To Örebro University

oru.seÖrebro University Publications
Change search
Refine search result
1 - 39 of 39
CiteExportLink to result list
Permanent link
Cite
Citation style
  • apa
  • ieee
  • modern-language-association-8th-edition
  • vancouver
  • Other style
More styles
Language
  • de-DE
  • en-GB
  • en-US
  • fi-FI
  • nn-NO
  • nn-NB
  • sv-SE
  • Other locale
More languages
Output format
  • html
  • text
  • asciidoc
  • rtf
Rows per page
  • 5
  • 10
  • 20
  • 50
  • 100
  • 250
Sort
  • Standard (Relevance)
  • Author A-Ö
  • Author Ö-A
  • Title A-Ö
  • Title Ö-A
  • Publication type A-Ö
  • Publication type Ö-A
  • Issued (Oldest first)
  • Issued (Newest first)
  • Created (Oldest first)
  • Created (Newest first)
  • Last updated (Oldest first)
  • Last updated (Newest first)
  • Disputation date (earliest first)
  • Disputation date (latest first)
  • Standard (Relevance)
  • Author A-Ö
  • Author Ö-A
  • Title A-Ö
  • Title Ö-A
  • Publication type A-Ö
  • Publication type Ö-A
  • Issued (Oldest first)
  • Issued (Newest first)
  • Created (Oldest first)
  • Created (Newest first)
  • Last updated (Oldest first)
  • Last updated (Newest first)
  • Disputation date (earliest first)
  • Disputation date (latest first)
Select
The maximal number of hits you can export is 250. When you want to export more records please use the Create feeds function.
  • 1.
    Andreoli, L.
    et al.
    University of Brescia | ASST Spedali Civili, Dept Clinical and Experimental Sciences | Unit of Rheumatology and Clinical Immunology, Brescia, Italy.
    Lini, D.
    University of Brescia | ASST Spedali Civili, Dept Clinical and Experimental Sciences | Unit of Rheumatology and Clinical Immunology, Brescia, Italy.
    Schreiber, K.
    Danish Hospital for Rheumatic Diseases, Rheumatology, Sønderborg, Denmark; University of Southern Denmark, Department of Regional Health Research (IRS), Odense, Denmark; Guys and St Thomas’ NHS Foundation Trust, Thrombosis and Hemostasis, London, United Kingdom.
    Sen, P.
    Maulana Azad Medical College, 2-Bahadurshah Zafar Marg, New Delhi, India.
    Ravichandran, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Stockholm, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Faculty of Medicine and Health, Örebro University, Department of Rheumatology, Örebro, Sweden.
    Toro Gutierrez, C. E.
    Pontifica Universidad Javeriana, Reference Center for Osteoporosis, Rheumatology and Dermatology, Cali, Colombia.
    Katchamart, W.
    Mahidol University, Division of Rheumatology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Bangkok, Thailand.
    Goo, P. A.
    Queen Savang Vadhana Memorial Hospital, Department of Medicine, Chonburi, Thailand.
    Shumnalieva, R.
    University Hospital “St. Ivan Rilski”, Department of Rheumatology, Sofia, Bulgaria.
    Chibuzo, O. C.
    University of Nigeria Teaching Hospital, Ituku-Ozalla/University of Nigeria, Enugu Campus, Department of Medicine, Enugu, Nigeria.
    Velikova, T.
    Sofia University St. Kliment Ohridski, Medical Faculty, Sofia, Bulgaria.
    Day, J.
    Institute of Medical Research, Walter and Eliza Hall, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia; Royal Melbourne Hospital, Department of Rheumatology, Parkville, Australia.
    Joshi, M.
    Byramjee Jeejeebhoy Government Medical College, Sassoon General Hospitals, Pune, India.
    Katsuyuki Shinjo, S.
    Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Division of Rheumatology, São Paulo, Brazil.
    Gracia-Ramos, A. E.
    National Medical Center “La Raza”, Instituto Mexicano del Seguro Social, Department of Internal Medicine, General Hospital, Mexico City, Mexico.
    Cavagna, L.
    University of Pavia, Rheumatology Unit, Dipartimento di Medicine Interna e Terapia Medica, Pavia, Italy.
    Kuwana, M.
    Nippon Medical School Graduate School of Medicine, Department of Allergy and Rheumatology, Tokyo, Japan.
    Knitza, J.
    Friedrich-Alexander-Universität Erlangen-Nürnberg, Medizinische Klinik 3 - Rheumatologie und Immunologie, Universitätsklinikum Erlangen, Erlangen, Germany.
    Makol, A.
    Mayo Clinic, Division of Rheumatology, Rochester, United States of America.
    Chen, Y. M.
    Taichung Veterans General Hospital, Division of Allergy, Immunology and Rheumatology, Department of Medical Research, Taichung, Taiwan, Republic of China.
    Chinoy, H.
    School of Biological Sciences, The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, Manchester, United Kingdom; Manchester University NHS Foundation Trust, National Institute for Health Research Manchester Biomedical Research Centre, Manchester, United Kingdom; Manchester Academic Health Science Centre, Department of Rheumatology, Salford Royal NHS Foundation Trust, Salford, United Kingdom.
    Agarwal, V.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, United States of America.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; New Cross Hospital, Royal Wolverhampton Trust, Department of Rheumatology, Wolverhampton, United Kingdom.
    COVID-19 VACCINE SAFETY DURING PREGNANCY AND BREASTFEEDING IN WOMEN WITH AUTOIMMUNE DISEASES: RESULTS FROM THE COVAD STUDY2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 56-57, article id OP0082Article in journal (Other academic)
    Abstract [en]

    Background: COVID-19 vaccine hesitancy among pregnant and breastfeeding women with autoimmune diseases (AID) is often attributed to the fear of adverse events (AE) and disease flares (DF). No data are available regarding COVID-19 vaccine safety in this population.

    Objectives: We aimed at describing delayed-onset (>7 days) vaccine-related AE (minor and major), DF, and related AID treatment modifications from the COVID-19 Vaccination in Autoimmune Diseases (COVAD) study.

    Methods: Among complete responses from 9201 participants as of June 21, 2022, 6787 (73.8%) were women. Six subgroups were identified upon diagnosis of AID vs healthy controls (HC) and their pregnancy/breastfeeding status at the time of any dose of vaccine (Figure 1).

    Results: Forty pregnant and 52 breastfeeding AID patients were identified and their vaccination rates (at least one dose) was 100% and 96.2%, respectively (Table 1). Overall AE, minor AE, and major AE were reported significantly more frequently by pregnant than non-pregnant patients (45% vs. 26%, p=0.01; 40% vs. 25.9%, p=0.03; 17.5% vs. 4.6%, p<0.01), but no difference was found in comparison with pregnant HC. No difference was observed between breastfeeding patients and HC. Post-vaccination DF were reported by 17.5% of pregnant and 20% of breastfeeding patients, and by 18% of age- and disease-matched control patients (n=2315). All DF in pregnant/breastfeeding patients were managed with glucocorticoids and a fifth of them required initiation or change in immunosuppressive treatment.

    Conclusion: This study provides the first insights into the safety of COVID-19 vaccination during the antenatal period in women with AID. While AEs were more commonly reported by pregnant patients with AID, these were no higher than among pregnant healthy controls without AID. These observations are reassuring, likely to strengthen physician-patient communication and overcome hesitancy as the benefits for the mother and fetus by passive immunization are likely to overweigh the potential risks of AE and DF.

    Reference: [1]Fazal ZZ, et al; COVAD Study Group. COVAD survey 2 long-term outcomes: unmet need and protocol. Rheumatol Int 2022; 42:2151-2158.

