1
|
Marvel J, Gargon E, Howse C, Chohan A, Mayhew M, Kenney G, Stone L, Fisher BA, Steenackers M, Williamson N, Perella C, Goswami P. The Development and Content Validation of the Sjögren's Related Quality of Life Instrument (SRQoL). Rheumatol Ther 2024; 11:1591-1609. [PMID: 39388050 DOI: 10.1007/s40744-024-00718-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
INTRODUCTION Several clinical outcome assessment (COA) instruments assess Sjögren's disease (Sjögren's) symptoms, but do not provide comprehensive assessment of the health-related quality of life (HRQoL) impact of Sjögren's. This study aimed to develop a patient-reported outcome (PRO) instrument for the assessment of HRQoL, intended for use in clinical trials and clinical practice in the assessment of treatment benefit. METHODS Review of study sponsor proprietary data and qualitative interviews informed the development of a conceptual model, the Sjögren's Related Quality of Life (SRQoL) and patient global impression of severity (PGI-S) and change (PGI-C) items. Combined concept elicitation and cognitive debriefing interviews with patients with Sjögren's explored their HRQoL impact experience and content validity of the SRQoL and PGI items. RESULTS Twenty participants were interviewed about their Sjögren's experience. Following inductive analysis of interviews, concepts were categorized into eight domains: emotional well-being (e.g., worry and stress; n = 20/20; 100%), sleep (e.g., daytime sleepiness and waking up during the night; n = 20/20; 100%), activities of daily living (e.g., difficulty looking at screens and difficulty driving; n = 20/20; 100%), cognition (e.g., concentration difficulties and word finding difficulties; n = 19/20; 95.0%), physical functioning (e.g., difficulty walking and difficulty exercising; n = 19/20; 95.0%), social and family functioning (e.g., dependent on others and relationship difficulties; n = 17/20; 85.0%), work (n = 15/20; 75.0%), and sexual functioning (n = 12/20; 60.0%). SRQoL and PGI items, instructions, response options, and recall period were well understood and relevant to participants. CONCLUSIONS The SRQoL is a new PRO instrument to assess Sjögren's impact on HRQoL, developed in accordance with regulatory guidance. This study provides considerable insight into the patient experience of Sjögren's and evidence to support the content validity of the SRQoL. Future research should evaluate the psychometric properties of the SRQoL to support its use in clinical trials and clinical practice and further validate its use as an assessment of treatment benefit.
Collapse
Affiliation(s)
| | | | - Chloe Howse
- Patient-Centered Outcomes, Adelphi Values Ltd., Bollington, UK
| | | | - Megan Mayhew
- Patient-Centered Outcomes, Adelphi Values Ltd., Bollington, UK
| | | | - Linda Stone
- Sjögren's UK (formerly British Sjögrens Syndrome Association), Birmingham, UK
| | - Benjamin A Fisher
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR), Birmingham Biomedical Research Centre and Department of Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | | | | |
Collapse
|
2
|
Feng R, Xiao X, Wang Y, Huang B, Chen J, Cheng G, Jin Y. Metabolic impact of low dose IL-2 therapy for primary Sjögren's Syndrome in a double-blind, randomized clinical trial. Clin Rheumatol 2024:10.1007/s10067-024-07165-2. [PMID: 39482484 DOI: 10.1007/s10067-024-07165-2] [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: 09/11/2024] [Revised: 09/22/2024] [Accepted: 09/26/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVES Low-dose interleukin 2 (Ld-IL2) is increasingly being explored as an immune-modulating treatment for autoimmune diseases which mainly affect T cell subsets. This study investigates the metabolic effects of Ld-IL2 therapy in patients with primary Sjögren's syndrome (pSS). METHOD A total of 60 patients were recruited to conduct a double-blind, randomized clinical trial. Of these patients, 50% (30/60) received Ld-IL2 therapy along with standard treatment for 12 weeks, followed by 12 weeks of follow-up. The effectiveness was evaluated by Sjögren's Tool for Assessing Response (STAR). An untargeted analysis was performed to profile hydrophilic metabolites. RESULTS Metabolic profiling revealed significant alterations post-treatment, notably in metabolites like acetyl-CoA, ascorbic acid, and glutathione, which are beneficial in managing autoimmune diseases. In addition, the levels of metabolite accumulation were correlated with variations in immune cell subsets (p < 0.05), particularly Tregs. Moreover, patients exhibiting a specific metabolic profile, including lower serum levels of isoleucine, ADP, Thymidine 5'-triphosphate, and other metabolites, had a high response rate (91.7%-98.6%), as indicated by the receiver operating characteristic (ROC) curve. CONCLUSIONS These findings suggest that Ld-IL2 therapy influences metabolic pathways in pSS, offering insights into the systemic effects of Ld-IL2 therapy beyond immune modulation. TRIAL REGISTRATION NUMBER ClinicalTrials.gov number, NCT02464319. Key Points • Metabolic alteration in pSS is significantly associated with Ld-IL2 therapy. • Metabolic changes correlate with variations in immune cell subsets, particularly Tregs. • Metabolic profiling could be a valuable tool in guiding Ld-IL2 therapy choices for pSS patients.
Collapse
Affiliation(s)
- Ruiling Feng
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Peking University People's Hospital, 11 Xizhimen South St., Beijing, 100044, China
| | - Xian Xiao
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Peking University People's Hospital, 11 Xizhimen South St., Beijing, 100044, China
| | - Yifan Wang
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Peking University People's Hospital, 11 Xizhimen South St., Beijing, 100044, China
| | - Bo Huang
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Peking University People's Hospital, 11 Xizhimen South St., Beijing, 100044, China
| | - Jiali Chen
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Peking University People's Hospital, 11 Xizhimen South St., Beijing, 100044, China
| | - Gong Cheng
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Peking University People's Hospital, 11 Xizhimen South St., Beijing, 100044, China
| | - Yuebo Jin
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Peking University People's Hospital, 11 Xizhimen South St., Beijing, 100044, China.
| |
Collapse
|
3
|
Georgel L, Benyoussef AA, Berrouiguet S, Guellec D, Carvajal Alegria G, Marhadour T, Jousse-Joulin S, Cochener-Lamard B, Labetoulle M, Gottenberg JE, Bourcier T, Nocturne G, Saraux A, Mariette X, Consigny M, Gravey M, Devauchelle-Pensec V, Seror R, Cornec D. Development of a web-based ecological momentary assessment tool to measure day-to-day variability of the symptoms in patients with Sjögren's disease. RMD Open 2024; 10:e004526. [PMID: 39357926 PMCID: PMC11448184 DOI: 10.1136/rmdopen-2024-004526] [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: 05/13/2024] [Accepted: 07/27/2024] [Indexed: 10/04/2024] Open
Abstract
OBJECTIVES To develop and validate a web-based ecological momentary assessment (EMA) tool to enhance symptoms monitoring among patients with Sjögren's disease (SjD). METHODS Consecutive adults with SjD were enrolled in this pilot observational study. Participants used the WebApp over a 3-month period, for the daily collection of individual EULAR Sjögren's Syndrome Patient Reported Index (ESSPRI) scales and separate assessment of eyes and mouth dryness, using 0-10 numerical scales. Primary outcome was the measure of the interdaily variability of symptoms. Data collected through the WebApp were compared with those obtained with paper-based questionnaires administered during a final visit, using distinct approaches (predicted error, maximum negative error and maximum positive error). User experience was assessed using the System Usability Scale (SUS) score. RESULTS Among the 45 participants, 41 (91.1%) were women. Median age was 57 years (IQR: 49-66). Daily variability of symptoms ranged between 0.5 and 0.8 points across the scales. Over the 3-month period, the predicted error ranged between -1.2 and -0.3 points of the numerical scales. The greatest differences were found for fatigue (-1.2 points (IQR: -2.3 to -0.2)) and ESSPRI score (-1.2 points (IQR: -1.7 to -0.3)). Over the last 2 weeks, the predicted error ranged between - 1.2 and 0.0 points. Maximum negative error ranged between -2.0 and -1.0 points, and maximum positive error between -0.3 and 0.0 points. Median SUS score was 90 (IQR: 85-95). CONCLUSION Our results demonstrate the usability and the relevance of our web-based EMA tool for capturing data that closely reflects daily experiences of patients with SjD.
Collapse
Affiliation(s)
- Laurie Georgel
- Rheumatology Department, Centre National de Référence des Maladies Auto-Immunes Rares (CERAINOM), CHU Brest, Brest, France
| | | | | | - Dewi Guellec
- Rheumatology Department, Centre National de Référence des Maladies Auto-Immunes Rares (CERAINOM), CHU Brest, Brest, France
- LBAI, Univ Brest, Inserm, Brest, France, Brest, France
| | | | - Thierry Marhadour
- Rheumatology Department, Centre National de Référence des Maladies Auto-Immunes Rares (CERAINOM), CHU Brest, Brest, France
| | - Sandrine Jousse-Joulin
- Rheumatology Department, Centre National de Référence des Maladies Auto-Immunes Rares (CERAINOM), CHU Brest, Brest, France
- LBAI, Univ Brest, Inserm, Brest, France, Brest, France
| | | | - Marc Labetoulle
- Department of Ophthalmology, Hôpital Bicêtre, Le Kremlin-Bicetre, Paris, France
| | | | | | - Gaétane Nocturne
- Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux universitaires Paris-Sud - Hôpital Bicêtre, Le Kremlin Bicêtre, France
- Université Paris-Sud, Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Alain Saraux
- Rheumatology Department, Centre National de Référence des Maladies Auto-Immunes Rares (CERAINOM), CHU Brest, Brest, France
- LBAI, Univ Brest, Inserm, Brest, France, Brest, France
| | - Xavier Mariette
- Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux universitaires Paris-Sud - Hôpital Bicêtre, Le Kremlin Bicêtre, France
- Université Paris-Sud, Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | | | | | - Valérie Devauchelle-Pensec
- Rheumatology Department, Centre National de Référence des Maladies Auto-Immunes Rares (CERAINOM), CHU Brest, Brest, France
- LBAI, Univ Brest, Inserm, Brest, France, Brest, France
| | - Raphaele Seror
- Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux universitaires Paris-Sud - Hôpital Bicêtre, Le Kremlin Bicêtre, France
- Université Paris-Sud, Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Divi Cornec
- Rheumatology Department, Centre National de Référence des Maladies Auto-Immunes Rares (CERAINOM), CHU Brest, Brest, France
- LBAI, Univ Brest, Inserm, Brest, France, Brest, France
| |
Collapse
|
4
|
Wang Y, Xie X, Han Y, Liu X, Guo Q, Zhang L, Ni X, Liu H. The value of parotid sialography in the diagnosis and staging of Sjogren's syndrome. J Dent Sci 2024; 19:2179-2185. [PMID: 39347041 PMCID: PMC11437596 DOI: 10.1016/j.jds.2024.03.007] [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: 01/29/2024] [Revised: 03/06/2024] [Indexed: 10/01/2024] Open
Abstract
Background/purpose 2016 American College of Rheumatology/European League Against Rheumatism (ACR/EULAR) primary Sjögren's syndrome (SS) diagnostic criteria did not incorporate radiographic examination while staging SS according to salivary gland imaging and serological autoantibody tests was not discussed. The aim is to study the value of parotid sialography for diagnosing SS, and to initially explore the method of staging SS based on the results of imaging and serological autoantibody tests. Materials and methods 287 patients' clinical records were included. The sensitivity and specificity of parotid sialography in the diagnosis of SS were investigated. SS patients were categorized into early stage (autoantibody positive, imaging does not support SS), active stage (autoantibody positive, imaging supports SS), and quiescent stage (autoantibody negative, imaging supports SS), clinical characteristics of different stages were compared. Results The sensitivity of parotid sialography for the diagnosis of SS was 82.6%, the specificity was 71.5%. 10-minute USFR of the patients in the active stage (0.18 ± 0.38 ml/10min) was significantly lower than that of early stage (0.34 ± 0.47 ml/10min) and quiescent stage (0.54 ± 0.52 ml/10min), P = 0.010, and the rate of confirmed SS was significantly higher in the active stage (82.9%) than that in the early stage (44.4%) and the quiescent stages (14.8%), P < 0.001. Conclusion Parotid sialography remains valuable in the diagnosis of SS. Performing imaging and serological autoantibody tests before lip gland biopsy may reduce invasive examinations for patients without significantly increasing the rate of missed diagnosis. According to imaging and serological autoantibody tests, SS can be categorized into early, active, and quiescent stages.
