rheumatology, F4-105, Academic Medical Centre/University of Amsterdam, Amsterdam, The Netherlands
| | - Danielle M Gerlag
- Department of Clinical Immunology and Rheumatology, F4-105, Academic Medical Centre/University of Amsterdam, Amsterdam, The Netherlands
| | - Eliza Pontifex
- Department of Rheumatology, St Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Douglas J Veale
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland.,Department of Rheumatology, St Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Paul P Tak
- Department of Clinical Immunology and Rheumatology, F4-105, Academic Medical Centre/University of Amsterdam, Amsterdam, The Netherlands
| | - Oliver FitzGerald
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland.,Department of Rheumatology, St Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Stephen R Pennington
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
21
|
Jadon DR, Nightingale AL, McHugh NJ, Lindsay MA, Korendowych E, Sengupta R. Serum soluble bone turnover biomarkers in psoriatic arthritis and psoriatic spondyloarthropathy. J Rheumatol 2014; 42:21-30. [PMID: 25362660 DOI: 10.3899/jrheum.140223] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Because psoriatic arthritis (PsA) is an inflammatory disease of joints, serum soluble biomarkers specific for chronic joint and bone inflammation may predict future disease severity and response to therapy, thereby informing stratified medicine approaches. The objectives of our systematic review were to determine whether serum soluble bone and cartilage turnover biomarkers are (1) associated with PsA or psoriatic spondyloarthropathy; and (2) associated with disease activity, disease severity, or clinical phenotype. Ten studies met eligibility criteria. Matrix metalloproteinase (MMP)-3, Dickkopf (DKK)-1, macrophage colony-stimulating factor (M-CSF), crosslinked telopeptide of collagen-1, and tumor necrosis factor-related apoptosis-inducing ligand were associated with PsA, with equivocal results for osteoprotegerin (OPG) and bone alkaline phosphatase (ALP). MMP-3, DKK-1, M-CSF, CPII:C2C (ratio of cartilage degradation vs byproduct formation), and possibly OPG were associated with PsA independently of psoriasis. C1-2C (a neoepitope released when type 2 cartilage is degraded by collagenases) was associated with both tender and swollen joint counts, and bone morphogenetic protein-4 with patient global assessment of disease, pain score, and the Bath Ankylosing Spondylitis Disease Activity Index. Bone ALP was associated with disease activity. M-CSF and receptor activator of nuclear factor-κB ligand were associated with several plain radiographic features. No studies have investigated biomarker associations specifically with axial PsA.
Collapse
Affiliation(s)
- Deepak R Jadon
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath.
| | - Alison L Nightingale
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| | - Neil J McHugh
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| | - Mark A Lindsay
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| | - Eleanor Korendowych
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| | - Raj Sengupta
- From the Royal National Hospital for Rheumatic Diseases, and the University of Bath, Bath, UK.D.R. Jadon, MRCP, Research Fellow, Rheumatology; E. Korendowych, FRCP, Consultant Rheumatologist; R. Sengupta, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases; A.L. Nightingale, PhD, Research Fellow; M.A. Lindsay, PhD, Professor, Pharmacy and Pharmacology, University of Bath; N.J. McHugh, FRCP, Consultant Rheumatologist, Royal National Hospital for Rheumatic Diseases, and the University of Bath
| |
Collapse
|
22
|
Siebuhr AS, He Y, Gudmann NS, Gram A, Kjelgaard-Petersen CF, Qvist P, Karsdal MA, Bay-Jensen AC. Biomarkers of cartilage and surrounding joint tissue. Biomark Med 2014; 8:713-31. [DOI: 10.2217/bmm.13.144] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The identification and clinical demonstration of efficacy and safety of osteo- and chondro-protective drugs are met with certain difficulties. During the last few decades, the pharmaceutical industry has, in the field of rheumatology, experienced disappointments associated with the development of disease modification. Today, the vast amount of patients suffering from serious, chronic joint diseases can only be offered treatments aimed at improving symptoms, such as pain and acute inflammation, and are not aimed at protecting the joint tissue. This huge, unmet medical need has been the driver behind the development of improved analytical techniques allowing better and more efficient clinical trial design, implementation and analysis. With this review, we aim to provide a brief and general overview of biochemical markers of joint tissue, with special focus on neoepitopes. Furthermore, we highlight recent studies applying biochemical markers in joint degenerative diseases. These disorders, including osteoarthritis, rheumatoid arthritis and spondyloarthropathies, are the most predominant disorders in Europe and the USA, and have enormous socioeconomical impact.