  • 2.
    Aoude, M.
    et al.
    Saint Joseph University of Beirut, Rheumatology Department, Beirut, Lebanon.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; The Royal Wolverhampton NHS Trust, Dept. of Rheumatology, Wolverhampton, United Kingdom.
    Hmamouchi, I.
    Temara Hospital, Rheumatology Unit, Temara, Morocco; Faculty of Medicine and Pharmacy, Mohammed V University, Laboratory of Biostatistics, Clinical Research and Epidemiology (LBRCE), Rabat, Morocco.
    Grignaschi, S.
    Fondazione I.R.C.C.S. Policlinico San Matteo, Rheumatology, Pavia, Italy; The University of Pavia, Department of Internal Medicine and Medical Therapeutics, Pavia, Italy.
    Cavagna, L.
    Fondazione I.R.C.C.S. Policlinico San Matteo, Rheumatology, Pavia, Italy; The University of Pavia, Department of Internal Medicine and Medical Therapeutics, Pavia, Italy.
    Kim, M.
    University of Illinois College of Medicine at Peoria, Department of Internal Medicine, Peoria, United States of America.
    R, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Lilleker, J. B.
    School of Biological Sciences University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Rochester, United Kingdom; Manchester Centre for Clinical Neurosciences, Clinical Neurosciences, Salford, United Kingdom.
    Sen, P.
    Maulana Azad Medical College(MAMC), Rheumatology, New Delhi, India.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, Navi Mumbai, India.
    Kardes, S.
    Istanbul University Faculty of Medicine, Department of Medical Ecology and Hydroclimatology, Istanbul, Turkey.
    Day, J.
    Royal Melbourne Hospital Neuroscience Foundation, Department of Rheumatology, Parkville, Australia; WEHI - Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia.
    Makol, A.
    Mayo Clinic, Division of Rheumatology, Rochester, United States of America.
    Milchert, M.
    Pomeranian Medical University, Department of Rheumatology, Internal Medicine, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Gheita, T. A.
    Faculty Of Medicine Kasr Al-Ainy Cairo University, Rheumatology Department, Cairo, Egypt.
    Salim, B.
    Fauji Foundation Hospital Road, Rheumatology Department, Rawalpindi, Pakistan.
    Velikova, T.
    Lozenetz University Hospital, Department of Clinical Immunology, Sofia, Bulgaria.
    Gracia-Ramos, A. E.
    MSS, Department of Internal Medicine, Ciudad de México, Mexico.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska University Hospital, Division of Rheumatology, Stockholm, Sweden; Örebro University, Department of Rheumatology, Örebro, Sweden.
    Selva-O'callaghan, A.
    La Vall d’Hebron, Systemic Autoimmune Diseases Unit, Internal Medicine Department, Barcelona, Spain.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Dept of Rheumatology, London, United Kingdom.
    Chatterjee, T.
    University of Illinois College of Medicine at Peoria, Department of Internal Medicine, Peoria, United States of America.
    Tan, A. L.
    Leeds General Infirmary, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom; University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom.
    Saavedra, M. A.
    Centro Medico Nacional La Raza, Departamento de Reumatología, Ciudad de México, Mexico.
    Shinjo, S. Katsuyuki
    Centro Medico Nacional La Raza, Departamento de Reumatología, Ciudad de México, Mexico.
    Knitza, J.
    University of Erlangen-Nuremberg, Medizinische Klinik 3 - Rheumatologie und Immunologie, Erlangen, Germany.
    Kuwana, M.
    Nippon Medical School, Department of Allergy and Rheumatology, Bunkyo City, Japan.
    Nune, A.
    Southport & Ormskirk Hospital NHS Trust, Southport, United Kingdom.
    Distler, O.
    University Hospital of Zürich, Rheumatology, Zürich, Switzerland.
    Chinoy, H.
    School of Biological Sciences University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Manchester, United Kingdom; NIHR Manchester Biomedical Research Unit, Manchester, United Kingdom; Salford Royal NHS Foundation Trust, Rheumatology, Manchester, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, United States of America.
    Ziade, N.
    Saint Joseph University of Beirut, Rheumatology Department, Beirut, Lebanon; Hôtel-Dieu de France, Rheumatology Department, Beirut, Lebanon.
    TREATMENT PATTERNS OF IDIOPATHIC INFLAMMATORY MYOPATHIES: RESULTS FROM AN INTERNATIONAL COHORT OF OVER 1,400 PATIENTS2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 105-106Article in journal (Other academic)
  • 3.
    Arkema, E. V.
    et al.
    Department of Medicine Solna, Clinical Epidemiology Unit, Karolinska Institutet, Stockholm, Sweden.
    Rossides, M.
    Department of Medicine Solna, Clinical Epidemiology Unit, Karolinska Institutet, Stockholm, Sweden.
    von Euler, Mia
    Clinical Science and Education and Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
    Svenungsson, E.
    Department of Medicine Solna, Rheumatology Unit, Karolinska Institutet, Stockholm, Sweden.
    Sjöwall, C.
    Department of Clinical and Experimental Medicine, Rheumatology/Division of Neuro and Inflammation Sciences, Linköping University, Linköping, Sweden.
    Simard, J. F.
    Department of Medicine Solna, Clinical Epidemiology Unit, Karolinska Institutet, Stockholm, Sweden; Department of Health Research and Policy, Division of Epidemiology, Stanford School of Medicine, Stanford, CA, United States; Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, CA, United States.
    Response to: 'Increased stroke incidence in systemic lupus erythematosus patients: Risk factors or disease itself?' by Bruzzese and Zullo2017In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 77, no 10, article id e72Article in journal (Other academic)
  • 4.
    Arkema, Elizabeth V.
    et al.
    Department of Medicine Solna, Clinical Epidemiology Unit, Karolinska Institutet, Solna, Sweden.
    Svenungsson, Elisabet
    Department of Medicine Solna, Rheumatology Unit, Karolinska Institutet, Stockholm, Sweden.
    von Euler, Mia
    Department of Clinical Science and Education, Karolinska Institutet, Solna, Sweden; Stroke Research Network at Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.
    Sjöwall, Christopher
    Clinical and Experimental Medicine, Rheumatology/Division of Neuro and Inflammation Sciences, Linköping University, Linköping, Sweden.
    Simard, Julia F.
    Department of Medicine Solna, Clinical Epidemiology Unit, Karolinska Institutet, Solna, Sweden; Department of Health Research and Policy, Division of Epidemiology, Stanford School of Medicine, Stanford, California, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford School of Medicine, Stanford, California, USA.
    Stroke in systemic lupus erythematosus: a Swedish population-based cohort study2017In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 76, no 9, p. 1544-1549Article in journal (Refereed)
    Abstract [en]

    OBJECTIVE: To study the occurrence of ischaemic and haemorrhagic stroke in systemic lupus erythematosus (SLE) compared with the general population by age, sex and time since SLE diagnosis METHODS: Adults with incident SLE were identified from the Swedish National Patient Register (NPR, n=3390) and general population comparators from the Total Population Register were matched on age, sex and county (n=16730). Individuals were followed prospectively until first of death, December 2013, emigration or incident stroke (identified from the NPR, Cause of Death Register and the Stroke Register). Incidence rates, rate differences and HR were estimated comparing SLE with non-SLE. Estimates were stratified by sex, age and time since diagnosis.

    RESULTS: We observed 126 strokes in SLE and 304 in the general population. Individuals with SLE had a twofold increased rate of ischaemic stroke compared with the general population (HR 2.2; 95% CI 1.7 to 2.8). The HR for intracerebral haemorrhage was 1.4 (95% CI 0.7 to 2.8). There was effect modification by sex and age, with the highest HRs for females and individuals <50 years old. The HR for ischaemic stroke was highest in the first year of follow-up (3.7; 95% CI 2.1 to 6.5).

    CONCLUSIONS: The relative risk of ischaemic stroke in SLE was more than doubled compared with the general population, and importantly, the highest relative risks were observed within the first year after SLE diagnosis. Thus, the first encounter with patients presents an opportunity for rheumatologists to screen for risk factors and intervene.

  • 5.
    Brolin, S.
    et al.
    Karolinska University Hospital, Department of Gastroenterology, Dermatology and Rheumatology, Stockholm, Sweden.
    Lövström, B.
    Karolinska University Hospital, Department of Gastroenterology, Dermatology and Rheumatology, Stockholm, Sweden.
    Welin, Elisabet
    Örebro University, School of Health Sciences.
    Gunnarsson, I.
    Karolinska University Hospital, Department of Gastroenterology, Dermatology and Rheumatology, Stockholm, Sweden; Karolinska Institutet, Rheumatology, Stockholm, Sweden.
    Pettersson, S.
    Karolinska University Hospital, Department of Gastroenterology, Dermatology and Rheumatology, Stockholm, Sweden.
    THE NEED FOR INFORMATION AMONG PATIENTS WITH ANCA ASSOCIATED VASCULITIS DIFFERS BETWEEN GROUPS2021In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 80, no Suppl. 1, p. 1023-1023, article id POS1476-HPArticle in journal (Other academic)
  • 6. Carli, C.
    et al.
    Ehlin, A. G. C.
    Klareskog, L.
    Lindblad, S.
    Montgomery, Scott M.
    Örebro University, Department of Clinical Medicine.
    Trends in disease modifying antirheumatic drug prescription in early rheumatoid arthritis are influenced more by hospital setting than patient or disease characteristics2006In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 65, no 8, p. 1102-1105Article in journal (Refereed)
    Abstract [en]

    Objective: To characterise temporal trends and factors associated with the prescription of disease modifying antirheumatic drugs (DMARDs) at the initial consultation in early rheumatoid arthritis (RA).