Collapse
Affiliation(s)
- Yutian Wang
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiaoyan Xie
- Department of Oral and Maxillofacial Radiology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ying Han
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xingyun Liu
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| | - Qianyun Guo
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| | - Lei Zhang
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xinyi Ni
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| | - Hongwei Liu
- Department of Oral Medicine, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
5
|
Gandolfo S, Bombardieri M, Pers JO, Mariette X, Ciccia F. Precision medicine in Sjögren's disease. THE LANCET. RHEUMATOLOGY 2024; 6:e636-e647. [PMID: 38723653 DOI: 10.1016/s2665-9913(24)00039-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/16/2024] [Accepted: 02/08/2024] [Indexed: 08/23/2024]
Abstract
Sjögren's disease is a clinically and pathophysiologically heterogeneous disease to which precision medicine, on the basis of clinical and biological heterogeneity, has been not always applicable. In patients with Sjögren's disease, the relationship between dysregulated biological pathways and symptoms such as fatigue and pain or clinical manifestations is often difficult to establish. This clinical and biological dissociation also poses challenges when defining appropriate clinical endpoints for clinical trials. In the last few years, however, research efforts have been focused on gaining a better understanding of the considerable heterogeneity of Sjögren's disease by developing stratification models aimed at clustering patients with this condition into homogenous subgroups characterised by distinctive molecular signatures, biomarkers, clinical features, and outcomes. In this Review, we discuss current evidence regarding clinical, laboratory, histological, and biomolecular stratification in Sjögren's disease and examine how available stratification data can guide precision medicine and inform the design of future clinical trials.
Collapse
Affiliation(s)
- Saviana Gandolfo
- Rheumatology Section, Ospedale San Giovanni Bosco, Naples, Italy.
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Jacques-Olivier Pers
- Lymphocytes B Autoimmunité et Immunothérapies, UMR1227, INSERM, CHU de Brest, University of Brest, Brest, France
| | - Xavier Mariette
- Rheumatology Department, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, Paris, France
| | - Francesco Ciccia
- Rheumatology Section, Department of Precision Medicine, University della Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
6
|
Fisher BA, Mariette X, Papas A, Grader-Beck T, Bootsma H, Ng WF, van Daele PLA, Finzel S, Noaiseh G, Elgueta S, Hermann J, McCoy SS, Akpek E, Bookman A, Sopala M, Montecchi-Palmer M, Luo WL, Scheurer C, Hueber W. Safety and efficacy of subcutaneous iscalimab (CFZ533) in two distinct populations of patients with Sjögren's disease (TWINSS): week 24 results of a randomised, double-blind, placebo-controlled, phase 2b dose-ranging study. Lancet 2024; 404:540-553. [PMID: 39096929 DOI: 10.1016/s0140-6736(24)01211-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/08/2024] [Accepted: 06/06/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Sjögren's disease is a chronic autoimmune disease with an unmet need for targeted therapies. The aim of the TWINSS study is to evaluate the safety and efficacy of iscalimab, a monoclonal antibody against CD40, in patients with active Sjögren's disease. METHODS This randomised, double-blind, placebo-controlled, phase 2b study, conducted at 71 sites in 23 countries, enrolled patients aged 18 years or older fulfilling the American College of Rheumatology/European Alliance of Associations for Rheumatology (EULAR) 2016 criteria. In the dose-ranging cohort 1, patients with a EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI) score of 5 or higher and a EULAR Sjögren's Syndrome Patient Reported Index (ESSPRI) score of 5 or higher were randomly assigned (1:1:1:1) to subcutaneous iscalimab 150 mg, 300 mg, 600 mg, or placebo. In the proof-of-concept cohort 2, patients with an ESSDAI score of less than 5, ESSPRI (dryness or fatigue) score of 5 or higher, and Impact of Dry Eye on Everyday Life score of 30 or higher were randomly assigned (1:1) to iscalimab 600 mg or placebo. The sponsor, investigator, site personnel, and patients were masked to the treatment assignment. The primary objectives were to demonstrate a dose-response relationship of iscalimab based on the change in ESSDAI from baseline to week 24 in cohort 1 by Multiple Comparison Procedure-Modelling (MCP-Mod), and to assess the effect of iscalimab 600 mg on ESSPRI at week 24 in cohort 2. All the efficacy analyses included all patients who were randomly assigned, and safety analysis included all patients who received at least one dose of study drug. This trial is registered with ClinicalTrials.gov (NCT03905525), and is complete. FINDINGS Between Oct 1, 2019, and Feb 28, 2022, 460 patients were screened; 173 patients were assigned to cohort 1 (44 to iscalimab 150 mg, 43 to 300 mg, 43 to 600 mg, and 43 to placebo) and 100 to cohort 2 (50 to each group). In cohort 1, the MCP step showed a significant dose-response relationship for placebo-adjusted ESSDAI change from baseline in one of four models (Linlog model, one-sided p=0·0041). ESSDAI decreased from baseline to week 24 with all three doses of iscalimab; 150 mg and 600 mg doses showed statistically significant improvement (placebo-adjusted least squares [LS] mean difference -3·0 [95% CI -4·9 to -1·1]; p=0·0025 for 150 mg and -2·9 [-4·9 to -1·0]; p=0·0037 for 600 mg). In cohort 2, ESSPRI showed a trend towards improvement with iscalimab 600 mg (placebo-adjusted LS mean change from baseline -0·57 points [95% CI -1·30 to 0·15]; p=0·12). Serious adverse events were reported in nine patients in cohort 1 (one [2%] of 43 in the placebo group, one [2%] of 44 in the iscalimab 150 mg group, three [7%] of 42 in the 300 mg group, four [9%] of 44 in the 600 mg group) and four patients in cohort 2 (two [4%] of 50 in each group). No deaths occurred over the 24-week period. INTERPRETATION The study met the primary objective of demonstrating a significant dose-response relationship with iscalimab in terms of disease activity at week 24. Iscalimab was well tolerated and showed initial clinical benefit over placebo in two distinct populations of patients with Sjögren's disease, to be confirmed in larger trials. FUNDING Novartis Pharma.
Collapse
Affiliation(s)
- Benjamin A Fisher
- Department of Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK; NIHR Birmingham Biomedical Research Centre, Birmingham, UK
| | - Xavier Mariette
- Department of Rheumatology, Université Paris-Saclay, Assistance Publique - Hôpitaux de Paris, Hôpital Bicêtre, INSERM UMR1184, Le Kremlin Bicêtre, France
| | - Athena Papas
- Division of Oral Medicine, Tufts School of Dental Medicine, Boston, MA, USA
| | - Thomas Grader-Beck
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Wan-Fai Ng
- NIHR Newcastle Clinical Research Facility, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK
| | - P L A van Daele
- Department of Internal Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Stephanie Finzel
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ghaith Noaiseh
- Division of Allergy, Clinical Immunology and Rheumatology, Department of Medicine, University of Kansas, Kansas City, KS, USA
| | - Sergio Elgueta
- Department of Rheumatology, Clinica Alemana de Valdivia, Valdivia, Chile; Clinical Research Chile SpA, Biomedical Research Centre, Valdivia, Chile
| | - Josef Hermann
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Sara S McCoy
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Esen Akpek
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | - Wen-Lin Luo
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | |
Collapse
|
7
|
Baldini C, Fulvio G, La Rocca G, Ferro F. Update on the pathophysiology and treatment of primary Sjögren syndrome. Nat Rev Rheumatol 2024; 20:473-491. [PMID: 38982205 DOI: 10.1038/s41584-024-01135-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/11/2024]
Abstract
Sjögren syndrome or Sjögren disease is a chronic form of autoimmune epithelitis characterized by lymphocytic infiltration of the exocrine glands, particularly the salivary and lacrimal glands, leading to progressive glandular dysfunction and subsequent xerostomia and xerophthalmia. Other common manifestations include pain and fatigue, various systemic manifestations and non-Hodgkin's lymphoma. Sjögren syndrome is therefore a complex and disabling disease associated with a reduced quality of life and with considerable long-term damage. Most of the available treatments are merely symptomatic with limited efficacy in both preventing glandular damage and suppressing systemic disease activity. In the past 10 years, great progress has been made in understanding the pathophysiology of Sjögren syndrome, opening new avenues towards a more targeted and individualized therapeutic approach to the disease. Indeed, several randomized controlled trials have just been completed or are poised to commence evaluating the effectiveness of novel drugs targeting both innate and adaptive immune pathways, including pro-inflammatory cytokines, the type I interferon system, B cell activation, B cell and T cell co-stimulation pathway, and ectopic germinal centre formation. Novel clinical trials are also ongoing exploring various targeted approaches (that is, IgG recycling inhibition, nuclease therapy and CAR-T cell therapy) for Sjögren syndrome.
Collapse
Affiliation(s)
- Chiara Baldini
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Giovanni Fulvio
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gaetano La Rocca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Ferro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
8
|
Mariette X. [Sjögren's disease: From pathophysiology to treatment]. Biol Aujourdhui 2024; 218:1-8. [PMID: 39007771 DOI: 10.1051/jbio/2024003] [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: 02/13/2024] [Indexed: 07/16/2024]
Abstract
Sjögren's disease (SjD) is a systemic autoimmune disorder characterized by a triad of key symptoms affecting almost all patients (salivary and lacrimal dryness, pain and fatigue) and extra-glandular systemic involvement affecting one to two-thirds of patients. Over the past decade, knowledge of the epidemiology, classification criteria, assessment of systemic activity and symptoms presented by patients has grown. In addition, advances in understanding the pathophysiology of SjD have enabled a more targeted therapeutic approach. Current management of SjD is based on EULAR treatment guidelines. But since these recommendations, new drugs targeting specific pathophysiological pathways of the disease, and essentially B lymphocyte activation, have shown efficacy in phase 2 trials. In this review, we will summarize the available evidence on systemic therapies, including: 1. advances in outcome assessment, 2. current evidence on targeted disease-modifying therapies and biologic drugs targeting primarily B lymphocytes, 3. an overview of promising drugs being tested in ongoing trials.
Collapse
Affiliation(s)
- Xavier Mariette
- Département de rhumatologie, Université Paris-Saclay, INSERM U1184, Centre d'immunologie des infections virales et des maladies auto-immunes, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, Service de Rhumatologie, 78 rue du Général Leclerc, 94275 Le Kremlin-Bicêtre, France
| |
Collapse
|
9
|
Boudjeniba C, Soret P, Trutschel D, Hamon A, Baloche V, Chassagnol B, Desvaux E, Bichat A, Aussy A, Moingeon P, Lefebvre C, Hubert S, Alarcon-Riquelmé M, Ng WF, Gottenberg JE, Schwikowski B, Bombardieri M, van Roon JAG, Mariette X, Guedj M, Birmele E, Laigle L, Becht E. Consensus gene modules strategy identifies candidate blood-based biomarkers for primary Sjögren's disease. Clin Immunol 2024; 264:110241. [PMID: 38735508 DOI: 10.1016/j.clim.2024.110241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 04/16/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024]
Abstract
Primary Sjögren disease (pSD) is an autoimmune disease characterized by lymphoid infiltration of exocrine glands leading to dryness of the mucosal surfaces and by the production of autoantibodies. The pathophysiology of pSD remains elusive and no treatment with demonstrated efficacy is available yet. To better understand the biology underlying pSD heterogeneity, we aimed at identifying Consensus gene Modules (CMs) that summarize the high-dimensional transcriptomic data of whole blood samples in pSD patients. We performed unsupervised gene classification on four data sets and identified thirteen CMs. We annotated and interpreted each of these CMs as corresponding to cell type abundances or biological functions by using gene set enrichment analyses and transcriptomic profiles of sorted blood cell subsets. Correlation with independently measured cell type abundances by flow cytometry confirmed these annotations. We used these CMs to reconcile previously proposed patient stratifications of pSD. Importantly, we showed that the expression of modules representing lymphocytes and erythrocytes before treatment initiation is associated with response to hydroxychloroquine and leflunomide combination therapy in a clinical trial. These consensus modules will help the identification and translation of blood-based predictive biomarkers for the treatment of pSD.