Collapse
Affiliation(s)
- Anne S Siebuhr
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Yi He
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Natasja S Gudmann
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Aurelie Gram
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | | | - Per Qvist
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Morten A Karsdal
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| | - Anne C Bay-Jensen
- Nordic Bioscience, Biomarkers & Research, Herlev Hovedgade 207, Herlev DK-2730, Denmark
| |
Collapse
|
23
|
Yeremenko N, Härle P, Cantaert T, van Tok M, van Duivenvoorde LM, Bosserhoff A, Baeten D. The cartilage protein melanoma inhibitory activity contributes to inflammatory arthritis. Rheumatology (Oxford) 2013; 53:438-47. [PMID: 24287514 DOI: 10.1093/rheumatology/ket382] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Melanoma inhibitory activity (MIA) is a small chondrocyte-specific protein with unknown function. MIA knockout mice (MIA(-/-)) have a normal phenotype with minor microarchitectural alterations of cartilage. Our previous study demonstrated that immunodominant epitopes of MIA are actively presented in an HLA-DR4-restricted manner in the inflamed RA joint. The objective of this study was to investigate the potential role of MIA as an autoantigen. METHODS Collagen-induced arthritis (CIA) and anti-collagen antibody-induced arthritis (CAIA) were induced in MIA(-/-) mice. Anti-type II collagen (anti-CII) antibodies were measured by ELISA. T cell proliferation and cytokine production were assessed by flow cytometry. RESULTS MIA(-/-) mice had a markedly reduced incidence and severity of CIA and CAIA compared with wild-type (WT) mice. Attenuation of disease was not related to defective binding of anti-CII antibodies to cartilage in the absence of MIA. However, MIA(-/-) mice had significantly reduced anti-CII IgG1 and IgG2a antibody levels accompanied by an increase in FoxP3-expressing CD25(+)CD4(+) regulatory T cells. This was paralleled by a significant reduction in CII-specific IFN-γ production by T cells in MIA(-/-) but not WT animals, suggesting a qualitative impact of MIA on the collagen-induced Th1 response. Furthermore, Ag-specific proliferation of T cells after restimulation with MIA in WT but not MIA(-/-) mice indicated the existence of MIA-specific T cells in the context of CIA. CONCLUSION These data support a role for MIA as an autoantigen during arthritis development. Whether MIA can influence the balance of pathogenic vs regulatory responses in human RA remains to be investigated.
Collapse
Affiliation(s)
- Nataliya Yeremenko
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
24
|
Ramonda R, Modesti V, Ortolan A, Scanu A, Bassi N, Oliviero F, Punzi L. Serological markers in psoriatic arthritis: promising tools. Exp Biol Med (Maywood) 2013; 238:1431-6. [PMID: 24146263 DOI: 10.1177/1535370213506435] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to identify specific biomarkers that could be used to screen for psoriatic arthritis (PsA), as well as to assess disease activity and treatment outcome in affected patients. Forty-three outpatients considered eligible for anti-TNF-α treatment (etanercept 50 mg/week) were enrolled. Serum samples of vascular endothelial growth factor (VEGF), metalloproteinase-3 (MMP3), pentraxin 3 (PTX3), and high-sensitive C-reactive protein (hs-CRP) were collected at baseline (t0) and after 6 (t6), 12 (t12), and 24 months (t24) of treatment. Baseline values were compared with those of a group of healthy controls matched for age and sex. Disease activity scores and functional tests (DAS28, BASDAI, PASI, BASFI, HAQ, VAS pain, and VAS patient global disease activity) after treatment were found to be significantly different from baseline values. At baseline, MMP3, hs-CRP and VEGF values in the PsA-patients were found to be significantly higher with respect to levels in the controls. There were no differences in the PTX3 values. MMP3 was significantly lower at t6 (P < 0.0001), t12 (P < 0.0001) and t24 (P < 0.0001). hs-CRP and VEGF were significantly lower, respectively, at t12 (P < 0.01; P < 0.05) and t24 (P < 0.05; P < 0.01). PTX3 was significantly higher at t24 (P < 0.05). A correlation was found between MMP3 and hs-CRP (r = 0.45, P = 0.0005). MMP3, hs-CRP, and VEGF appear to be useful for the early detection of PsA and to monitor disease progression. The rise in PTX3 did not appear to be linked to the inflammatory state of the disease but might be an expression of the atherosclerotic process frequently observed in PsA.