    Methods: Data from 2584 patients with early RA at 19 hospitals were extracted from the Swedish Rheumatoid Arthritis Register for the period 1997–2001. Disease characteristics and DMARD prescription at first consultation with the rheumatologist were investigated using cross tabulation and logistic regression.

    Results: DMARD prescriptions, particularly for methotrexate, increased from 1997 to 2001 independently of patient characteristics. Stratification by hospital type showed that patients in district hospitals were less likely to be prescribed DMARDs than those in university hospitals (adjusted odds ratio (OR) = 0.53 (95% confidence interval (CI) 0.40 to 0.69), p<0.001), independently of confounding factors. Association of the DAS28 with the likelihood of DMARD prescription was greater among patients attending district hospitals (OR = 1.65 (1.34 to 2.02), p<0.001) than those at university hospitals (OR = 1.23 (1.07 to 1.41), p = 0.003) and county hospitals (OR = 1.34 (1.01 to 1.63), p = 0.003). Interaction testing indicated that the difference was significant (p = 0.007).

    Conclusions: Temporal trends in DMARD prescription indicate an increasingly aggressive approach to disease management among Swedish rheumatologists. However, the association of hospital type with DMARD prescription suggests that the adoption of research findings in clinical care varies considerably.

  • 7.
    Emamikia, S.
    et al.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine, Solna, Sweden.
    Gentline, C.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine, Solna, Sweden.
    Enman, Y.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine, Solna, Sweden.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine, Solna, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    CAN WE ENHANCE ADHERENCE TO MEDICATIONS IN PATIENTS WITH SYSTEMIC LUPUS ERYTHEMATOSUS? RESULTS FROM A QUALITATIVE STUDY2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 439-439, article id POS0371Article in journal (Other academic)
  • 8.
    Fanouriakis, Antonis
    et al.
    Rheumatology and Clinical Immunology Unit, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece.
    Kostopoulou, Myrto
    Rheumatology and Clinical Immunology Unit, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece.
    Andersen, Jeanette
    Lupus Europe, Copenhagen, Denmark.
    Aringer, Martin
    Division of Rheumatology, Department of Medicine III, University Medical Center & Faculty of Medicine Carl Gustav Carus at the TU Dresden, Dresden, Germany.
    Arnaud, Laurent
    Department of Rheumatology, Hôpitaux Universitaires de Strasbourg, INSERM UMR-S 1109, Centre National de Référence des Maladies Auto-immunes Systémiques Rares (RESO), Strasbourg, France.
    Bae, Sang-Cheol
    Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Hanyang University Institute for Rheumatology Research and Hanyang Institute of Bioscience and Biotechnology, Seoul, South Korea.
    Boletis, John
    Department of Nephrology and Renal Transplantation Unit, "Laiko" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
    Bruce, Ian N.
    Centre for Epidemiology Versus Arthritis, University of Manchester, Manchester, UK; National Institute for Health Research Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
    Cervera, Ricard
    Department of Autoimmune Diseases, Hospital Clinic, Barcelona, Spain.
    Doria, Andrea
    Rheumatology Unit, Department of Medicine, University of Padova, Padova, Italy.
    Dörner, Thomas
    Department of Rheumatology and Clinical Immunology, Charite Universitätsmedizin Berlin; Deutsches Rheumaforschungszentrum, Berlin, Germany.
    Furie, Richard A.
    Division of Rheumatology, Northwell Health, Great Neck, New York City, New York, USA.
    Gladman, Dafna D.
    Lupus Program, Centre for Prognosis Studies in the Rheumatic Disease, Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada.
    Houssiau, Frederic A.
    Service de Rhumatologie, Cliniques Universitaires Saint-Luc and Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium.
    Inês, Luís Sousa
    Department of Rheumatology, Centro Hospitalar e Universitario de Coimbra, Coimbra, Portugal; School of Health Sciences, Universidade da Beira Interior, Covilha, Portugal.
    Jayne, David
    Department of Medicine, University of Cambridge, Cambridge, UK.
    Kouloumas, Marios
    Cyprus League Against Rheumatism, Aglantzia, Cyprus.
    Kovács, László
    Department of Rheumatology and Immunology, Faculty of Medicine, University of Szeged, Hungary.
    Mok, Chi Chiu
    Department of Medicine, Tuen Mun Hospital, Hong Kong, China.
    Morand, Eric F.
    Centre for Inflammatory Diseases, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia.
    Moroni, Gabriella
    Department of Biomedical Sciences Humanitas University, Nephrology and Dialysis Division, IRCCS Humanitas Research Hospital, Milan, Italy.
    Mosca, Marta
    Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
    Mucke, Johanna
    Department of Rheumatology & Hiller Research Unit Rheumatology, UKD, Heinrich-Heine University, Düsseldorf, Germany.
    Mukhtyar, Chetan B.
    Vasculitis Service, Rheumatology Department, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK.
    Nagy, György
    Hospital of the Hospitaller Order of Saint John of God, Budapest, Hungary; Department of Rheumatology and Clinical Immunology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary; Heart and Vascular Center, Semmelweis University, Budapest, Hungary.
    Navarra, Sandra
    Section of Rheumatology, Department of Medicine, University of Santo Tomas, Manila, Philippines.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden; Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
    Pego-Reigosa, José M.
    Rheumatology Department, Complejo Hospitalario Universitario de Vigo, IRIDIS (Investigation in Rheumatology and Immune-Mediated Diseases) - VIGO Group, Galicia Sur Health Research Institute, Vigo, Spain.
    Petri, Michelle
    Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
    Pons-Estel, Bernardo A.
    Grupo Oroño, Centro Regional de Enfermedades Autoinmunes y Reumáticas (GO-CREAR), Rosario, Argentina.
    Schneider, Matthias
    Department of Rheumatology & Hiller Research Unit Rheumatology, UKD, Heinrich-Heine University, Düsseldorf, Germany.
    Smolen, Josef S.
    Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria.
    Svenungsson, Elisabet
    Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
    Tanaka, Yoshiya
    First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan.
    Tektonidou, Maria G.
    Rheumatology Unit, First Department of Propaedeutic Internal Medicine, "Laiko" General Hospital, Medical School, National and Kapodistrian University of Athens, Joint Academic Rheumatology Program, Athens, Greece.
    Teng, Yk Onno
    Centre of Expertise for Lupus-, Vasculitis- and Complement-mediated Systemic autoimmune diseases, Department of Internal Medicine - section Nephrology, Leiden University Medical Center, Leiden, The Netherlands.
    Tincani, Angela
    Rheumatology and Clinical Immunology, Department of Clinical and Experimental Sciences, ASST Spedali Civili and University of Brescia, Brescia, Italy.
    Vital, Edward M.
    Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
    van Vollenhoven, Ronald F.
    Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Centers, Amsterdam, The Netherlands.
    Wincup, Chris
    Department of Rheumatology, King's College Hospital, London, UK.
    Bertsias, George
    Rheumatology, Clinical Immunology and Allergy, University Hospital of Heraklion, Greece, University Hospital of Heraklion, Heraklion, Greece.
    Boumpas, Dimitrios T.
    Rheumatology and Clinical Immunology Unit, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece; Laboratory of Autoimmunity and Inflammation, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Joint Academic Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Greece, Medical School, University of Cyprus, Nicosia, Cyprus.
    EULAR recommendations for the management of systemic lupus erythematosus: 2023 update2024In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 83, no 1, p. 15-29Article in journal (Refereed)
    Abstract [en]

    OBJECTIVES: To update the EULAR recommendations for the management of systemic lupus erythematosus (SLE) based on emerging new evidence.

    METHODS: An international Task Force formed the questions for the systematic literature reviews (January 2018-December 2022), followed by formulation and finalisation of the statements after a series of meetings. A predefined voting process was applied to each overarching principle and recommendation. Levels of evidence and strengths of recommendation were assigned, and participants finally provided their level of agreement with each item.