Collapse
Affiliation(s)
- Cheïma Boudjeniba
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France; Laboratoire MAP5 UMR 8145, Université Paris Cité, Paris, France; Computational Systems Biomedicine Lab, Institut Pasteur, Université Paris Cité, F-75015 Paris, France
| | - Perrine Soret
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Diana Trutschel
- Computational Systems Biomedicine Lab, Institut Pasteur, Université Paris Cité, F-75015 Paris, France
| | | | - Valentin Baloche
- Department of Rheumatology and Clinical Immunology, Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Bastien Chassagnol
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Emiko Desvaux
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Antoine Bichat
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Audrey Aussy
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Philippe Moingeon
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Céline Lefebvre
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Sandra Hubert
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Marta Alarcon-Riquelmé
- GENYO, Centre for Genomics and Oncological Research, Pfizer, University of Granada, Spain
| | - Wan-Fai Ng
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | | | - Benno Schwikowski
- Computational Systems Biomedicine Lab, Institut Pasteur, Université Paris Cité, F-75015 Paris, France
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London, School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Joel A G van Roon
- Department of Rheumatology and Clinical Immunology, Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Xavier Mariette
- Department of Rheumatology, Université Paris-Saclay, INSERM UMR1184, AP-HP, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Mickaël Guedj
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Etienne Birmele
- Institut de Recherche Mathématique Avancée, UMR 7501 Université de Strasbourg et CNRS, Strasbourg, France
| | - Laurence Laigle
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France
| | - Etienne Becht
- Translational Medicine, Servier, Research and Development, Gif-Sur-Yvette, France; Centre de Recherche sur l'Inflammation, INSERM UMRS1149, Paris, France.
| |
Collapse
|
10
|
Pontarini E, Sciacca E, Chowdhury F, Grigoriadou S, Rivellese F, Murray-Brown WJ, Lucchesi D, Fossati-Jimack L, Nerviani A, Jaworska E, Ghirardi GM, Giacomassi C, Emery P, Ng WF, Sutcliffe N, Everett C, Fernandez C, Tappuni A, Seror R, Mariette X, Porcher R, Cavallaro G, Pulvirenti A, Verstappen GM, de Wolff L, Arends S, Bootsma H, Lewis MJ, Pitzalis C, Bowman SJ, Bombardieri M. Serum and Tissue Biomarkers Associated With Composite of Relevant Endpoints for Sjögren Syndrome (CRESS) and Sjögren Tool for Assessing Response (STAR) to B Cell-Targeted Therapy in the Trial of Anti-B Cell Therapy in Patients With Primary Sjögren Syndrome (TRACTISS). Arthritis Rheumatol 2024; 76:763-776. [PMID: 38073013 DOI: 10.1002/art.42772] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/26/2023] [Accepted: 12/04/2023] [Indexed: 02/17/2024]
Abstract
OBJECTIVE This study aimed to identify peripheral and salivary gland (SG) biomarkers of response/resistance to B cell depletion based on the novel concise Composite of Relevant Endpoints for Sjögren Syndrome (cCRESS) and candidate Sjögren Tool for Assessing Response (STAR) composite endpoints. METHODS Longitudinal analysis of peripheral blood and SG biopsies was performed pre- and post-treatment from the Trial of Anti-B Cell Therapy in Patients With Primary Sjögren Syndrome (TRACTISS) combining flow cytometry immunophenotyping, serum cytokines, and SG bulk RNA sequencing. RESULTS Rituximab treatment prevented the worsening of SG inflammation observed in the placebo arm, by inhibiting the accumulation of class-switched memory B cells within the SG. Furthermore, rituximab significantly down-regulated genes involved in immune-cell recruitment, lymphoid organization alongside antigen presentation, and T cell co-stimulatory pathways. In the peripheral compartment, rituximab down-regulated immunoglobulins and auto-antibodies together with pro-inflammatory cytokines and chemokines. Interestingly, patients classified as responders according to STAR displayed significantly higher baseline levels of C-X-C motif chemokine ligand-13 (CXCL13), interleukin (IL)-22, IL-17A, IL-17F, and tumor necrosis factor-α (TNF-α), whereas a longitudinal analysis of serum T cell-related cytokines showed a selective reduction in both STAR and cCRESS responder patients. Conversely, cCRESS response was better associated with biomarkers of SG immunopathology, with cCRESS-responders showing a significant decrease in SG B cell infiltration and reduced expression of transcriptional gene modules related to T cell costimulation, complement activation, and Fcγ-receptor engagement. Finally, cCRESS and STAR response were associated with a significant improvement in SG exocrine function linked to transcriptional evidence of SG epithelial and metabolic restoration. CONCLUSION Rituximab modulates both peripheral and SG inflammation, preventing the deterioration of exocrine function with functional and metabolic restoration of the glandular epithelium. Response assessed by newly developed cCRESS and STAR criteria was associated with differential modulation of peripheral and SG biomarkers, emerging as novel tools for patient stratification.
Collapse
Affiliation(s)
| | | | | | | | - Felice Rivellese
- Queen Mary University of London and Bart's Health NHS Trust, London, UK
| | | | | | | | | | | | | | | | | | - Wan Fai Ng
- Newcastle University and NIHR Newcastle Clinical Research Facility, Newcastle upon Tyne, UK
| | | | | | | | - Anwar Tappuni
- Queen Mary University of London and Bart's Health NHS Trust, London, UK
| | - Raphael Seror
- Université' Paris-Saclay, and AP-HP, Hôpital Bicêtre, Le Kremlin, Bicêtre, France
| | - Xavier Mariette
- Université' Paris-Saclay, and AP-HP, Hôpital Bicêtre, Le Kremlin, Bicêtre, France
| | - Raphael Porcher
- Université Paris Cité, Centre de Recherche Épidémiologie et Statistiques Paris, France
| | | | | | - Gwenny M Verstappen
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Liseth de Wolff
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Suzanne Arends
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Miles J Lewis
- Queen Mary University of London and Bart's Health NHS Trust, London, UK
| | | | - Simon J Bowman
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | |
Collapse
|
11
|
Lu L, Dai T, Zhao Y, Qu H, Sun QA, Xia H, Wang W, Li G. The value of MRI-based radiomics for evaluating early parotid gland injury in primary Sjögren's syndrome. Clin Rheumatol 2024; 43:1675-1682. [PMID: 38538907 DOI: 10.1007/s10067-024-06935-2] [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: 06/27/2023] [Revised: 09/12/2023] [Accepted: 03/10/2024] [Indexed: 04/16/2024]
Abstract
OBJECTIVE This study aimed to evaluate the value of machine learning models (ML) based on MRI radiomics in diagnosing early parotid gland injury in primary Sjögren's syndrome (pSS). METHODS A total of 164 patients (114 in the training cohort and 50 in the testing cohort) with pSS (n=82) or healthy controls (HC) (n=82) were enrolled. Itksnap software was used to perform two-dimensional segmentation of the bilateral parotid glands on T1-weighted (T1WI) and fat-suppressed T2-weighted imaging (fs-T2WI) images. A total of 1548 texture features of the parotid glands were extracted using radiomics software. A radiomics score (Radscore) was constructed and calculated. A t-test was used to compare the Radscore between the two groups. Finally, five machine learning models were trained and tested to identify early pSS parotid injury, and the performance of the machine learning models was evaluated by calculating the acceptance operating curve (ROC) and other parameters. RESULTS The Radscores between the pSS and HC groups showed significant statistical differences (p<0.001). Among the five machine learning models, the Extra Trees Classifier (ETC) model performed high predictive efficacy in identifying early pSS parotid injury, with an AUC of 0.87 in the testing set. CONCLUSION MRI radiomics-based machine learning models can effectively diagnose early parotid gland injury in primary Sjögren's syndrome.
Collapse
Affiliation(s)
- Lu Lu
- Medical College, Yangzhou University, Yangzhou, 255000, China
| | - Tiantian Dai
- Medical College, Yangzhou University, Yangzhou, 255000, China
| | - Yi Zhao
- Department of Radiology, Medical Imaging Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 255000, China
| | - Hang Qu
- Department of Radiology, Medical Imaging Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 255000, China
| | - Qi An Sun
- Department of Radiology, Medical Imaging Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 255000, China
| | - Hongyi Xia
- Department of Radiology, Medical Imaging Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 255000, China
| | - Wei Wang
- Medical College, Yangzhou University, Yangzhou, 255000, China.
- Department of Radiology, Medical Imaging Center, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 255000, China.
| | - Guoqing Li
- Medical College, Yangzhou University, Yangzhou, 255000, China
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 255000, China
| |
Collapse
|
12
|
Price EJ, Benjamin S, Bombardieri M, Bowman S, Carty S, Ciurtin C, Crampton B, Dawson A, Fisher BA, Giles I, Glennon P, Gupta M, Hackett KL, Larkin G, Ng WF, Ramanan AV, Rassam S, Rauz S, Smith G, Sutcliffe N, Tappuni A, Walsh SB. British Society for Rheumatology guideline on management of adult and juvenile onset Sjögren disease. Rheumatology (Oxford) 2024:keae152. [PMID: 38621708 DOI: 10.1093/rheumatology/keae152] [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: 11/06/2023] [Accepted: 03/02/2024] [Indexed: 04/17/2024] Open
Abstract
Sjögren disease (SD) is a chronic, autoimmune disease of unknown aetiology with significant impact on quality of life. Although dryness (sicca) of the eyes and mouth are the classically described features, dryness of other mucosal surfaces and systemic manifestations are common. The key management aim should be to empower the individual to manage their condition-conserving, replacing and stimulating secretions; and preventing damage and suppressing systemic disease activity. This guideline builds on and widens the recommendations developed for the first guideline published in 2017. We have included advice on the management of children and adolescents where appropriate to provide a comprehensive guideline for UK-based rheumatology teams.