Collapse
Affiliation(s)
- Roberta Ramonda
- Rheumatology Unit, Department of Medicine DIMED, University of Padova, 35128 Padova, Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Chandran V, Shen H, Pollock RA, Pellett FJ, Carty A, Cook RJ, Gladman DD. Soluble Biomarkers Associated with Response to Treatment with Tumor Necrosis Factor Inhibitors in Psoriatic Arthritis. J Rheumatol 2013; 40:866-71. [DOI: 10.3899/jrheum.121162] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Objective.To identify soluble biomarkers associated with response to therapy with tumor necrosis factor inhibitors (TNFi) in patients with psoriatic arthritis (PsA).Methods.The study was conducted at a PsA clinic where patients are assessed every 6 months, and serum samples are collected and stored once a year at the time of clinical assessment. Forty patients with active PsA who gave serum samples prior to treatment with TNFi and after at least 3 months of therapy were identified. Patients were classified as TNFi responders if tender joint count was < 3, swollen joint count was 0, and Psoriasis Area and Severity Index score was < 4 at the time the second sample was collected. The following biomarkers were tested by ELISA: TNF superfamily 14, matrix metalloprotease-3 (MMP-3), receptor activator of nuclear factor kappa-B ligand, osteoprotegerin, cartilage oligomeric matrix protein (COMP), CPII, C2C and C1-2C, CS-846, and highly sensitive C-reactive protein. Paired t tests and logistic regression was used for statistical analyses.Results.After a mean treatment duration of 11 months with TNFi (etanercept 28 patients, adalimumab 6, golimumab 4, infliximab 2), 29 patients were classified as TNFi responders. Baseline level of MMP-3 was independently associated with responder status (OR 1.067 for each 1-unit increase, p = 0.045). A reduction in MMP-3 levels with therapy increased the odds of achieving response (OR 1.213 for each 1-unit change, p = 0.030), whereas a reduction in COMP decreased the odds (OR 0.587, for each 100-unit increase, p = 0.039). None of the other biomarkers was associated with response.Conclusion.Baseline as well as reduction in serum MMP-3 and increase in serum COMP are independently associated with response to TNFi therapy in patients with PsA.
Collapse
|
26
|
Szentpetery A, McKenna MJ, Murray BF, Ng CT, Brady JJ, Morrin M, Radovits B, Veale DJ, Fitzgerald O. Periarticular bone gain at proximal interphalangeal joints and changes in bone turnover markers in response to tumor necrosis factor inhibitors in rheumatoid and psoriatic arthritis. J Rheumatol 2013; 40:653-62. [PMID: 23457381 DOI: 10.3899/jrheum.120397] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) and psoriatic arthritis (PsA) are characterized by periarticular bone erosion; periarticular bone formation is a feature in PsA. The effect of anti-tumor necrosis factor-α (TNF-α) on periarticular bone remodeling is unclear in both diseases. Our aim was to assess the response of bone turnover markers (BTM) and hand bone mineral density (BMD) to anti-TNF over 3 years in RA and PsA. METHODS We measured serum bone-specific alkaline phosphatase (bone ALP), procollagen type-I N-propeptide (PINP), intact osteocalcin, C-terminal cross-linking telopeptides (CTX-I), urinary N-terminal cross-linking telopeptide of type-I collagen (NTX-I), and free deoxypyridinoline crosslinks (fDPD) at baseline, 1, 12, and 36 months. BMD measurements (hands/spine/hip) were obtained at 3 timepoints. RESULTS We recruited 62 patients (RA 35; PsA 27). BTM correlated significantly with hand BMD but not with central BMD. Low hand BMD was associated with RA and increased BTM. Following anti-TNF therapy, hip BMD declined while spine and hand BMD were unchanged. Periarticular BMD at proximal interphalangeal (PIP) joints increased while it decreased at metacarpophalangeal joints. Bone ALP increased steadily and was always higher in PsA. PINP and intact osteocalcin increased to a lesser extent, but resorption markers did not change. CONCLUSION At baseline, hand BMD was inversely associated with BTM. Bone formation rather than resorption markers better showed the bone response to anti-TNF. Despite a lack of effect on central BMD, the modest effect of anti-TNF on PIP BMD may provide evidence that BTM reflect specifically bone remodeling activity at periarticular sites of inflammation in RA and PsA.