    RESULTS: The Task Force agreed on 5 overarching principles and 13 recommendations, concerning the use of hydroxychloroquine (HCQ), glucocorticoids (GC), immunosuppressive drugs (ISDs) (including methotrexate, mycophenolate, azathioprine, cyclophosphamide (CYC)), calcineurin inhibitors (CNIs, cyclosporine, tacrolimus, voclosporin) and biologics (belimumab, anifrolumab, rituximab). Advice is also provided on treatment strategies and targets of therapy, assessment of response, combination and sequential therapies, and tapering of therapy. HCQ is recommended for all patients with lupus at a target dose 5 mg/kg real body weight/day, considering the individual's risk for flares and retinal toxicity. GC are used as 'bridging therapy' during periods of disease activity; for maintenance treatment, they should be minimised to equal or less than 5 mg/day (prednisone equivalent) and, when possible, withdrawn. Prompt initiation of ISDs (methotrexate, azathioprine, mycophenolate) and/or biological agents (anifrolumab, belimumab) should be considered to control the disease and facilitate GC tapering/discontinuation. CYC and rituximab should be considered in organ-threatening and refractory disease, respectively. For active lupus nephritis, GC, mycophenolate or low-dose intravenous CYC are recommended as anchor drugs, and add-on therapy with belimumab or CNIs (voclosporin or tacrolimus) should be considered. Updated specific recommendations are also provided for cutaneous, neuropsychiatric and haematological disease, SLE-associated antiphospholipid syndrome, kidney protection, as well as preventative measures for infections, osteoporosis, cardiovascular disease.

    CONCLUSION: The updated recommendations provide consensus guidance on the management of SLE, combining evidence and expert opinion.

  • 9.
    Fornaro, M.
    et al.
    Rheumatology Unit, Department of Precision and Regenerative Medicine and Ionian Area, Bari, Italy.
    Venerito, V.
    Rheumatology Unit, Department of Precision and Regenerative Medicine and Ionian Area, Bari, Italy.
    Iannone, F.
    Rheumatology Unit, Department of Precision and Regenerative Medicine and Ionian Area, Bari, Italy.
    Ravichandran, N.
    Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.
    Nikiphorou, E.
    Centre for Rheumatic Diseases, King’s College London, London, United Kingdom; Rheumatology Department, King’s College Hospital, London, United Kingdom.
    Joshi, M.
    Byramjee Jeejeebhoy Government Medical College and Sassoon, General Hospitals, Pune, India.
    Tan, A. L.
    NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals Trust, Leeds, United Kingdom; Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.
    Saha, S.
    Mymensingh Medical College, Mymensingh, Bangladesh, Bangladesh.
    Shinjo, S. Katsuyuki
    Division of Rheumatology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, Navi Mumbai, Maharashtra, India.
    Ziade, N.
    Mahatma Gandhi Mission Medical College, Navi Mumbai, Maharashtra, India.
    Velikova, T.
    Department of Clinical Immunology, Medical Faculty, University Hospital “Lozenetz”, Sofia University St. Kliment Ohridski, Sofia, Bulgaria.
    Jagtap, K.
    Seth Gordhandhas Sunderdas Medical College and King Edwards Memorial Hospital, Mumbai, Maharashtra, India.
    Milchert, M.
    Department of Internal Medicine, Rheumatology, Diabetology, Geriatrics and Clinical Immunology, Omeranian Medical University in Szczecin, Szczecin, Poland.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden; Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
    Gracia-Ramos, A. E.
    Department of Internal Medicine, General Hospital, National Medical Center “La Raza”, Instituto Mexicano del Seguro Social, Av. Jacaranda S/N, Col. La Raza, Del. Azcapotzalco, Mexico City, Mexico.
    Cavagna, L.
    Rheumatology Unit, Dipartimento di Medicine Interna e Terapia Medica, Università degli studi di Pavia, Pavia, Italy.
    Kuwana, M.
    Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan.
    Knitza, J.
    Medizinische Klinik 3 – Rheumatologie und Immunologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
    Makol, A.
    Division of Rheumatology, Mayo Clinic, Rochester Minnesota, United States of America.
    Dzifa, D.
    Department of Medicine and Therapeutics, University of Ghana School of Medicine and Dentistry, College of Health Sciences, Korle-Bu, Accra, Ghana.
    Toro Gutierrez, C. E.
    Reference Center for Osteoporosis, Rheumatology and Dermatology, Pontifica Universidad Javeriana, Cali, Colombia.
    Vinicio Caballero, C.
    Department of Medicine, Hospital Universidad del Norte, Barranquilla, Atlantico, Colombia.
    Distler, O.
    Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
    Day, J.
    Department of Rheumatology, Royal Melbourne Hospital, Parkville VIC, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville VIC, Australia.
    Chinoy, H.
    Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom; National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom; Department of Rheumatology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, United Kingdom.
    Aggarwal, R.
    Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh Pennsylvania, United States of America.
    Gupta, L.
    Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, United Kingdom; City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, United Kingdom.
    MULTIMORBIDITY AND PROMIS HEALTH OUTCOMES IN PATIENTS WITH IDIOPATHIC INFLAMMATORY MYOPATHIES: DATA FROM A LARGE, GLOBAL E-SURVEY (COVAD STUDY)2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 942-943, article id POS1216Article in journal (Other academic)
    Abstract [en]

    Background: Prevalence of comorbidities and their impact on health outcomes in Idiopathic inflammatory myopathies (IIMs) is limited.

    Objectives: This study aimed to explore the prevalence of multimorbidity in patients with IIMs, other autoimmune rheumatic diseases (AIRDs) and Healthy controls (HCs). We further explore the impact of comorbidities on patients’ physical, mental, and social health assessed by the Patient-Reported Outcome Measurement Information System (PROMIS instruments).

    Methods: Data for this study were acquired from the COVAD 2 e-survey hosted by a study group consisting of 167 collaborators in 110 countries. Basic multimorbidity (BM) was defined as the co-occurrence of two or more comorbidities in an individual, while complex multimorbidity (CM) signified the co-occurrence of 3 or more chronic conditions affecting 3 or more different organ systems. PROMIS global physical health (PGP), mental health (PGM), fatigue 4a (F4a) and physical function short form (SF10) were analysed using descriptive statistics and linear regression models. Hierarchical Clustering on Principal Components was performed to outline the grouping.

    Results: Of 10740 complete respondents, 1558 IIMs, 4591 AIRDs and 3652 HCs were analysed. Individuals with IIMs exhibited high burden of any comorbidity (OR: 1.62 vs AIRDs and 2.95 vs HCs,p<0.01), BM (OR 1.66 vs AIRDs and 3.52 vs HCs,p<0.01), CM (OR: 1.69 vs AIRDs and 6.23 vs HCs,p<0.01), and mental health disorders (MHDs) (OR 1.33 vs AIRDs and 2.63 vs HCs,p<0.01).

    IIM patients with comorbidities (and MHDs) had worse physical function (low PGP, PGM, SF10 and higher F4a scores, all p<0.001). Worse physical function (PGP) was predicted by age (0.35; 0.030), active disease (-1.51; <0.001), BM (-1.11; <0.001), and MHDs (-1.47; <0.001). PGM was impacted by age (0.51; 0.004), active disease (-1.34, <0.001), BM (-0.75; 0.001) and MHDs (-2.22; <0.001). Determinants of SF10a were age (-3.86; <0.001), active disease (-7.03, <0.001), female (2.85, <0.001), BM (-2.95; <0.001) and MHDs (-2.37; <0.001). Fatigue (F4a) was impacted by age (-0.96, <0.001), active disease (1.45, <0.001), country human development index (0.95; 0.036), BM (1.11; <0.001); and MHDs (2.17; <0.001).

    Four distinct clusters (Figure 1A, Table 1) were identified i.e., cluster 0: lower burden of comorbidities and good health status; cluster 1: older patients, whit higher burden of comorbidities and poor health status, cluster 2: patients with higher prevalence of MHDs, lower PGP and PGM; and higher F4a scores; and lastly Cluster 3 that comprised older patients with an average burden of comorbidities and overall good health status according to PROMIS scores.