Collapse
Affiliation(s)
- Elizabeth J Price
- Department of Rheumatology, Great Western Hospital NHS Foundation Trust, Swindon, UK
| | - Stuart Benjamin
- The Academy Library and Information Service, Great Western Hospital NHS Foundation Trust, Swindon, UK
| | - Michele Bombardieri
- Department of Rheumatology, Barts and The London School of Medicine and Dentistry, Barts Health NHS Trust, London, UK
- Centre for Experimental Medicine and Rheumatology, The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Simon Bowman
- Department of Rheumatology, Milton Keynes University Hospital, Milton Keynes, UK
- Department of Rheumatology, University Hospitals Birmingham NHSFT, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Sara Carty
- Department of Rheumatology, Great Western Hospital NHS Foundation Trust, Swindon, UK
| | - Coziana Ciurtin
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Bridget Crampton
- Patient Representative, Sjogren's UK Helpline Lead, Sjogren's UK (British Sjögren's Syndrome Association), Birmingham, UK
| | - Annabel Dawson
- Patient Representative, Sjogren's UK (British Sjögren's Syndrome Association), Birmingham, UK
| | - Benjamin A Fisher
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and Department of Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Ian Giles
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Peter Glennon
- General Practice, NHS Staffordshire & Stoke on Trent ICB, Stafford, UK
| | - Monica Gupta
- Department of Rheumatology, Gartnavel General Hospital, Glasgow, UK
| | - Katie L Hackett
- Department of Social Work, Education and Community Wellbeing, Northumbria University, Newcastle upon Tyne, UK
| | | | - Wan-Fai Ng
- Translational and Clinical Research Institute & Newcastle NIHR Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK
- Department of Rheumatology, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Athimalaipet V Ramanan
- Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, Bristol, UK
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Saad Rassam
- Haematology and Haemato-Oncology, KIMS Hospital, Maidstone, Kent, UK
| | - Saaeha Rauz
- Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Birmingham and Midland Eye Centre, Sandwell and West Birmingham NHS Trust, Birmingham, UK
| | - Guy Smith
- Department of Ophthalmology, Great Western Hospital NHS Foundation Trust, Swindon, UK
| | | | - Anwar Tappuni
- Institute of Dentistry, Queen Mary University of London, London, UK
| | - Stephen B Walsh
- London Tubular Centre, University College London, London, UK
| |
Collapse
|
13
|
Zehrfeld N, Witte T, Ernst D. [Update on Sjögren's syndrome : Diagnostics, treatment, and challenges]. Z Rheumatol 2024; 83:217-228. [PMID: 38498147 DOI: 10.1007/s00393-024-01493-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
Sjögren's syndrome (SjS) is the most common connective tissue disease with a prevalence of 1:200. Predominantly affecting women, SjS is associated with destruction of the exocrine glands, leading to xerophthalmia and xerostomia. In over 50% of patients, there are also extraglandular manifestations, leading to multiple organ manifestations including polyneuropathies and interstitial lung disease as well as symptoms such as fatigue and arthralgia. Diagnostic procedures include biomarkers, in particular anti-SS-A/Ro antibodies, histology of salivary glands, and salivary gland sonography. There are currently no licensed immunosuppressive drugs for SjS, so current treatment is often based on off-label use of drugs. The European League Against Rheumatism (EULAR) has recently published treatment recommendations based on the prevailing organ manifestations. Several promising controlled trials with novel compounds and concepts are currently in progress.
Collapse
Affiliation(s)
| | | | - Diana Ernst
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625, Hannover, Deutschland.
| |
Collapse
|
14
|
Nguyen Y, Beydon M, Foulquier N, Gordon R, Bouillot C, Hammitt KM, Bowman SJ, Mariette X, McCoy SS, Cornec D, Seror R. Identification of outcome domains in primary Sjögren's disease: A scoping review by the OMERACT Sjögren disease working group. Semin Arthritis Rheum 2024; 65:152385. [PMID: 38340608 DOI: 10.1016/j.semarthrit.2024.152385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVES Sjögren's disease (SjD) is a heterogenous disease with a wide range of manifestations, ranging from symptoms of dryness, fatigue, and pain, to systemic involvement. Considerable advances have been made to evaluate systemic activity or patient-reported outcomes, but most of the instruments were not able to assess all domains of this multifaceted disease. The aim of this scoping review was to generate domains that have been assessed in randomized controlled trials, as the first phase of the Outcome Measures in Rheumatology (OMERACT) process of core domain set development. METHODS We systematically searched Medline (Pubmed) and EMBASE between 2002 and March 2023 to identify all randomized controlled trials assessing relevant domains, using both a manual approach and an artificial intelligence software (BIBOT) that applies natural language processing to automatically identify relevant abstracts. Domains were mapped to core areas, as suggested by the OMERACT 2.1 Filter. RESULTS Among the 5,420 references, we included 60 randomized controlled trials, focusing either on overall disease manifestations (53%) or on a single organ/symptom: dry eyes (17%), xerostomia (15%), fatigue (12%), or pulmonary function (3%). The most frequently assessed domains were perceived dryness (52% for overall dryness), fatigue (57%), pain (52%), systemic disease activity (45%), lacrimal gland function (47%) and salivary function (55%), B-cell activation (60%), and health-related quality of life (40%). CONCLUSION Our scoping review highlighted the heterogeneity of SjD, in the study designs and domains. This will inform the OMERACT SjD working group to select the most appropriate core domains to be used in SjD clinical trials and to guide the future agenda for outcome measure research in SjD.
Collapse
Affiliation(s)
- Yann Nguyen
- Service de Rhumatologie, Assistance Publique - Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM), UMR1184, Université Paris-Saclay, Le Kremlin Bicêtre, Paris, France
| | - Maxime Beydon
- Service de Rhumatologie, Assistance Publique - Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | | | - Rachael Gordon
- Department of Medicine, Division of Rheumatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | - Simon J Bowman
- Rheumatology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Xavier Mariette
- Service de Rhumatologie, Assistance Publique - Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM), UMR1184, Université Paris-Saclay, Le Kremlin Bicêtre, Paris, France
| | - Sara S McCoy
- Division of Rheumatology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, USA
| | - Divi Cornec
- LBAI, UMR1227, Univ Brest, Inserm, Brest, France; INSERM, UMR1227, Lymphocytes B, Autoimmunité et Immunothérapies, Université de Bretagne Occidentale, Service de Rhumatologie, CHU de Brest, Brest, France
| | - Raphaèle Seror
- Service de Rhumatologie, Assistance Publique - Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM), UMR1184, Université Paris-Saclay, Le Kremlin Bicêtre, Paris, France.
| |
Collapse
|
15
|
Nguyen Y, Nocturne G, Henry J, Ng WF, Belkhir R, Desmoulins F, Bergé E, Morel J, Perdriger A, Dernis E, Devauchelle-Pensec V, Sène D, Dieudé P, Couderc M, Fauchais AL, Larroche C, Vittecoq O, Salliot C, Hachulla E, Le Guern V, Gottenberg JE, Mariette X, Seror R. Identification of distinct subgroups of Sjögren's disease by cluster analysis based on clinical and biological manifestations: data from the cross-sectional Paris-Saclay and the prospective ASSESS cohorts. THE LANCET. RHEUMATOLOGY 2024; 6:e216-e225. [PMID: 38437852 PMCID: PMC10949202 DOI: 10.1016/s2665-9913(23)00340-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 03/06/2024]
Abstract
BACKGROUND Sjögren's disease is a heterogenous autoimmune disease with a wide range of symptoms-including dryness, fatigue, and pain-in addition to systemic manifestations and an increased risk of lymphoma. We aimed to identify distinct subgroups of the disease, using cluster analysis based on subjective symptoms and clinical and biological manifestations, and to compare the prognoses of patients in these subgroups. METHODS This study included patients with Sjögren's disease from two independent cohorts in France: the cross-sectional Paris-Saclay cohort and the prospective Assessment of Systemic Signs and Evolution of Sjögren's Syndrome (ASSESS) cohort. We first used an unsupervised multiple correspondence analysis to identify clusters within the Paris-Saclay cohort using 26 variables comprising patient-reported symptoms and clinical and biological manifestations. Next, we validated these clusters using patients from the ASSESS cohort. Changes in disease activity (measured by the European Alliance of Associations for Rheumatology [EULAR] Sjögren's Syndrome Disease Activity Index [ESSDAI]), patient-acceptable symptom state (measured by the EULAR Sjögren's Syndrome Patient Reported Index [ESSPRI]), and lymphoma incidence during follow-up were compared between clusters. Finally, we compared our clusters with the symptom-based subgroups previously described by Tarn and colleagues. FINDINGS 534 patients from the Paris-Saclay cohort (502 [94%] women, 32 [6%] men, median age 54 years [IQR 43-64]), recruited between 1999 and 2022, and 395 patients from the ASSESS cohort (370 [94%] women, 25 [6%] men, median age 53 years [43-63]), recruited between 2006 and 2009, were included in this study. In both cohorts, hierarchical cluster analysis revealed three distinct subgroups of patients: those with B-cell active disease and low symptom burden (BALS), those with high systemic disease activity (HSA), and those with low systemic disease activity and high symptom burden (LSAHS). During follow-up in the ASSESS cohort, disease activity and symptom states worsened for patients in the BALS cluster (67 [36%] of 186 patients with ESSPRI score <5 at month 60 vs 92 [49%] of 186 at inclusion; p<0·0001). Lymphomas occurred in patients in the BALS cluster (five [3%] of 186 patients; diagnosed a median of 70 months [IQR 42-104] after inclusion) and the HSA cluster (six [4%] of 158 patients; diagnosed 23 months [13-83] after inclusion). All patients from the Paris-Saclay cohort with a history of lymphoma were in the BALS and HSA clusters. This unsupervised clustering classification based on symptoms and clinical and biological manifestations did not correlate with a previous classification based on symptoms only. INTERPRETATION On the basis of symptoms and clinical and biological manifestations, we identified three distinct subgroups of patients with Sjögren's disease with different prognoses. Our results suggest that these subgroups represent different heterogeneous pathophysiological disease mechanisms, stages of disease, or both. These findings could be of interest when stratifying patients in future therapeutic trials. FUNDING Fondation pour la Recherche Médicale, French Ministry of Health, French Society of Rheumatology, Innovative Medicines Initiative 2 Joint Undertaking, Medical Research Council UK, and Foundation for Research in Rheumatology.
Collapse
Affiliation(s)
- Yann Nguyen
- Department of Rheumatology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, Paris, France; Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Université Paris-Saclay, Paris, France
| | - Gaëtane Nocturne
- Department of Rheumatology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, Paris, France; Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Université Paris-Saclay, Paris, France
| | - Julien Henry
- Department of Rheumatology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, Paris, France
| | - Wan-Fai Ng
- Faculty of Medical Sciences, Clinical and Translational Research Institute, Newcastle University, NIHR Newcastle Biomedical Research Centre and NIHR Newcastle Clinical Research Facility, Newcastle upon Tyne NHS Hospitals Foundation Trust, Newcastle upon Tyne, UK
| | - Rakiba Belkhir
- Department of Rheumatology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, Paris, France
| | - Frédéric Desmoulins
- Department of Rheumatology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, Paris, France
| | - Elisabeth Bergé
- Department of Rheumatology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, Paris, France
| | - Jacques Morel
- Rheumatology Department, CHU de Montpellier, PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Aleth Perdriger
- Rheumatology Department, CHU Rennes, Université Rennes, Rennes, France
| | - Emmanuelle Dernis
- Department of Rheumatology and Clinical Immunology, General Hospital, Le Mans, France
| | - Valérie Devauchelle-Pensec
- Department of Rheumatology, CHU de Brest, INSERM 1227, LBAI, Université de Bretagne Occidentale, Centre de Référence des Maladies Auto-Immunes Rares de l'Adulte, Brest, France
| | - Damien Sène
- Department of Internal Medicine, Hôpital Lariboisière, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Philippe Dieudé
- Department of Rheumatology, Hôpital Bichat-Claude Bernard, Assistance Publique - Hôpitaux de Paris, INSERM UMR1152, Paris-Cité University, Paris, France
| | - Marion Couderc
- Department of Rheumatology, CHU de Clermont-Ferrand, INSERM UMR 1240, Clermont Auvergne University, Clermont-Ferrand, France
| | - Anne-Laure Fauchais
- Department of Internal Medicine, University Hospital of Limoges, Limoges, France
| | - Claire Larroche
- Department of Internal Medicine, Assistance Publique - Hôpitaux de Paris, Hôpital Avicenne, Bobigny, France
| | - Olivier Vittecoq
- Department of Rheumatology, Rouen University Hospital, Rouen, France
| | - Carine Salliot
- Department of Rheumatology, Centre Hospitalier Universitaire d'Orléans, Orléans, France
| | - Eric Hachulla
- Department of Internal Medicine and Clinical Immunology, Hôpital Claude Huriez, University of Lille, Lille, France
| | - Véronique Le Guern
- National Referral Centre for Rare Autoimmune and Systemic Diseases, Department of Internal Medicine, Hôpital Cochin, Assistance Publique - Hôpitaux de Paris Centre, Université Paris Cité, Paris, France
| | - Jacques-Eric Gottenberg
- Rheumatology Department, EA 3432, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, France
| | - Xavier Mariette
- Department of Rheumatology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, Paris, France; Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Université Paris-Saclay, Paris, France
| | - Raphaèle Seror
- Department of Rheumatology, Hôpital Bicêtre, Assistance Publique - Hôpitaux de Paris, Université Paris-Saclay, Paris, France; Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Université Paris-Saclay, Paris, France.