Collapse
Affiliation(s)
- Agnes Szentpetery
- Department of Rheumatology, the Metabolism Laboratory, St. Vincent's University Hospital, Dublin, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lories RJ, de Vlam K. Is psoriatic arthritis a result of abnormalities in acquired or innate immunity? Curr Rheumatol Rep 2012; 14:375-82. [PMID: 22527951 DOI: 10.1007/s11926-012-0257-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Psoriatic arthritis is a common chronic inflammatory joint disease in which both inflammation and tissue damage contribute to the patient's outcome. Abnormal activation of the innate and the adaptive immune system contributes to the chronic disease process. Novel insights into these immune pathways are further corroborated by genetic evidence. In this review, we compare the current paradigm of psoriasis to mechanisms that likely play a role in psoriatic arthritis and provide an overview of the role of immune mechanisms in the different features of the disease.
Collapse
Affiliation(s)
- Rik J Lories
- Laboratory for Skeletal Development and Joint Disorders, Department of Development and Regeneration, KU Leuven, Herestraat 49, 300 Leuven, Belgium.
| | | |
Collapse
|
28
|
Wagner CL, Visvanathan S, Elashoff M, McInnes IB, Mease PJ, Krueger GG, Murphy FT, Papp K, Gomez-Reino JJ, Mack M, Beutler A, Gladman D, Kavanaugh A. Markers of inflammation and bone remodelling associated with improvement in clinical response measures in psoriatic arthritis patients treated with golimumab. Ann Rheum Dis 2012; 72:83-8. [PMID: 22975755 PMCID: PMC3551220 DOI: 10.1136/annrheumdis-2012-201697] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Objective To determine serum biomarker associations with clinical response to golimumab treatment in patients with psoriatic arthritis (PsA). Methods GO–REVEAL was a randomised, placebo-controlled study of golimumab in patients with active PsA. Samples were collected from 100 patients at baseline, week 4 and week 14, and analysed for serum-based biomarkers and protein profiling (total 92 markers); data were correlated with clinical measures at week 14. Results Serum levels of a subset of proteins (apolipoprotein C III, ENRAGE, IL-16, myeloperoxidase, vascular endothelial growth factor, pyridinoline, matrix metalloproteinase 3, C-reactive protein (CRP), carcinoembryonic antigen, intercellular adhesion molecule 1 and macrophage inflammatory protein 1α) at baseline or week 4 were strongly associated with American College of Rheumatology 20% improvement (ACR20) response and/or disease activity score in 28 joints (DAS28) at week 14. A smaller subset of proteins was significantly associated with a 75% improvement in the psoriasis area and severity index score (PASI75) at week 14, (adiponectin, apolipoprotein CIII, serum glutamic oxaloacetic transaminase, and tumour necrosis factor α). Subsets of proteins were identified as potentially predictive of clinical response for each of the clinical measures, and the power of these biomarker panels to predict clinical response to golimumab treatment was stronger than for CRP alone. Conclusions This analysis provides insight into several panels of markers that may have utility in identifying PsA patients likely to have ACR20, DAS28, or PASI75 responses following golimumab treatment.
Collapse
Affiliation(s)
- Carrie L Wagner
- Department of Immunology, Janssen Research & Development, LLC, Spring House, PA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
FITZGERALD OLIVER, CHANDRAN VINOD. Update on Biomarkers in Psoriatic Arthritis: A Report from the GRAPPA 2010 Annual Meeting: Table 1. J Rheumatol 2012; 39:427-30. [DOI: 10.3899/jrheum.111241] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Biomarkers may be used to screen patients with psoriasis for psoriatic arthritis (PsA) and to assess disease activity and severity. Candidate biomarkers should fulfil the key features of the OMERACT (Outcome Measures in Rheumatology) filter, that is, truth, discrimination, and feasibility. A number of biomarkers are currently being investigated in psoriatic disease for important clinical outcomes. Serum high sensitivity C-reactive protein, cartilage oligomeric matrix protein, interleukin 6 (IL-6), osteoprotegerin, matrix metalloprotease-3 (MMP-3), and the ratio of C-propeptide of type II collagen (CPII) to collagen fragment neoepitopes Col2–3/4 (long mono) (C2C) show promise as serum biomarkers that distinguish subjects with PsA from those with psoriasis alone. Serum MMP-3 and melanoma inhibitory activity, synovial fluid IL-1, IL-1 receptor-α, IL-6, IL-8, and chemokine CCL3 and synovial tissue CD3-positive T cells may prove useful as biomarkers of PsA activity. Circulating osteoclast precursors, Dickkopf-1, macrophage colony stimulating factor, receptor activator of nuclear factor-κB ligand, and bone alkaline phosphatase are strong candidates as biomarkers of radiographic change. Prospective studies to identify biomarkers for psoriatic disease are high on the research agenda of the Group for Research and Assessment of Psoriasis and PsA.