    Dermatomyositis, anti-synthetase syndrome, necrotizing autoimmune myopathy were similarly represented in all clusters, whilst inclusion body myositis and polymyositis were more predominant in clusters 1 (40.6% and 17.2%) and 3 (32 % and 17.5%), while overlap myositis was more represented in cluster 2 (25.6%) and 0 (32.7%) (Figure 1B).

    Conclusion: Patients with IIMs have a higher burden of comorbidities that adversely impact physical and mental health, calling for optimized approaches for holistic patient management.

  • 10.
    Giannopoulou, N.
    et al.
    University of Nicosia, General Hospital of Paphos, Nicosia, Cyprus.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; Royal Wolverhampton Hospitals NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom; City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Department of Rheumatology, Birmingham, United Kingdom; The University of Manchester, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester, United Kingdom.
    Andreoli, L.
    Azienda Socio-Sanitaria Territoriale Spedali Civili, Rheumatology and Clinical Immunology Unit, Brescia, Italy; University of Brescia, Department of Clinical and Experimental Sciences, Brescia, Italy.
    Lini, D.
    Azienda Socio-Sanitaria Territoriale Spedali Civili, Rheumatology and Clinical Immunology Unit, Brescia, Italy; University of Brescia, Department of Clinical and Experimental Sciences, Brescia, Italy.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Rheumatology Department, London, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Department of Medicine, Pittsburgh, United States of America.
    Agarwal, V.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    COVID-19 VACCINE SAFETY DURING PREGNANCY IN WOMEN WITH SYSTEMIC LUPUS ERYTHEMATOSUS2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 1495-1496, article id AB0591Article in journal (Other academic)
    Abstract [en]

    Background: Vaccinations comprise a part of the antenatal care of pregnant women, including patients with systemic lupus erythematosus (SLE) who are at increased risk of adverse pregnancy outcomes (APOs). While COVID-19 vaccination has been shown to be safe in patients with SLE, data on vaccine-associated adverse events (AEs) during the antenatal and lactation period are scarce or lacking.

    Objectives: To investigate the association between COVID-19 vaccination and AEs in pregnant SLE patients.

    Methods: A total of 9201 complete responses were extracted on June 21st, 2022 from the COVID-19 Vaccination in Autoimmune Diseases (COVAD) 2 database, a global e-survey involving 157 collaborators from 106 countries. Among respondents, 6787 (73.8%) were women. We identified 70 (1.1%) women who were exposed to at least one COVID-19 vaccine dose during pregnancy, among those 11 with SLE. Delayed onset (>7 days) vaccine-related AEs were extracted and triangulated with disease activity, treatment changes due to flare after vaccination, and COVID-19 infections in vaccinated pregnant women with SLE. Additionally, information on health-related quality of life and physical function was recorded using PROMIS at the time of survey completion.

    Results: The age of patients ranged from 28 to 39 years; 5/11 women were of Asian origin. None of these patients reported major vaccine AEs, including four patients with self-reported active SLE prior to the vaccination. None of them reported any change in the status of their autoimmune disease, and no hospitalisation or special treatment was recorded. Six women experienced minor vaccine AEs; two of them had active disease prior to vaccination. Four patients reported COVID-19 infection; two of them while they were pregnant and post-vaccination and two prior to pregnancy and vaccination. All four patients experienced symptoms of their disease, but no overt SLE flare was reported. At the time of survey completion, all patients reported their general health as being good to excellent in all aspects evaluated. Importantly, no APOs were reported.

    None of the patients reported thrombotic events post-vaccination, which provides some reassurance regarding COVID-19 vaccination in a patient population with a high risk for cardiovascular comorbidity and thrombosis, especially in the presence of antiphospholipid antibodies or in patients diagnosed with the antiphospholipid syndrome, a considerable portion within SLE populations. Moreover, it was reassuring to note an absence of association between experienced vaccine AEs and active disease prior to vaccination. Although minor AEs were common, they did not impair daily functioning, and the symptoms resolved in all patients after a median of 3 (IQR: 2.5–5.0) days.

    Conclusion: Our report adds relevant evidence concerning the sensitive issue of COVID-19 vaccine AEs and flares in SLE patients during the antenatal and lactation period. Despite the small sample size, the findings provide some reassurance and can contribute to informed decisions regarding vaccination in patients with SLE and high-risk pregnancies due to their background autoimmune disease. Based on the present data, the risk/benefit ration of COVID-19 vaccination appears favourable, with vaccines both providing passive immunisation to the fetus and active immunisation to the mother with no signals of exacerbation of the mother’s autoimmune disease.

  • 11.
    Gomez, A.
    et al.
    Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Karolinska University Hospital, Medical Unit of Gastroenterology, Dermatology, and Rheumatology, Stockholm, Sweden.
    Jägerback, S.
    Danderyd University Hospital, Division of Rheumatology, Stockholm, Sweden.
    Sjowall, C.
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping, Sweden.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Karolinska University Hospital, Medical Unit of Gastroenterology, Dermatology, and Rheumatology, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    LOW-DOSE BELIMUMAB AND ANTIMALARIAL AGENTS PREVENT RENAL FLARES IN SYSTEMIC LUPUS ERYTHEMATOSUS: RESULTS FROM FOUR RANDOMISED CLINICAL TRIALS2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 1467-1468, article id AB0538Article in journal (Other academic)
    Abstract [en]

    Background: Lupus nephritis (LN) is one of the most severe manifestations in systemic lupus erythematosus (SLE), constituting a substantial cause of end-stage kidney disease, dialysis, and mortality. Prompt and adequate treatment of LN, and prevention of renal flares are key components of disease management towards improved outcomes in patients with SLE.

    Objectives: We aimed to determine the effect of the use of antimalarial agents (AMA) and different doses and pharmaceutical forms of belimumab on preventing renal flares in patients with active SLE.

    Methods: We pooled data from the BLISS-52, BLISS-76, BLISS-SC and BLISS-Northeast Asia randomised clinical trials of belimumab (N=3225), that included patients with seropositive (antinuclear antibody titres ≥1:80 and/or anti-dsDNA levels ≥30 IU/mL), active SLE yet no severe ongoing renal disease. Participants were allocated to receive intravenous (IV) belimumab 1 mg/kg (N=559), IV belimumab 10 mg/kg (N=1033), subcutaneous (SC) belimumab 200 mg (N=556) or placebo (N=1077) in addition to standard therapy. Additionally, we classified patients as AMA users if they had received hydroxychloroquine, chloroquine, mepacrine, or quinine sulphate in stable doses for at least 30 days prior to the trial commencement. The outcome of the present post-hoc analysis was development of renal flares, defined according to the analysis plan within the BLISS programme. The hazard of renal flare was assessed with Cox proportional hazards regression models, adjusted for age, sex, ethnicity, previous renal involvement, baseline proteinuria and glomerular filtration rate, and use of glucocorticoids and immunosuppressants.

    Results: In total, 192 patients developed a renal flare after a median of 197 days. In multivariable Cox regression analysis, use of AMA was associated with a lower risk of renal flares (HR: 0.64; 95% CI: 0.54–0.96; p=0.026). Compared with placebo, the risk of renal flares was lower among patients receiving IV belimumab 1 mg/kg (HR: 0.44; 95% CI: 0.25–0.79; p=0.006) and IV belimumab 10 mg/kg (HR: 0.63; 95% CI: 0.45–0.87; p=0.005), but not SC belimumab 200 mg (HR: 0.90; 95% CI: 0.57–1.42; p=0.648). When analysing all study arms with and without antimalarials separately, patients receiving IV belimumab 1 mg/kg along with AMA experienced the lowest rate of renal flares (18.5 (7.4–38.1) cases per 1000 person-years). Using patients who received placebo but not AMA as the reference comparator, patients receiving IV belimumab 1 mg/kg (OR: 0.30; 95% CI: 0.13–0.70; p=0.005) and patients receiving IV belimumab 10 mg (OR: 0.45; 95% CI: 0.27–0.75; p=0.002) were protected against renal flares only when belimumab use was combined with AMA.

    Conclusion: In this RCT setting, belimumab and AMA protected against renal flares in patients with active seropositive SLE yet no ongoing severe renal involvement. The protective effect of IV belimumab against renal flares appeared optimal when belimumab was combined with AMA. The prominent effect of low-dose belimumab motivates investigation of the efficacy of intermediate doses of belimumab.