| |
Collapse
|
16
|
Yang H, Sun C, Wang X, Wang T, Xie C, Li Z. Identification of ferroptosis-related diagnostic markers in primary Sjögren's syndrome based on machine learning. Med Oral Patol Oral Cir Bucal 2024; 29:e203-e210. [PMID: 37823298 PMCID: PMC10945879 DOI: 10.4317/medoral.26190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/03/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Primary Sjogren's syndrome (pSS) is a common autoimmune disorder that affects up to 0.3-3% of the global population. Ferroptosis has recently been identified to play a significant role in autoimmune diseases. However, the molecular mechanisms of ferroptosis in the initiation and progression of pSS remains unclear. MATERIAL AND METHODS To investigate the molecular mechanisms underlying the occurrence and progression of pSS, we utilized a comprehensive approach by integrating data obtained from the Gene Expression Omnibus (GEO) database with data from the FerrDb database to identify the ferroptosis-related differentially expressed genes (DEGs). Furthermore, we implemented an innovative transcriptomic analysis method utilizing a computer-aided algorithm to establish a network between hub genes associated with ferroptosis and the immune microenvironment in pSS patients. RESULTS Our results revealed significant differences in the gene expression profiles of pSS samples compared to normal tissues, with 1,830 significantly up-regulated genes and 1,310 significantly down-regulated genes. In addition, our results showed a significant increase in the proportions of B cells and CD4+ T cells in pSS samples compared to normal tissues. AND then, our analysis revealed that a combination of six ferroptosis-related genes, including TBK1, SLC1A4, PIK3CA, ENO3, EGR1, and ATG5, could serve as optimal markers for the diagnosis of pSS. The combined analysis of these six genes accurately diagnosed the occurrence of pSS. CONCLUSIONS This study offers valuable insights into the pathogenesis of pSS and highlights the importance of targeting ferroptosis-related DEGs, which suggests a novel treatment strategy for pSS.
Collapse
Affiliation(s)
- H Yang
- Department of Rheumatology and Immunology the First Affiliated Hospital of Bengbu Medical College No. 287 Changhuai Road, Bengbu, 233004, China
| | | | | | | | | | | |
Collapse
|
17
|
Berry J, Tarn J, Lendrem D, Casement J, Ng WF. What can patients tell us in Sjögren's syndrome? RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2024; 5:34-41. [PMID: 38571930 PMCID: PMC10985711 DOI: 10.1515/rir-2024-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 10/04/2023] [Indexed: 04/05/2024]
Abstract
In Sjögren's Syndrome (SS), clinical heterogeneity and discordance between disease activity measures and patient experience are key obstacles to effective therapeutic development. Patient reported outcome measures (PROMs) are useful tools for understanding the unmet needs from the patients' perspective and therefore they are key for the development of patient centric healthcare systems. Initial concern about the subjectivity of PROMs has given way to methodological rigour and clear guidance for the development of PROMs. To date, several studies of patient stratification using PROMs have identified similar symptom-based subgroups. There is evidence to suggest that these subgroups may represent different disease endotypes with differing responses to therapeutic interventions. Stratified medicine approaches, alongside sensitive outcome measures, have the potential to improve our understanding of SS pathobiology and therapeutic development. The inclusion of PROMs is important for the success of such approaches. In this review we discuss the opportunities of using PROMs in understanding the pathogenesis of and therapeutic development for SS.
Collapse
Affiliation(s)
- Joe Berry
- Translational and Clinical Research Institute, Newcastle University, Newcastle uponTyne, UK
| | - Jessica Tarn
- Translational and Clinical Research Institute, Newcastle University, Newcastle uponTyne, UK
| | - Dennis Lendrem
- Translational and Clinical Research Institute, Newcastle University, Newcastle uponTyne, UK
| | - John Casement
- Translational and Clinical Research Institute, Newcastle University, Newcastle uponTyne, UK
| | - Wan-Fai Ng
- Translational and Clinical Research Institute, Newcastle University, Newcastle uponTyne, UK
- National Institute for Health and Care Research (NIHR) Newcastle Biomedical Research Centre& NIHR Newcastle Clinical Research Facility, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle uponTyne, UK
| |
Collapse
|
18
|
Beydon M, McCoy S, Nguyen Y, Sumida T, Mariette X, Seror R. Epidemiology of Sjögren syndrome. Nat Rev Rheumatol 2024; 20:158-169. [PMID: 38110617 DOI: 10.1038/s41584-023-01057-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 12/20/2023]
Abstract
Sjögren syndrome is a phenotypically varied autoimmune disorder that can occur alone in primary Sjögren syndrome or in association with other connective tissue diseases (CTDs), including rheumatoid arthritis, systemic lupus erythematosus (SLE) and systemic sclerosis (SSc). The estimation of the prevalence and incidence of Sjögren syndrome varies depending on diagnostic criteria and study design, making it difficult to estimate geographical and temporal trends. Nonetheless, disease phenotype is influenced by geographical origin, which is a risk factor for systemic activity. Whether mortality in primary Sjögren syndrome is increased compared with that of the general population is not yet known, but extra-glandular manifestations, in particular lymphomas, are clear risk factors for mortality. In CTDs associated with Sjögren syndrome, lymphoma risk seems higher than that of patients with CTD alone, and there is potentially lower disease activity in SLE with Sjögren syndrome and in SSc with Sjögren syndrome than in SLE or SSc alone.
Collapse
Affiliation(s)
- Maxime Beydon
- Department of Rheumatology, Bicêtre AP-HP Hôpital, Université Paris-Saclay, Paris, France
| | - Sara McCoy
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yann Nguyen
- Department of Rheumatology, Bicêtre AP-HP Hôpital, Université Paris-Saclay, Paris, France
- Center for Immunology of Viral Infections and Auto-Immune Diseases, INSERM U1184, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Takayuki Sumida
- Department of Internal Medicine, University of Tsukuba, Tsukuba, Japan
| | - Xavier Mariette
- Department of Rheumatology, Bicêtre AP-HP Hôpital, Université Paris-Saclay, Paris, France
- Center for Immunology of Viral Infections and Auto-Immune Diseases, INSERM U1184, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Raphaèle Seror
- Department of Rheumatology, Bicêtre AP-HP Hôpital, Université Paris-Saclay, Paris, France.
- Center for Immunology of Viral Infections and Auto-Immune Diseases, INSERM U1184, Université Paris Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
19
|
Dörner T, Kaul M, Szántó A, Tseng JC, Papas AS, Pylvaenaeinen I, Hanser M, Abdallah N, Grioni A, Santos Da Costa A, Ferrero E, Gergely P, Hillenbrand R, Avrameas A, Cenni B, Siegel RM. Efficacy and safety of remibrutinib, a selective potent oral BTK inhibitor, in Sjögren's syndrome: results from a randomised, double-blind, placebo-controlled phase 2 trial. Ann Rheum Dis 2024; 83:360-371. [PMID: 37932009 PMCID: PMC10894844 DOI: 10.1136/ard-2023-224691] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/10/2023] [Indexed: 11/08/2023]
Abstract
OBJECTIVES To evaluate the safety and efficacy of remibrutinib in patients with moderate-to-severe Sjögren's syndrome (SjS) in a phase 2 randomised, double-blind trial (NCT04035668; LOUiSSE (LOU064 in Sjögren's Syndrome) study). METHODS Eligible patients fulfilling 2016 American College of Rheumatology/European League Against Rheumatism (EULAR) criteria for SjS, positive for anti-Ro/Sjögren's syndrome-related antigen A antibodies, with moderate-to-severe disease activity (EULAR Sjögren's Syndrome Disease Activity Index (ESSDAI) (based on weighted score) ≥ 5, EULAR Sjögren's Syndrome Patient Reported Index (ESSPRI) ≥ 5) received remibrutinib (100 mg) either one or two times a day, or placebo for the 24-week study treatment period. The primary endpoint was change from baseline in ESSDAI at week 24. Key secondary endpoints included change from baseline in ESSDAI over time, change from baseline in ESSPRI over time and safety of remibrutinib in SjS. Key exploratory endpoints included changes to the salivary flow rate, soluble biomarkers, blood transcriptomic and serum proteomic profiles. RESULTS Remibrutinib significantly improved ESSDAI score in patients with SjS over 24 weeks compared with placebo (ΔESSDAI -2.86, p=0.003). No treatment effect was observed in ESSPRI score (ΔESSPRI 0.17, p=0.663). There was a trend towards improvement of unstimulated salivary flow with remibrutinib compared with placebo over 24 weeks. Remibrutinib had a favourable safety profile in patients with SjS over 24 weeks. Remibrutinib induced significant changes in gene expression in blood, and serum protein abundance compared with placebo. CONCLUSIONS These data show preliminary efficacy and favourable safety of remibrutinib in a phase 2 trial for SjS.
Collapse
Affiliation(s)
- Thomas Dörner
- Dept. Med./Rheumatology and Clinical Immunology, Charite Univ. Hospital, Berlin, Germany
| | - Martin Kaul
- Novartis Institutes for BioMedical Research, Basel, Switzerland
- Independent consultant, Neustadt, Germany
| | - Antónia Szántó
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Athena S Papas
- Oral Medicine, Tufts University School of Dental Medicine, Boston, Massachusetts, USA
| | | | - Malika Hanser
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nasri Abdallah
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Andrea Grioni
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Enrico Ferrero
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Peter Gergely
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | - Bruno Cenni
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | |
Collapse
|
20
|
Zhou X, Xu D, Li M, Zeng X. New investigational drugs to treat Sjogren's syndrome: lessons learnt from immunology. Expert Opin Investig Drugs 2024; 33:105-114. [PMID: 38293750 DOI: 10.1080/13543784.2024.2312216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/26/2024] [Indexed: 02/01/2024]
Abstract
INTRODUCTION Sjögren's syndrome is a heterogeneous autoimmune condition that impairs quality of life because of dryness, fatigue, pain, and systemic involvements. Current treatment largely depends on empirical evidence, with no effective therapy approved. Clinical trials on targeted drugs often fail to report efficacy due to common factors. AREAS COVERED This review summarizes the pathogenesis and what caused the failure of new investigational drugs in clinical trials, highlighting solutions for more effective investigations, with greater consistency between research outcomes, clinical use, and patient needs. EXPERT OPINION Unlinked pathobiology with symptoms resulted in misidentified targets and disappointing trials. Useful stratification tools are necessary for the heterogeneous SS patients. Composite endpoints or improvements in ESSDAI scores are needed, considering the high placebo response, and the unbalance between symptom burden and disease activity. Compared to classic biologics, targeted cell therapy will be a more promising field of investigation in the coming years.