Collapse
|
30
|
Connolly M, Mullan RH, McCormick J, Matthews C, Sullivan O, Kennedy A, FitzGerald O, Poole AR, Bresnihan B, Veale DJ, Fearon U. Acute-phase serum amyloid A regulates tumor necrosis factor α and matrix turnover and predicts disease progression in patients with inflammatory arthritis before and after biologic therapy. ACTA ACUST UNITED AC 2011; 64:1035-45. [PMID: 22076945 DOI: 10.1002/art.33455] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To investigate the relationship between acute-phase serum amyloid A (A-SAA) and joint destruction in inflammatory arthritis. METHODS Serum A-SAA and C-reactive protein (CRP) levels, the erythrocyte sedimentation rate (ESR), and levels of matrix metalloproteinase 1 (MMP-1), MMP-2, MMP-3, MMP-9, MMP-13, tissue inhibitor of metalloproteinases 1 (TIMP-1), vascular endothelial growth factor (VEGF), and type I and type II collagen-generated biomarkers C2C and C1,2C were measured at 0-3 months in patients with inflammatory arthritis commencing anti-tumor necrosis factor α (anti-TNFα) therapy and were correlated with 1-year radiographic progression. The effects of A-SAA on MMP/TIMP expression on RA fibroblast-like synoviocytes (FLS), primary human chondrocytes, and RA/psoriatic arthritis synovial explant cultures were assessed using real-time polymerase chain reaction, enzyme-linked immunosorbent assay, antibody protein arrays, and gelatin zymography. RESULTS Serum A-SAA levels were significantly (P < 0.05) correlated with MMP-3, the MMP-3:TIMP-1 ratio, C1,2C, C2C, and VEGF. The baseline A-SAA level but not the ESR or the CRP level correlated with the 28-joint swollen joint count and was independently associated with 1-year radiographic progression (P = 0.038). A-SAA increased MMP-1, MMP-3, MMP-13, and MMP/TIMP expression in RA FLS and synovial explants (P < 0.05). In chondrocytes, A-SAA induced MMP-1, MMP-3, and MMP-13 messenger RNA and protein expression (all P < 0.01), resulting in a significant shift in MMP:TIMP ratios (P < 0.05). Gelatin zymography revealed that A-SAA induced MMP-2 and MMP-9 activity. Blockade of the A-SAA receptor SR-B1 (A-SAA receptor scavenger receptor-class B type 1) inhibited MMP-3, MMP-2, and MMP-9 expression in synovial explant cultures ex vivo. Importantly, we demonstrated that A-SAA has the ability to induce TNFα expression in RA synovial explant cultures (P < 0.05). CONCLUSION A-SAA may be involved in joint destruction though MMP induction and collagen cleavage in vivo. The ability of A-SAA to regulate TNFα suggests that A-SAA signaling pathways may provide new therapeutic strategies for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Mary Connolly
- St. Vincent's University Hospital, Dublin Academic Medical Centre, The Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Avgerinou G, Tousoulis D, Siasos G, Oikonomou E, Maniatis K, Papageorgiou N, Paraskevopoulos T, Miliou A, Koumaki D, Latsios G, Therianiou A, Trikas A, Kampoli AM, Stefanadis C. Anti-tumor necrosis factor α treatment with adalimumab improves significantly endothelial function and decreases inflammatory process in patients with chronic psoriasis. Int J Cardiol 2011; 151:382-3. [PMID: 21764467 DOI: 10.1016/j.ijcard.2011.06.112] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 06/25/2011] [Indexed: 01/08/2023]
|
32
|