  • 12.
    Gomez, A.
    et al.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Sjöwall, C.
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping, Sweden.
    OBESITY AND TOBACCO SMOKING ARE INDEPENDENTLY ASSOCIATED WITH POOR PATIENT-REPORTED OUTCOMES IN PATIENTS WITH SYSTEMIC LUPUS ERYTHEMATOSUS FROM A SWEDISH TERTIARY REFERRAL CENTRE2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 1402-1402, article id AB0549Article in journal (Other academic)
  • 13.
    Grignaschi, S.
    et al.
    Fondazione I.R.C.C.S. Policlinico San Matteo, Rheumatology, Pavia, Italy; The University of Pavia, Department of Internal Medicine and Medical Therapeutics, Pavia, Italy.
    Cavagna, L.
    Fondazione I.R.C.C.S. Policlinico San Matteo, Rheumatology, Pavia, Italy; The University of Pavia, Department of Internal Medicine and Medical Therapeutics, Pavia, Italy.
    Kim, M.
    University of Illinois College of Medicine at Peoria, Department of Internal Medicine, Peoria, United States of America.
    R, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Lilleker, J. B.
    School of Biological Sciences University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Rochester, United Kingdom; Manchester Centre for Clinical Neurosciences, Clinical Neurosciences, Salford, United Kingdom.
    Sen, P.
    Maulana Azad Medical College (MAMC), Rheumatology, New Delhi, India.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, Navi Mumbai, India.
    Kardes, S.
    Istanbul University Faculty of Medicine, Department of Medical Ecology and Hydroclimatology, Istanbul, Turkey.
    Day, J.
    Royal Melbourne Hospital Neuroscience Foundation, Department of Rheumatology, Parkville, Australia; WEHI - Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia.
    Makol, A.
    Mayo Clinic, Division of Rheumatology, Rochester, United States of America.
    Milchert, M.
    Pomeranian Medical University, Department of Rheumatology, Internal Medicine, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Gheita, T. A.
    Faculty Of Medicine Kasr Al-Ainy Cairo University, Rheumatology Department, Cairo, Egypt.
    Salim, B.
    Fauji Foundation Hospital Road, Rheumatology Department, Rawalpindi, Pakistan.
    Velikova, T.
    Lozenetz University Hospital, Department of Clinical Immunology, Sofia, Bulgaria.
    Gracia-Ramos, A. E.
    IMSS, Department of Internal Medicine, Ciudad de México, Mexico.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska University Hospital, Division of Rheumatology, Stockholm, Sweden; Örebro University, Department of Rheumatology, Örebro, Sweden.
    Selva-O'callaghan, A.
    La Vall d’Hebron, Systemic Autoimmune Diseases Unit, Internal Medicine Department, Barcelona, Spain.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Dept of Rheumatology, London, United Kingdom.
    Chatterjee, T.
    University of Illinois College of Medicine at Peoria, Department of Internal Medicine, Peoria, United States of America.
    Tan, A. L.
    Leeds General Infirmary, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom; University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom.
    Saavedra, M. A.
    Centro Medico Nacional La Raza, Departamento de Reumatología, Ciudad de México, Mexico.
    Shinjo, S. Katsuyuki
    Faculdade de Medicina da Universidade de São Paulo, Division of Rheumatology, Sao Paulo, Brazil.
    Ziade, N.
    Saint Joseph University of Beirut, Rheumatology Department, Bayrut, Lebanon; Hôtel-Dieu de France, Rheumatology Department, Bayrut, Lebanon.
    Knitza, J.
    University of Erlangen-Nuremberg, Medizinische Klinik 3 - Rheumatologie und Immunologie, Erlangen, Germany.
    Kuwana, M.
    Nippon Medical School, Department of Allergy and Rheumatology, Bunkyo City, Japan.
    Nune, A.
    Southport & Ormskirk Hospital NHS Trust, Southport, United Kingdom.
    Distler, O.
    University Hospital of Zürich, Rheumatology, Zürich, Switzerland.
    Chinoy, H.
    School of Biological Sciences University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Manchester, United Kingdom; NIHR Manchester Biomedical Research Unit, Manchester, United Kingdom; Salford Royal NHS Foundation Trust, Rheumatology, Manchester, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, United States of America.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; The Royal Wolverhampton NHS Trust, Dept. of Rheumatology, Wolverhampton, United Kingdom.
    HIGH FATIGUE SCORES IN PATIENTS WITH IDIOPATHIC INFLAMMATORY MYOPATHIES: A MULTIGROUP COMPARATIVE STUDY FROM THE COVAD E-SURVEY2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 748-748, article id POS0899Article in journal (Other academic)
  • 14.
    Gupta, L.
    et al.
    Royal Wolverhampton Hospitals NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom; Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Hoff, L. S.
    Universidade de Sao Paulo Faculdade de Medicina FMUSP, Division of Rheumatology, Sao Paulo, Brazil.
    R, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Sen, P.
    Maulana Azad Medical College, Undergraduate, New Delhi, India.
    Katsuyuki Shinjo, S.
    Universidade de Sao Paulo Faculdade de Medicina FMUSP, Division of Rheumatology, Sao Paulo, Brazil.
    Day, J.
    Royal Melbourne Hospital, Department of Rheumatology, Parkville, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia.
    Lilleker, J. B.
    School of Biological Sciences, The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Manchester, United Kingdom; Manchester Centre for Clinical Neurosciences, Neurology, Salford, United Kingdom.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, Navi Mumbai, India.
    Kardes, S.
    Istanbul University Faculty of Medicine, Department of Medical Ecology and Hydroclimatology, Istanbul, Turkey.
    Kim, M.
    University of Illinois College of Medicine at Peoria, Department of Internal Medicine, Peoria, United States of America.
    Makol, A.
    Mayo Clinic, Division of Rheumatology, Rochester, United States of America.
    Milchert, M.
    Pomeranian Medical University in Szczecin, Department of Internal Medicine, Rheumatology, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Gheita, T. A.
    Kasr Al Ainy School of Medicine, Cairo University, Department of Rheumatology, Cairo, Egypt.
    Salim, B.
    Fauji Foundation Hospital, Department of Clinical Immunology, Rawalpindi, Pakistan.
    Velikova, T.
    University Hospital “Lozenetz”, Sofia University, Department of Clinical Immunology, Sofia, Bulgaria.
    Gracia-Ramos, A. E.
    General Hospital, National Medical Center “La Raza”, Instituto Mexicano del Seguro Social, Department of Internal Medicine, Mexico City, Mexico.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Stockholm, Sweden; Örebro University Faculty of Medicine and Health, Department of Rheumatology, Örebro, Sweden.
    Selva-O'callaghan, A.
    Vall D’hebron General Hospital, Universitat Autonoma de Barcelona, Department of Internal Medicine, Barcelona, Spain.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Department of Rheumatology, London, United Kingdom.
    Chatterjee, T.
    University of Illinois College of Medicine at Peoria, Department of Internal Medicine, Peoria, United States of America.
    Tan, A. L.
    Leeds Teaching Hospitals Trust, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom; University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom.
    Nune, A.
    Southport and Ormskirk Hospital NHS Trust, Southport, United Kingdom.
    Cavagna, L.
    Fondazione I.R.C.C.S. Policlinico San Matteo, Division of Rheumatology, Pavia, Italy; University of Pavia, Department of Internal Medicine and Medical Therapeutics, Pavia, Italy.
    Saavedra, M. A.
    Hospital de Especialidades Dr. Antonio Fraga Mouret, Centro Médico Nacional La Raza, IMSS, Department of Rheumatology, Mexico City, Mexico.
    Ziade, N.
    Saint-Joseph University, Department of Rheumatology, Beirut, Lebanon; Hotel-Dieu de France Hospital, Department of Rheumatology, Beirut, Lebanon.
    Knitza, J.
    Friedrich-Alexander-Universität Erlangen-Nürnberg, Department of Rheumatology and Immunology, Erlangen, Germany.
    Kuwana, M.
    Nippon Medical School Graduate School of Medicine, Department of Allergy and Rheumatology, Tokyo, Japan.
    Distler, O.
    University Hospital Zurich, University of Zurich, Department of Rheumatology, Zurich, Switzerland.
    Chinoy, H.
    School of Biological Sciences, The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Manchester, United Kingdom; The University of Manchester, National Institute for Health Research Manchester Biomedical Research Centre, Manchester, United Kingdom; Manchester Academic Health Science Centre, Department of Rheumatology, Salford, United Kingdom.
    Agarwal, V.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, United States of America.
    COVID-19 SEVERITY AND VACCINE BREAKTHROUGH INFECTIONS IN IDIOPATHIC INFLAMMATORY MYOPATHIES, OTHER SYSTEMIC AUTOIMMUNE AND INFLAMMATORY DISEASES, AND HEALTHY INDIVIDUALS: RESULTS FROM THE COVID-19 VACCINATION IN AUTOIMMUNE DISEASES (COVAD) STUDY.2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 334-336, article id POS0201Article in journal (Other academic)
  • 15.
    Gupta, L.
    et al.
    Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, Department of Rheumatology, Wolverhampton, United Kingdom; Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Department of Clinical Immunology and Rheumatology, Lucknow, India; Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Manchester, United Kingdom; Sandwell and West Birmingham Hospitals NHS Trust, City Hospital, Birmingham, United Kingdom.
    Ravichandran, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Venerito, V.
    Department of Emergency and Transplantation, University of Bari, Rheumatology Unit, Bari, Italy.
    Dey, M.
    University of Liverpool, Institute of Life Course and Medical Sciences, Liverpool, United Kingdom; Queen Elizabeth Hospital, Department of Rheumatology, London, United Kingdom.
    Sen, P.
    Maulana Azad Medical College, MBBS, New Delhi, India.
    Saha, S.
    Mymensingh Medical College, MBBS, Mymensingh, Bangladesh.
    Tan, A. L.
    Leeds Teaching Hospitals Trust, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom; University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom.
    Shinjo, S. Katsuyuki
    Universidade de Sao Paulo, Division of Rheumatology, Faculdade de Medicina FMUSP, Sao Paulo, Brazil.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, MBBS, Navi Mumbai, India.
    Joshi, M.
    Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospitals, MBBS, Pune, India.
    Ziade, N.
    Saint-Joseph University, Rheumatology Department, Beirut, Lebanon; Hotel-Dieu de France Hospital, Rheumatology Department, Beirut, Lebanon.
    Velikova, T.
    University Hospital “Lozenetz, Sofia University St. Kliment Ohridski, Department of Clinical Immunology, Medical Faculty, Sofia, Bulgaria.
    Jagtap, K.
    Seth Gordhandhas Sunderdas Medical College and King Edwards Memorial Hospital, MBBS, Mumbai, India.
    Milchert, M.
    Pomeranian Medical University in Szczecin, Department of Internal Medicine, Rheumatology, Diabetology, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Gracia-Ramos, A. E.
    General Hospital, National Medical Center “La Raza”, Instituto Mexicano del Seguro Social, Department of Internal Medicine, Mexico City, Mexico.
    Cavagna, L.
    Università degli studi di Pavia, Rheumatology Unit, Dipartimento di Medicine Interna e Terapia Medica, Pavia, Lombardy, Italy.
    Kuwana, M.
    Nippon Medical School Graduate School of Medicine, Department of Allergy and Rheumatology, Tokyo, Japan.
    Knitza, J.
    Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Medizinische Klinik 3 – Rheumatologie und Immunologie, Erlangen, Deutschland, Germany.
    Makol, A.
    Mayo Clinic, Division of Rheumatology, Rochester, Minnesota, United States of America.
    Dzifa, D.
    University of Ghana Medical School, College of Health Sciences, Rheumatology Unit, Department of Medicine and Therapeutics, Accra, Ghana.
    Gutierrez, C. E. Toro
    Pontificia Universidad Javeriana - Cali, General Director, Reference Center for Osteoporosis, Rheumatology and Dermatology, Cali, Colombia.
    Caballero, C. Vinicio
    University Hospital North, Department of Medicine, Soledad, Colombia.
    Distler, O.
    University Hospital of Zürich, Department of Rheumatology, Zürich, Switzerland.
    Day, J.
    WEHI - Walter and Eliza Hall Institute of Medical Research, Department of Rheumatology, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia; Royal Melbourne Hospital, Department of Rheumatology, Parkville, Australia.
    Chinoy, H.
    Manchester Academic Health Science Centre The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester, United Kingdom; The University of Manchester, National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Department of Rheumatology, Salford, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Department of Clinical Immunology and Rheumatology, Pittsburgh, United States of America.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Rheumatology Department, London, United Kingdom.
    COMORBIDITIES, COMPLEX MULTIMORBIDITY AND PROMIS HEALTH OUTCOMES AMONGAUTOIMMUNE RHEUMATIC DISEASES: DATA FROM THE COVAD STUDY2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 555-556, article id POS0573Article in journal (Other academic)
    Abstract [en]