Collapse
Affiliation(s)
- Xingyu Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Dong Xu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
21
|
de Wolff L, Arends S, Verstappen GM, Bootsma H. Minimal added value of separate dryness assessments compared with overall dryness in ESSPRI in patients with Sjögren's disease. Rheumatology (Oxford) 2024; 63:e29-e31. [PMID: 37632773 PMCID: PMC10836974 DOI: 10.1093/rheumatology/kead441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/12/2023] [Accepted: 08/04/2023] [Indexed: 08/28/2023] Open
Affiliation(s)
- Liseth de Wolff
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Suzanne Arends
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Gwenny M Verstappen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
22
|
Bentley D, Fisher BA, Barone F, Kolb FA, Attley G. A randomized, double-blind, placebo-controlled, parallel group study on the effects of a cathepsin S inhibitor in primary Sjögren's syndrome. Rheumatology (Oxford) 2023; 62:3644-3653. [PMID: 36864622 PMCID: PMC10629789 DOI: 10.1093/rheumatology/kead092] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/06/2023] [Indexed: 03/04/2023] Open
Abstract
OBJECTIVES Primary SS (pSS) is a chronic autoimmune disorder characterized by mucosal dryness and systemic symptoms. We tested the effects of inhibition of cathepsin S using the potent and selective inhibitor RO5459072 on disease activity and symptoms of pSS. METHODS This was a randomized, double-blind, placebo-controlled, parallel-group, Phase IIA study to investigate the effects of RO5459072 (100 mg twice daily; 200 mg per day). Seventy-five patients with pSS were randomized 1:1 to receive either RO5459072 or placebo for 12 weeks. The primary outcome was the proportion of patients with a ≥3 point reduction from baseline in EULAR SS Disease Activity Index (ESSDAI) score. We also investigated the effects of RO5459072 on quality of life, exocrine gland function, biomarkers related to SS, and safety and tolerability. RESULTS The proportion of patients showing an improvement in ESSDAI score was not significantly different between the RO5459072 and placebo arms. No clinically meaningful treatment effects were observed in favour of RO5459072 for all secondary outcomes. Analysis of soluble biomarkers indicated target engagement between RO5459072 and cathepsin S. There were modest decreases in the number of circulating B cells and T cells in the RO5459072 group, although these did not reach significance. RO5459072 was safe and well-tolerated. CONCLUSIONS There was no clinically relevant improvement in ESSDAI score (primary endpoint), and no apparent benefit in favour of RO5459072 in any of the secondary clinical endpoints. Further work is needed in order to understand the mechanisms of MHC-II-mediated immune stimulation in pSS. TRIAL REGISTRATION ClinicalTrials.gov; NCT02701985.
Collapse
Affiliation(s)
| | - Benjamin A Fisher
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- National Institute of Health Research (NIHR) Birmingham Biomedical Research Centre and Department of Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | - Gemma Attley
- Roche Pharma Research and Early Development, Little Falls, NJ, USA
| |
Collapse
|
23
|
Arends S, Verstappen GM, de Wolff L, Pringle S, Kroese FGM, Vissink A, Bootsma H. Why do drug treatments fail in Sjögren's disease? Considerations for treatment, trial design and interpretation of clinical efficacy. Expert Rev Clin Immunol 2023; 19:1187-1194. [PMID: 37551702 DOI: 10.1080/1744666x.2023.2234641] [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: 05/01/2023] [Accepted: 07/05/2023] [Indexed: 08/09/2023]
Abstract
INTRODUCTION Despite ongoing efforts to develop effective therapeutics, no disease-modifying drugs have been officially licensed for the indication of Sjögren's disease (SjD). This is partly due to heterogeneity in disease manifestations, which complicates drug target selection, trial design and interpretation of clinical efficacy in SjD. AREAS COVERED Here, we summarize developments and comment on challenges in 1) identifying the right target for treatment, 2) selection of the primary study endpoint for trials and definition of clinically relevant response to treatment, 3) inclusion criteria and patient stratification, 4) distinguishing between disease activity and damage and 5) establishing the effect of treatment considering measurement error, natural variation, and placebo or nocebo responses. EXPERT OPINION Targets that are involved in both the immune cell response and dysregulation of glandular epithelial cells (e.g. B-lymphocytes, type-I interferon) are of particular interest to treat both glandular and extra-glandular manifestations of SjD. The recent development of composite study endpoints (CRESS and STAR) may be a crucial step forward in the search for clinically effective systemic treatment of patients with SjD. Important additional areas for future research are symptom-based and/or molecular pathway-based patient stratification, prevention of irreversible damage, and establishing the effect of treatment.
Collapse
Affiliation(s)
- Suzanne Arends
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Gwenny M Verstappen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Liseth de Wolff
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Sarah Pringle
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Frans G M Kroese
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Arjan Vissink
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
24
|
Soyfer EM, Fleischman AG. Myeloproliferative neoplasms - blurring the lines between cancer and chronic inflammatory disorder. Front Oncol 2023; 13:1208089. [PMID: 37361587 PMCID: PMC10288874 DOI: 10.3389/fonc.2023.1208089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Myeloproliferative Neoplasm (MPN) is a group of chronic blood cancers that arise from a hematopoietic stem cell (HSC) clone with somatic mutations causing constitutive activation of myeloid cytokine receptor signaling. In addition to elevated blood cell counts, MPN typically presents with increased inflammatory signaling and inflammation symptoms. Therefore, while being a clonally derived neoplasm, MPN has much in common with chronic non-cancerous inflammatory conditions, such as rheumatoid arthritis, lupus, and many more. MPN and chronic inflammatory disease (CID) share similar chronicity, symptoms, dependency on the immune system, environmental triggers, and treatments. Overall, we will highlight the similarities between an MPN and CID. We highlight that while MPN is classified as a cancer, its behavior is more aligned to that of a chronic inflammatory disease. We propose that MPN should inhabit a fluid/spectrum between auto-inflammatory disease and cancer.
Collapse
Affiliation(s)
- Eli M. Soyfer
- School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Angela G. Fleischman
- School of Medicine, University of California, Irvine, Irvine, CA, United States
- Division of Hematology/Oncology, University of California (UC) Irvine Health, Irvine, CA, United States
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
25
|
de Frémont GM, Costedoat-Chalumeau N, Lazaro E, Belkhir R, Guettrot-Imbert G, Morel N, Nocturne G, Molto A, Goulenok T, Diot E, Perard L, Ferreira-Maldent N, Le Besnerais M, Limal N, Martis N, Abisror N, Debouverie O, Richez C, Sobanski V, Maurier F, Sauvetre G, Levesque H, Timsit MA, Tieulié N, Orquevaux P, Bienvenu B, Mahevas M, Papo T, Lartigau-Roussin C, Chauvet E, Berthoux E, Sarrot-Reynauld F, Raffray L, Couderc M, Silva NM, Jourde-Chiche N, Belhomme N, Thomas T, Poindron V, Queyrel-Moranne V, Delforge J, Le Ray C, Pannier E, Mariette X, Le Guern V, Seror R. Pregnancy outcomes in women with primary Sjögren's syndrome: an analysis of data from the multicentre, prospective, GR2 study. THE LANCET. RHEUMATOLOGY 2023; 5:e330-e340. [PMID: 38251600 DOI: 10.1016/s2665-9913(23)00099-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Adverse pregnancy outcomes in women with primary Sjögren's syndrome have only been evaluated retrospectively using heterogeneous methods and with contradictory results. We aimed to describe adverse pregnancy, delivery, and birth outcome risks in pregnant women with primary Sjögren's syndrome compared with those of a matched general population in France, and to identify factors predictive of disease flares or adverse pregnancy outcomes. METHODS We conducted a multicentre, prospective, cohort study in France using the GR2 (Groupe de Recherche sur la Grossesse et les Maladies Rares) registry. Women from the GR2 study were eligible if they had conceived before March, 2021, had primary Sjögren's syndrome according to the American College of Rheumatology and European Alliance of Associations for Rheumatology (EULAR) 2016 classification criteria, and had an ongoing pregnancy at 12 weeks of gestation. In women who entered in the registry with pregnancies before 18 weeks of gestation, we sought to identify factors associated with primary Sjögren's syndrome flare (≥3-point increase in EULAR Sjögren's Syndrome Disease Activity Index [ESSDAI] score) or adverse pregnancy outcomes (fetal or neonatal death, placental insufficiency leading to a preterm delivery [<37 weeks of gestation], or small-for-gestational-age birthweight). A matched controlled study compared adverse pregnancy, delivery, and birth outcome rates between pregnant women with primary Sjögren's syndrome from the GR2 registry and matched controls from the general population included in the last French perinatal survey (Enquête Nationale Périnatale 2016). FINDINGS 1944 pregnancies were identified in the GR2 cohort, of which 106 pregnancies in 96 women with primary Sjögren's syndrome were included in this analysis. The median age at pregnancy onset was 33 years (IQR 31-36). 87 (83%) of 105 pregnancies (with ethnicity data) were in White women, 18 (17%) were in Black women; 92 (90%) of 102 had previous systemic activity (ESSDAI score of ≥1; data missing in four pregnancies), and 48 (45%) of 106 had systemic activity at inclusion. Of 93 pregnancies included at week 18 of gestation or earlier, primary Sjögren's syndrome flares occurred in 12 (13%). No baseline parameters were associated with primary Sjögren's syndrome flare. Four twin pregnancies and one medical termination were excluded from the adverse pregnancy outcome analysis; of the remaining 88, adverse pregnancy outcomes occurred in six (7%). Among pregnancies in women with data for antiphospholipid antibodies (n=55), antiphospholipid antibody positivity was more frequent among pregnancies with adverse outcomes (two [50%] of four pregnancies) compared with those without adverse outcomes (two [4%] of 51 pregnancies; p=0·023). Anti-RNP antibody positivity was also more frequent among pregnancies with adverse outcomes than those without, although this was not statistically significant. In the matched controlled study, adverse pregnancy outcomes occurred in nine (9%) of 105 pregnancies in women with primary Sjögren's syndrome and 28 (7%) of the 420 matched control pregnancies; adverse pregnancy outcomes were not significantly associated with primary Sjögren's syndrome (odds ratio 1·31, 95% CI 0·53-2·98; p=0·52). INTERPRETATION Pregnancies in women with primary Sjögren's syndrome had very good prognoses for mothers and fetuses, with no overall increase in adverse pregnancy outcome risk compared with the general population. Women with antiphospholipid antibodies or anti-RNP antibodies require close monitoring, because these factors might be associated with a higher risk of adverse pregnancy outcomes. FUNDING Lupus France, Association des Sclérodermiques de France, Association Gougerot Sjögren, Association Francophone Contre la Polychondrite Chronique Atrophiante, AFM-Telethon, Société Nationale Française de Médecine Interne, Société Française de Rhumatologie, Cochin Hospital, French Health Ministry, Fondation for Research in Rheumatology, Association Prix Véronique Roualet, Union Chimique Belge.