Catterall JB, Stabler TV, Flannery CR, Kraus VB. Changes in serum and synovial fluid biomarkers after acute injury (NCT00332254). Arthritis Res Ther 2010; 12:R229. [PMID: 21194441 PMCID: PMC3046542 DOI: 10.1186/ar3216] [Citation(s) in RCA: 190] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 09/01/2010] [Accepted: 12/31/2010] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Acute trauma involving the anterior cruciate ligament is believed to be a major risk factor for the development of post-traumatic osteoarthritis 10 to 20 years post-injury. In this study, to better understand the early biological changes which occur after acute injury, we investigated synovial fluid and serum biomarkers. METHODS We collected serum from 11 patients without pre-existing osteoarthritis from a pilot intervention trial (5 placebo and 6 drug treated) using an intra-articular interleukin-1 receptor antagonist (IL-1Ra) therapy, 9 of which also supplied matched synovial fluid samples at presentation to the clinic after acute knee injury (mean 15.2 ± 7.2 days) and at the follow-up visit for reconstructive surgery (mean 47.6 ± 12.4 days). To exclude patients with pre-existing osteoarthritis (OA), the study was limited to individuals younger than 40 years of age (mean 23 ± 3.5) with no prior history of joint symptoms or trauma. We profiled a total of 21 biomarkers; 20 biomarkers in synovial fluid and 13 in serum with 12 biomarkers measured in both fluids. Biomarkers analyzed in this study were found to be independent of treatment (P > 0.05) as measured by Mann-Whitney and two-way ANOVA. RESULTS We observed significant decreases in synovial fluid (sf) biomarker concentrations from baseline to follow-up for (sf)C-Reactive protein (CRP) (P = 0.039), (sf)lubricin (P = 0.008) and the proteoglycan biomarkers: (sf)Glycosaminoglycan (GAG) (P = 0.019), and (sf)Alanine-Arginine-Glycine-Serine (ARGS) aggrecan (P = 0.004). In contrast, we observed significant increases in the collagen biomarkers: (sf)C-terminal crosslinked telopeptide type II collagen (CTxII) (P = 0.012), (sf)C1,2C (P = 0.039), (sf)C-terminal crosslinked telopeptide type I collagen (CTxI) (P = 0.004), and (sf)N-terminal telopeptides of type I collagen (NTx) (P = 0.008). The concentrations of seven biomarkers were significantly higher in synovial fluid than serum suggesting release from the signal knee: IL-1β (P < 0.0001), fetal aggrecan FA846 (P = 0.0001), CTxI (P = 0.0002), NTx (P = 0.012), osteocalcin (P = 0.012), Cartilage oligomeric matrix protein (COMP) (P = 0.0001) and matrix metalloproteinase (MMP)-3 (P = 0.0001). For these seven biomarkers we found significant correlations between the serum and synovial fluid concentrations for only CTxI (P = 0.0002), NTx (P < 0.0001), osteocalcin (P = 0.0002) and MMP-3 (P = 0.038). CONCLUSIONS These data strongly suggest that the biology after acute injury reflects that seen in cartilage explant models stimulated with pro-inflammatory cytokines, which are characterized by an initial wave of proteoglycan loss followed by subsequent collagen loss. As the rise of collagen biomarkers in synovial fluid occurs within the first month after injury, and as collagen loss is thought to be irreversible, very early treatment with agents to either reduce inflammation and/or reduce collagen loss may have the potential to reduce the onset of future post-traumatic osteoarthritis. TRIAL REGISTRATION The samples used in this study were derived from a clinical trial NCT00332254 registered with ClinicalTrial.gov.
Collapse
Affiliation(s)
- Jonathan B Catterall
- Division of Rheumatology, Department of Medicine, Duke University, Genome Science Research Building I, 905 South LaSalle Street, Durham, NC 27710, USA
| | - Thomas V Stabler
- Division of Rheumatology, Department of Medicine, Duke University, Genome Science Research Building I, 905 South LaSalle Street, Durham, NC 27710, USA
| | - Carl R Flannery
- Tissue Repair Research Unit, BioTherapeutics Division, Pfizer Inc., 200 Cambridge Park Drive, Cambridge, MA 02140, USA
| | - Virginia B Kraus
- Division of Rheumatology, Department of Medicine, Duke University, Genome Science Research Building I, 905 South LaSalle Street, Durham, NC 27710, USA
| |
Collapse
|