    Background: Comorbidities have a profound impact on the QoL of patients living with autoimmune rheumatic diseases (AIRDs). Unfortunately, global data on the burden of comorbidities and its impact on health outcomes in this vulnerable group is scarce.

    Objectives: We studied the prevalence, distribution and clustering of comorbidities and multimorbidity among patients with AIRDs and healthy controls (HCs) and its impact on health outcomes, utilizing data from the ongoing 2nd COVAD study.

    Methods: The COVAD study is a global e-survey that embodies patient voice while empowering collaborators and young researchers. The study group of 157 physicians across 106 countries from February-June 2022 captured details of AIRDs, autoimmune and non-autoimmune comorbidities, and validated patient reported outcomes. Human Development Index (UNDP 2021-22) of country of residence was taken as a surrogate marker for socioeconomic status (SES).

    Basic multimorbidity (BM), Complex multimorbidity (CM), Autoimmune multimorbidity (AM) are defined as the co-occurrence of ≥2 non-rheumatic comorbidities, ≥3 non-rheumatic chronic conditions affecting ≥3 different organ systems [1] and ≥3 autoimmune diseases (AIDs) in an individual respectively.

    PROMIS global physical health (PGP), mental health (PGM), fatigue 4a (F4a) and physical function short form (SF10) scores were calculated for the different groups and compared using descriptive statistics, linear regression and cluster analysis (hierarchical followed by K means).

    Results: Of 17,612 total respondents, 6149 (62.7%) had underlying AIRDs and 3652 (37.3%) were HCs, with female (80.8%) and Caucasian (53.9%) predominance in the former.

    All types of multimorbidity were more frequent in AIRDs than HCs, including any comorbidity (77.1% versus 25.0%; OR: 2.9; 2.7-3.2), BM (21.0% vs 6.2%; 4.0; 3.4-4.6), and CM (3.1% vs 0.5%; 6.4; 3.9-10.4), and with prevalence increasing with age (p<0.001) (Figure 1A, B). Comorbidity prevalence was the highest among Americans and Australians (72% each).

    Patients with AIRDs had poorer health outcomes than HCs, including lower PGP, PGM, SF10, F4a scores (all p<0.001). Among AIRDs, those with comorbidities had lower physical function and PROMIS scores (PGP, PGM, and SF10), and reported fatigue more often (all p<0.001).

    Female gender, and underlying BM and AM particularly predisposed patients to worse physical health (lower PGP, lower SF10a) and mental health outcomes (lower PGM). While advanced age (-1.815; <0.001), and lower SES (0.871; 0.027) specifically predicted poorer physical function (lower SF10a). Fatigue (higher F4a) was seen more frequently among women (1.711; <0.001), and those with BM (1.142; 0.002); AM (1.768; 0.011), and higher SEC (0.478; 0.016).

    Cluster analysis of patients with AIRDs revealed 2 clusters (Figure 1C 1D); cluster 1 with low PGP, PGM, SF10 and high F4a; cluster 2 with high PGP, PGM, SF10 and low F4a. The clusters differed predominantly based on the frequency of comorbidities; any comorbidity (59.7% vs 41.8%; p<0.001), BM (28.5% vs 14.7%; 0.001); CM (4.5% vs 1.9%; <0.001), and AM (10.0% vs 4.0%; <0.001).

    Conclusion: Comorbidities complicate three-quarters of individuals living with AIRDs, and have an outsized impact on self-reported physical function, perceived fatigue, and QoL. Substantial regional differences call for further exploration of key drivers of this important aspect to allow optimized multidisciplinary and holistic care in anticipation of poorer outcomes.

    Reference: [1]Harrison C, Britt H, Miller G, Henderson J. Examining different measures of multimorbidity, using a large prospective cross-sectional study in Australian general practice. BMJ Open. 2014 Jul 1;4(7):e004694.