Collapse
Affiliation(s)
- Grégoire Martin de Frémont
- APHP, Hôpital Bicêtre, Service de Rhumatologie, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Inserm UMR 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Nathalie Costedoat-Chalumeau
- APHP, Hôpital Cochin, Service de Médecine Interne, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Paris, France; Centre de Recherche Epidémiologie et Biostatistiques de Sorbonne Paris Cité, Université de Paris, Paris, France
| | - Estibaliz Lazaro
- CHU de Bordeaux, Service de Médecine Interne et Maladies Infectieuses, Bordeaux, France
| | - Rakiba Belkhir
- APHP, Hôpital Bicêtre, Service de Rhumatologie, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Inserm UMR 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Gaëlle Guettrot-Imbert
- APHP, Hôpital Cochin, Service de Médecine Interne, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Paris, France
| | - Nathalie Morel
- APHP, Hôpital Cochin, Service de Médecine Interne, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Paris, France
| | - Gaétane Nocturne
- APHP, Hôpital Bicêtre, Service de Rhumatologie, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Inserm UMR 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Anna Molto
- Centre de Recherche Epidémiologie et Biostatistiques de Sorbonne Paris Cité, Université de Paris, Paris, France; APHP, Hôpital Cochin, Service de Rhumatologie, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Paris, France
| | | | - Elisabeth Diot
- CHU de Tours, Service de Médecine Interne, Tours, France
| | - Laurent Perard
- Hôpital Saint-Joseph, Service de Médecine Interne, Lyon, France
| | | | | | - Nicolas Limal
- APHP, Hôpital Henri-Mondor, Service de Médecine Interne, Créteil, France
| | - Nihal Martis
- CHU de Nice, Hôpital Archet, Service de Médecine Interne, Nice, France
| | - Noémie Abisror
- APHP, Hôpital Saint-Antoine, Service de Médecine Interne, Paris, France
| | | | | | - Vincent Sobanski
- CHU de Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares, Inserm U1286, Université de Lille, Lille, France
| | - François Maurier
- Hôpitaux Privés de Metz, Service de Médecine Interne, Metz, France
| | | | - Hervé Levesque
- CHU de Rouen, Service de Médecine Interne, Rouen, France
| | | | | | | | - Boris Bienvenu
- Hôpital Saint-Joseph, Service de Médecine Interne, Marseille, France
| | - Matthieu Mahevas
- APHP, Hôpital Henri-Mondor, Service de Médecine Interne, Créteil, France
| | - Thomas Papo
- APHP, Hôpital Bichat, Service de Médecine Interne, Paris, France
| | | | - Elodie Chauvet
- Polyclinique Médipôle Saint-Roch, Service de Médecine Interne, Cabestany, France
| | - Emilie Berthoux
- Hôpital Saint-Joseph, Service de Médecine Interne, Lyon, France
| | | | - Loïc Raffray
- CHU Félix-Guyon, Service de Médecine Interne, Saint-Denis de la Réunion, France
| | - Marion Couderc
- CHU de Clermont-Ferrand, Service de Rhumatologie, Clermont-Ferrand, France
| | | | - Noémie Jourde-Chiche
- APHM, CHU de la Conception, Centre de Néphrologie et Transplantation Rénale, C2VN, Inserm 1263, Institut National de la Recherche Agronomique (INRA) 1260, Faculté de Pharmacie, Marseille, France
| | | | - Thierry Thomas
- CHU de Saint-Etienne, Service de Médecine Interne, Saint-Etienne, France
| | - Vincent Poindron
- CHU de Strasbourg, Service de Médecine Interne, Strasbourg, France
| | | | - Juliette Delforge
- APHP, Hôpital Jean-Verdier, Service de Médecine Interne, Bobigny, France
| | - Camille Le Ray
- INSERM UMR 1153, Obstetrical, Perinatal and Pediatric Epidemiology Research Team (Epopé), Center for Epidemiology and Statistics, FHU PREMA, Université Paris Cité, Paris, France
| | - Emmanuelle Pannier
- APHP, Hôpital Cochin Port Royal, Maternité Port Royal, Service d'Obstétrique, Université de Paris, Paris, France
| | - Xavier Mariette
- APHP, Hôpital Bicêtre, Service de Rhumatologie, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Inserm UMR 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Véronique Le Guern
- APHP, Hôpital Cochin, Service de Médecine Interne, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Paris, France
| | - Raphaèle Seror
- APHP, Hôpital Bicêtre, Service de Rhumatologie, Centre de Référence Maladies Auto-Immunes et Systémiques Rares d'Île-de-France, Inserm UMR 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
26
|
Thorlacius GE, Björk A, Wahren-Herlenius M. Genetics and epigenetics of primary Sjögren syndrome: implications for future therapies. Nat Rev Rheumatol 2023; 19:288-306. [PMID: 36914790 PMCID: PMC10010657 DOI: 10.1038/s41584-023-00932-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2023] [Indexed: 03/14/2023]
Abstract
In primary Sjögren syndrome (pSS), chronic inflammation of exocrine glands results in tissue destruction and sicca symptoms, primarily of the mouth and eyes. Fatigue, arthralgia and myalgia are also common symptoms, whereas extraglandular manifestations that involve the respiratory, nervous and vascular systems occur in a subset of patients. The disease predominantly affects women, with an estimated female to male ratio of 14 to 1. The aetiology of pSS, however, remains incompletely understood, and effective treatment is lacking. Large-scale genetic and epigenetic investigations have revealed associations between pSS and genes in both innate and adaptive immune pathways. The genetic variants mediate context-dependent effects, and both sex and environmental factors can influence the outcome. As such, genetic and epigenetic studies can provide insight into the dysregulated molecular mechanisms, which in turn might reveal new therapeutic possibilities. This Review discusses the genetic and epigenetic features that have been robustly connected with pSS, putting them into the context of cellular function, carrier sex and environmental challenges. In all, the observations point to several novel opportunities for early detection, treatment development and the pathway towards personalized medicine.
Collapse
Affiliation(s)
- Gudny Ella Thorlacius
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Albin Björk
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Rheumatology, Academic Specialist Center, Stockholm, Sweden
| | - Marie Wahren-Herlenius
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
27
|
Arends S, van der Geest KS, Sandovici M. Assessment of treatment efficacy in polymyalgia rheumatica. THE LANCET. RHEUMATOLOGY 2023; 5:e171-e173. [PMID: 38251513 DOI: 10.1016/s2665-9913(23)00056-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 03/08/2023]
Affiliation(s)
- Suzanne Arends
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, Netherlands.
| | - Kornelis Sm van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, Netherlands
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, Netherlands
| |
Collapse
|
28
|
Wratten S, Abetz-Webb L, Arenson E, Griffiths P, Bowman S, Hueber W, Ndife B, Kuessner D, Goswami P. Development and testing of an alternative responder definition for EULAR Sjögren's Syndrome Patient Reported Index (ESSPRI). RMD Open 2023; 9:e002721. [PMID: 36931685 PMCID: PMC10030922 DOI: 10.1136/rmdopen-2022-002721] [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: 09/08/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
OBJECTIVES Dryness, fatigue and joint/muscle pain are typically assessed in Sjögren's trials using European Alliance of Associations for Rheumatology Sjögren's Syndrome Patient Reported Index (ESSPRI). A Patient Acceptable Symptom State of <5 and a Minimal Clinically Important Improvement (MCII)/responder definition (RD) of ≥1 point or 15% on ESSPRI have previously been defined. This study explored alternative RDs to better discriminate between active treatment and placebo in trials. METHODS Anchor-based and distribution-based methods were used to derive RD thresholds in blinded phase IIb trial data (N=190) and confirm these in blinded data pooled from three early phase II trials (N=126). The populations consisted of individuals with moderate-to-severe systemic primary Sjögren's. Anchors were prioritised by ESSPRI correlations and used in similar conditions. Triangulated estimates were discussed with experts (N=3). The revised RD was compared with the original using unblinded data to assess placebo and treatment responder rates. RESULTS Patients were predominantly female (>90%), white (90%), with mean age of 50 years. Receiver operating characteristic estimates supported an MCII threshold of 1.5-1.6 in the phase II data, whereas correlation-weighted mean change estimates supported a low/minimal symptom severity threshold of ≥2. A low/minimal symptom severity of ≤3 showed the greatest sensitivity/specificity balance. Analyses in the pooled data supported these thresholds (MCII: 1.5-2.1; low/minimal symptom severity: 2.7-3.7). Unblinded analyses confirmed the revised RD reduced placebo rates. CONCLUSIONS Completing a trial with an improvement of ≥1.5 points compared with baseline and an ESSPRI score of ≤3 points is a relevant RD for moderate-to-severe systemic Sjögren's and reduces placebo rates.
Collapse
Affiliation(s)
| | | | - Ethan Arenson
- Patient-Centered Outcomes, Adelphi Values, Bollington, UK
| | - Pip Griffiths
- Patient-Centered Outcomes, Adelphi Values, Bollington, UK
| | - Simon Bowman
- Rheumatology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Wolfgang Hueber
- Department of Immunology, Novartis Pharma, Basel, Switzerland
| | - Briana Ndife
- Department of Oncology, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Daniel Kuessner
- Department of Immunology, Novartis Pharma, Basel, Switzerland
| | | |
Collapse
|
29
|
He J, Chen J, Miao M, Zhang R, Cheng G, Wang Y, Feng R, Huang B, Luan H, Jia Y, Jin Y, Zhang X, Shao M, Wang Y, Zhang X, Li J, Zhao X, Wang H, Liu T, Xiao X, Zhang X, Su Y, Mu R, Ye H, Li R, Liu X, Liu Y, Li C, Liu H, Hu F, Guo J, Liu W, Zhang WB, Jacob A, Ambrus JL, Ding C, Yu D, Sun X, Li Z. Efficacy and Safety of Low-Dose Interleukin 2 for Primary Sjögren Syndrome: A Randomized Clinical Trial. JAMA Netw Open 2022; 5:e2241451. [PMID: 36355371 PMCID: PMC9650609 DOI: 10.1001/jamanetworkopen.2022.41451] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
IMPORTANCE Primary Sjögren syndrome (pSS) is a systemic autoimmune disease associated with dysregulated immune cells, with no efficient therapy. There is a need to study potential therapeutic approaches. OBJECTIVE To investigate the efficacy, safety, and immune response of low-dose interleukin 2 (LD-IL-2) in the treatment of pSS. DESIGN, SETTING, AND PARTICIPANTS A double-blind, placebo-controlled randomized clinical trial was conducted with a 2-group superiority design from June 2015 to August 2017. Sixty patients, aged 18 to 70 years, were recruited from Peking University People's Hospital. Efficacy analyses were based on the intention-to-treat (ITT) principle. Data were analyzed from December 2018 to March 2020. INTERVENTIONS Patients with pSS were treated with LD-IL-2 or placebo for 12 weeks and accompanied by 12 weeks of follow-up. MAIN OUTCOMES AND MEASURES The primary end point was defined as a 3-point or greater improvement on the European League Against Rheumatism Sjögren's Syndrome Disease Activity Index (ESSDAI) by week 24. The secondary end points included other clinical responses, safety, and changes of immune cell subsets at week 12 and 24. RESULTS Sixty patients with pSS were recruited, with 30 in the LD-IL-2 group (mean [SD] age, 47.6 [12.8] years; 30 [100%] women) and 30 in the placebo group (mean [SD] age, 51.0 [11.9] years; 30 [100%] women), and 57 completed the trial. More patients in the LD-IL-2 group (20 [66.7%]) achieved ESSDAI score reduction of at least 3 points than in the placebo group (8 [26.7%]) at week 24 (P = .004). There were greater resolutions of dryness, pain, and fatigue in the LD-IL-2 group than placebo group at week 12 (dryness: difference, -18.33 points; 95% CI, -28.46 to -8.21 points; P = .001; pain: difference, -10.33 points; 95% CI, -19.38 to -1.29 points; P = .03; fatigue: difference, -11.67 points; 95% CI, -20.65 to -2.68 points; P = .01). No severe adverse events were observed in either group. In addition, the LD-IL-2 group showed a significant decrease in infection compared with the placebo group (1 [3.3%] vs 9 [30.0%]; P = .006). Immunological analysis revealed that LD-IL-2 promoted an expansion of regulatory T cells and regulatory CD24highCD27+ B cells. CONCLUSIONS AND RELEVANCE In this randomized clinical trial, LD-IL-2 was effective and well tolerated in patients with pSS, and it restored immune balance, with enhanced regulatory T cells and CD24highCD27+ B cells. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02464319.
Collapse
Affiliation(s)
- Jing He
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Jiali Chen
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Miao Miao
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Ruijun Zhang
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Gong Cheng
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Yifan Wang
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Ruiling Feng
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Bo Huang
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Huijie Luan
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Yuan Jia
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Yuebo Jin
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Xiaoying Zhang
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Miao Shao
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Yu Wang
- Center for Applied Statistics and School of Statistics, Renmin University of China, Beijing, China
| | - Xia Zhang
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Jing Li
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Xiaozhen Zhao
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Han Wang
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Tian Liu
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Xian Xiao
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Xuewu Zhang
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Yin Su
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Rong Mu
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Hua Ye
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Ru Li
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Xu Liu
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Yanying Liu
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Chun Li
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Huixin Liu
- Department of Clinical Epidemiology and Biostatistics, Peking University People’s Hospital, Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Jianping Guo
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Wanli Liu
- Institute for Immunology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Wen-Bin Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Center for Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing, People’s Republic of China
| | | | | | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Ian Frazer Centre for Children’s Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
30
|
Ritter J, Chen Y, Stefanski AL, Dörner T. Current and future treatment in primary Sjögren's syndrome - A still challenging development. Joint Bone Spine 2022; 89:105406. [PMID: 35537697 DOI: 10.1016/j.jbspin.2022.105406] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/14/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022]
Abstract
Primary Sjögren's syndrome (pSS) is a chronic autoimmune disease characterized by sicca symptoms, systemic manifestations and constitutional symptoms substantially diminishing patient's quality of life. In this review, we summarize recent recommendations for management of pSS patients and current clinical studies in pSS addressing unmet medical needs. Expanding knowledge about disease pathogenesis and the introduction of validated outcome measures, such as capturing disease activity (ESSDAI) and patient-reported outcomes (ESSPRI) have shaped recent developments. In contrast, lack of evidence for current treatment options remarkably limits the management of pSS patients as reflected by the 2019 updated EULAR recommendations for management of Sjögren's syndrome. In this context, symptomatic treatment is usually appropriate for sicca symptoms, whereas systemic treatment is reserved for moderate to severe organ manifestations including care by a multidisciplinary team in centers of expertise. Most promising targets for new treatment modalities are based on immunopathological insights and include direct B cell targeting strategies, targeting co-stimulation by CD40/CD40L blocking, inhibition of key cytokine activity (BLyS/BAFF, type I interferon) and intracellular signaling pathways.
Collapse
Affiliation(s)
- Jacob Ritter
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Yidan Chen
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Gesellschaft, Berlin, Germany
| | - Ana-Luisa Stefanski
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Gesellschaft, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Gesellschaft, Berlin, Germany.
| |
Collapse
|
31
|
JAK/STAT Pathway Targeting in Primary Sjögren Syndrome. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2022; 3:95-102. [PMID: 36788973 PMCID: PMC9895869 DOI: 10.2478/rir-2022-0017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/25/2022] [Indexed: 11/06/2022]
Abstract
Primary Sjögren's syndrome (pSS) is an autoimmune systemic disease mainly affecting exocrine glands and resulting in disabling symptoms, as dry eye and dry mouth. Mechanisms underlying pSS pathogenesis are intricate, involving multiplanar and, at the same time, interlinked levels, e.g., genetic predisposition, epigenetic modifications and the dysregulation of both immune system and glandular-resident cellular pathways, mainly salivary gland epithelial cells. Unravelling the biological and molecular complexity of pSS is still a great challenge but much progress has been made in recent years in basic and translational research field, allowing the identification of potential novel targets for therapy development. Despite such promising novelties, however, none therapy has been specifically approved for pSS treatment until now. In recent years, growing evidence has supported the modulation of Janus kinases (JAK) - signal transducers and activators of transcription (STAT) pathways as treatment strategy immune mediated diseases. JAK-STAT pathway plays a crucial role in autoimmunity and systemic inflammation, being involved in signal pathways of many cytokines. This review aims to report the state-of-the-art about the role of JAK-STAT pathway in pSS, with particular focus on available research and clinical data regarding the use of JAK inhibitors in pSS.
Collapse
|
32
|
Ridgewell D, Thalayasingam N, Ng WF. Sjögren's syndrome: shedding light on emerging and key drug targets. Expert Opin Ther Targets 2022; 26:869-882. [PMID: 36576336 DOI: 10.1080/14728222.2022.2157259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Sjögren's syndrome (SS) is an immune-mediated inflammatory condition characterized by sicca syndrome, musculoskeletal pain, and fatigue. Extra-glandular manifestations are common and there is a markedly increased risk of lymphoma development. SS is associated with high health-economic burden driven largely by the symptom burden on patients. Currently, there is no approved disease-modifying treatment and management is based on empirical evidence. Progress in the understanding of SS pathogenesis has led to an expanding portfolio of more targeted therapies under development. AREAS COVERED This review summarizes the key development in targeted biological therapies in SS including emerging targets. It also highlights the challenges in therapeutic development in SS such as disease heterogeneity and defining appropriate disease assessment tools to evaluate therapeutic efficacy. EXPERT OPINION Early trials in SS failed to meet their primary outcomes which may in part due to the use of inappropriate or insensitive study endpoints. Recent trials targeting B-cells, B-T cell co-stimulation and IFN signaling have shown promising results. Development of composite endpoints including patient reported outcomes and objective disease measure may provide a more holistic approach to disease assessment. The impact of these new tools on therapeutic development that benefit patients remains to be fully evaluated.
Collapse
Affiliation(s)
- Dominic Ridgewell
- Musculoskeletal Theme, NIHR Newcastle Biomedical Research Centre and NIHR Newcastle Clinical Research Facility, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Nishanthi Thalayasingam
- Musculoskeletal Theme, NIHR Newcastle Biomedical Research Centre and NIHR Newcastle Clinical Research Facility, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Wan-Fai Ng
- Musculoskeletal Theme, NIHR Newcastle Biomedical Research Centre and NIHR Newcastle Clinical Research Facility, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK.,Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
33
|
Zeng L, He Q, Yang K, Hao W, Yu G, Chen H. A Systematic Review and Meta-Analysis of 19 Randomized Controlled Trials of Iguratimod Combined With Other Therapies for Sjogren’s Syndrome. Front Immunol 2022; 13:924730. [PMID: 35967307 PMCID: PMC9367640 DOI: 10.3389/fimmu.2022.924730] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo explore the efficacy and safety of Iguratimod intervention in Primary Sjogren’s syndrome (pSS).MethodsMany databases were searched to collect the RCTs. Three independent reviewers extracted data and assessed the quality of the studies based on the Cochrane Handbook. The statistical analysis was done by RevMan 5.3 and STATA. The quality of evidence was evaluated by GRADE tool.ResultsTwenty-nine RCTs with 2258 participants were included in this review. The meta-analysis shows that: iguratimod experiment group can reduce the ESSPRI score (WMD -1.93 [-2.33, -1.52], P<0.00001), ESSDAI score (WMD -1.39 [-1.81, -0.98], P<0.00001), Schirmer’s test (WMD 1.77 [0.85, 2.70], P=0.0002), RF (WMD -5.78 [-7.59, -3.97], P<0.00001), and decrease the ESR level (WMD -7.05 [-9.84, -4.26], P<0.00001). Meanwhile, the summary result showed the addiction of Iguratimod may not increase the adverse events. The adverse events were mainly gastrointestinal discomfort, abnormal liver function, and rash and itching. The quality of evidence of adverse events was moderate. Referring to minimal clinically important difference (MCID), the improvement of ESSPRI is clinically significant, and the improvement of ESSDAI for patients older than 60 years old may be clinically significant.ConclusionBased on current evidence, iguratimod can effectively reduce ESSPRI score, ESSDAI score, Schirmer’s test score and decrease systemic inflammatory response (such as ESR level and RF level) without increasing the probability of adverse events. The recommended course of treatment is at least 12 weeks.Systematic Review Registrationhttps://www.crd.york.ac.uk/prospero/display_record.php?, identifier CRD42020220770.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Qi He, ; Kailin Yang,
| | - Qi He
- People's Hospital of Ningxiang City, Ningxiang City, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Qi He, ; Kailin Yang,
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha City, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Qi He, ; Kailin Yang,
| | - Wensa Hao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Ganpeng Yu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha City, China
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Qi He, ; Kailin Yang,
| |
Collapse
|
34
|
Martin-Gutierrez L, Wilson R, Castelino M, Jury EC, Ciurtin C. Multi-Omic Biomarkers for Patient Stratification in Sjogren's Syndrome-A Review of the Literature. Biomedicines 2022; 10:1773. [PMID: 35892673 PMCID: PMC9332255 DOI: 10.3390/biomedicines10081773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Sjögren's syndrome (SS) is a heterogeneous autoimmune rheumatic disease (ARD) characterised by dryness due to the chronic lymphocytic infiltration of the exocrine glands. Patients can also present other extra glandular manifestations, such as arthritis, anaemia and fatigue or various types of organ involvement. Due to its heterogenicity, along with the lack of effective treatments, the diagnosis and management of this disease is challenging. The objective of this review is to summarize recent multi-omic publications aiming to identify biomarkers in tears, saliva and peripheral blood from SS patients that could be relevant for their better stratification aiming at improved treatment selection and hopefully better outcomes. We highlight the relevance of pro-inflammatory cytokines and interferon (IFN) as biomarkers identified in higher concentrations in serum, saliva and tears. Transcriptomic studies confirmed the upregulation of IFN and interleukin signalling in patients with SS, whereas immunophenotyping studies have shown dysregulation in the immune cell population frequencies, specifically CD4+and C8+T activated cells, and their correlations with clinical parameters, such as disease activity scores. Lastly, we discussed emerging findings derived from different omic technologies which can provide integrated knowledge about SS pathogenesis and facilitate personalised medicine approaches leading to better patient outcomes in the future.
Collapse
Affiliation(s)
- Lucia Martin-Gutierrez
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK; (L.M.-G.); (E.C.J.)
| | - Robert Wilson
- Department of Rheumatology, University College London Hospitals NHS Trust, London NW1 2PG, UK; (R.W.); (M.C.)
| | - Madhura Castelino
- Department of Rheumatology, University College London Hospitals NHS Trust, London NW1 2PG, UK; (R.W.); (M.C.)
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK; (L.M.-G.); (E.C.J.)
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London WC1E 6JF, UK
| |
Collapse
|
35
|
Discriminative power of salivary gland ultrasound in relation to symptom-based endotypes in suspected and definite primary Sjögren's Syndrome. Semin Arthritis Rheum 2022; 56:152075. [PMID: 35907342 DOI: 10.1016/j.semarthrit.2022.152075] [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: 05/31/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVES Salivary gland ultrasound (SGUS) is emerging as essential tool in primary Sjögren's Syndrome (pSS), but its link to symptom-based endotypes is unknown. Therefore, we explored SGUS outcomes in relation to endotypes in patients with definite and suspected pSS. METHODS Definite pSS patients (n = 171) fulfilling the 2016 ACR/EULAR classification criteria, and suspected pSS patients (n = 119), positive for at least one criterion, were included in the Belgian Sjögren's Syndrome Transition Trial (BeSSTT). Stratification into endotypes according to the Newcastle Sjögren's Stratification Tool resulted in low symptom burden (LSB), pain dominant with fatigue (PDF), dryness dominant with fatigue (DDF) and high symptom burden (HSB). SGUS was assessed with Hocevar score (0-48). The dataset was randomly divided into a discovery (n = 203) and replication (n = 87) cohort. RESULTS SGUS had strong discriminative power for pSS classification (AUC=0.74), especially in DDF (AUC=0.89). In definite pSS, Hocevar scores in DDF were high compared to other endotypes (38 (20-44) versus 18 (9-33); p < 0.001). Patients with highest SGUS-scores showed more sicca and laboratory abnormalities. Moreover, a subset of young, anti-SSA/Ro positive patients not fulfilling classification criteria showed clear SGUS abnormalities. Replication showed similar results. CONCLUSIONS SGUS-scores were significantly higher in definite pSS with DDF endotype, providing the first evidence of imaging abnormalities in salivary glands matching distinct biological profiles ascribed to pSS endotypes. Additionally, a subset of patients with potential early disease was detected based on presence of anti-SSA antibodies and high SGUS-scores. These results underscore the role of SGUS as powerful tool both in pSS classification and stratification.
Collapse
|