  • 16.
    Holloway, A.
    et al.
    Kings College Hospital, Rheumatology Department, London, United Kingdom.
    Lee, S. Y.
    Kings College Hospital, Rheumatology Department, London, United Kingdom.
    Nikiphorou, E.
    Kings College Hospital, Rheumatology Department, London, United Kingdom; Kings College Hospital, Rheumatology Department, London, United Kingdom.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Faculty of Medicine and Health, Örebro University, Department of Rheumatology, Örebro, Sweden; Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Ravichandran, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Day, J.
    The Royal Melbourne Hospital, Department of Rheumatology, Parkville, Australia; WEHI - Walter and Eliza Hall Institute of Medical Research, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia.
    Joshi, M.
    Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospitals, Pune, India.
    Saha, S.
    Mymensingh Medical College, Mymensingh, Bangladesh.
    Shaharir, S. S.
    Universiti Kebangsaan Malaysia, Faculty of Medicine, Bangi, Malaysia.
    Katchamart, W.
    Faculty of Medicine Siriraj Hospital, Mahidol University, Division of Rheumatology, Department of Medicine, Bangkok, Thailand.
    Goo, P. A.
    Queen Savang Vadhana Memorial Hospital, Department of Medicine, Chonburi, Thailand.
    Traboco, L.
    St. Luke’s Medical Center - Global City, Department of Medicine, Section of Rheumatology, Taguig, Philippines.
    Chen, Y. M.
    Taichung Veterans General Hospital, Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung City, Taiwan, Republic of China; Taichung Veterans General Hospital, Department of Medical Research, Taichung, Taiwan, Republic of China.
    Sen, P.
    Maulana Azad Medical College (MAMC), New Delhi, India.
    Lilleker, J. B.
    The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester, United Kingdom; Salford Royal NHS Foundation Trust, Manchester Centre for Clinical Neurosciences, Manchester, United Kingdom.
    Nune, A.
    Southport & Ormskirk Hospital NHS Trust, Southport, United Kingdom.
    Pauling, J.
    University of Bristol, Bristol Medical School Translational Health Sciences, Bristol, United Kingdom; North Bristol NHS Trust, Department of Rheumatology, Bristol, United Kingdom.
    Tan, A. L.
    Leeds Teaching Hospitals NHS Trust, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom; University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom.
    Ziade, N.
    Saint Joseph University of Beirut, Rheumatology Department, Beirut, Lebanon; Hôtel-Dieu de France Hospital, Rheumatology Department, Beirut, Lebanon.
    Milchert, M.
    Pomeranian Medical University, Department of Internal Medicine, Rheumatology, Diabetology, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Gracia-Ramos, A. E.
    General HospitalNational Medical Center “La Raza”, Instituto Mexicano del Seguro Social, Department of Internal Medicine, Mexico City, Mexico.
    Vinicio Caballero, C.
    Hospital Universidad del Norte, Department of Medicine, Barranquilla, Colombia.
    Agarwal, V.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, United States of America.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester, United Kingdom; Royal Wolverhampton Hospitals NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom; City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Department of Rheumatology, Birmingham, United Kingdom.
    Wincup, C.
    Kings College Hospital, Rheumatology Department, London, United Kingdom; Kings College Hospital, Rheumatology Department, London, United Kingdom.
    EVALUATING GLOBAL PATTERNS IN TREATMENT AND PREVALENCE OF COMORBIDITIES IN SYSTEMIC LUPUS ERYTHEMATOSUS2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 1456-1458, article id AB0522Article in journal (Other academic)
    Abstract [en]

    Background: Regional disparities in the management of systemic lupus erythematosus (SLE) are frequently described. Governance, funding, logistic barriers, and physician choice may be important determinants though scarce data from underrepresented regions limits our understanding.

    Objectives: To evaluate global patterns in treatment of SLE and identify the prevalence of comorbidities.

    Methods: We identified SLE patients from the COVAD 2 database, consisting of over 20,000 respondents worldwide. Healthy controls (HC) were included to assess population comorbidity levels. Demographics, treatment i.e., corticosteroids (CS), antimalarials, immunosuppressants (IS), cyclophosphamide and biologics plus comorbidity data was recorded. Country Human Development Index (HDI) classification, a composite index formulated by the United Nations to rank countries into tiers of development, was utilised.

    Results: 3323 HCs and 1167 SLE patients were included in analysis. Patients from low/medium HDI (lmHDI) countries were younger than those from high/very high HDI (hvhHDI) countries (median age 32, IQR 27-41 vs 41, IQR 32-52 years, p<0.0001). Disease duration was shorter in lmHDI countries (median 5, IQR 3-10 vs 10, IQR 5-19 years, p<0.0001).

    A higher proportion of SLE patients from lmHDI countries were on CS (73% vs 59%, p=0.0002), antimalarials (81% vs 68%, p=0.0002) and IS (66% vs 53%, p=0.0009) compared with patients from hvhHDI countries. Choice of IS varied with azathioprine prescribed more frequently in lmHDI countries (p=0.049). Biologics use was more common in hvhHDI countries (7% vs 2%, p=0.0055). Comorbidity prevalence was similar between groups, however when adjusted for age, patients with chronic kidney disease were significantly younger in lmHDI countries (36.67 vs 44.64 years, p=0.015), as were patients with coronary artery disease (35.7 vs. 44.6 years, p=0.015) and hypertension (41.5 vs 49.8 years, p=0.003). Results are detailed in Table 1.

    Conclusion: To our knowledge, this is the largest study evaluating treatment and comorbidity incidence in SLE populations based on country HDI. We identified striking differences in pharmacological management globally. Cardiovascular comorbidities were seen in younger patients and earlier in the disease course in lmHDI countries, suggestive of premature organ damage. This could be due to limited global access to high-cost medication and increasing access may improve outcomes. Our results call for review of cardiovascular risk guidelines and regional approaches to preventive action as well as pharmacological and non-pharmacological management of patients with established cardiovascular comorbidity.

  • 17.
    Jägerback, S.
    et al.
    Karolinska Institutet and Karolinska University Hospital, Stockholm, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Danderyd University Hospital, Division of Rheumatology, Danderyd, Sweden.
    Gomez, A.
    Karolinska Institutet and Karolinska University Hospital, Stockholm, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Stockholm, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    PREDICTORS OF RENAL FLARES IN SYSTEMIC LUPUS ERYTHEMATOSUS: A POST-HOC ANALYSIS OF FOUR PHASE III CLINICAL TRIALS OF BELIMUMAB2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 34-34, article id OP0052Article in journal (Other academic)
    Abstract [en]

    Background: In patients with systemic lupus erythematosus (SLE), renal involvement is associated with high morbidity, and renal flares are major contributors to poor long-term prognosis. Identification of patients at risk of developing renal flares despite immunosuppressant therapy is imperative to optimise management.

    Objectives: To identify predictors of renal flares in patients receiving treatment for active extra-renal SLE.

    Methods: Data from BLISS-52 (NCT00424476), BLISS-76 (NCT00410384), BLISS Northeast Asia (NEA; NCT01345253), and BLISS-SC (NCT01484496) were used (N=3225). The trials included patients with active, seropositive SLE and excluded active severe renal SLE. Participants were assigned to belimumab or placebo, on top of non-biologic standard therapy. We investigated anti-dsDNA, anti-Sm, anti-ribosomal P, and anti-cardiolipin (aCL) antibodies, low C3 and low C4 levels, B cell activating factor (BAFF), serum albumin, serum creatinine, and proteinuria at baseline as potential predictors of renal flares during a 52-week follow-up. We used Cox regression models to evaluate traditional disease features as predictors of renal flares in the pooled trial population. We also performed subgroup analysis of belimumab and placebo recipients. Covariates in the adjusted models included age, sex, ethnicity, body mass index, organ damage, baseline extra-renal activity (SLEDAI-2K score excluding renal and immunological descriptors), current or former renal SLE history (renal BILAG A–D), baseline use of glucocorticoids, antimalarials, and immunosuppressants, and use of belimumab.

    Results: The mean age of the study population was 36.7 years, 94% were women, 54.6% had current or former renal involvement at baseline, and 192 developed a renal flare after a median follow-up time of 197 (IQR: 85–330) days from baseline. Patients with current or former renal involvement at baseline displayed a >9-fold increased hazard to develop a new renal flare (HR: 9.4; 95% CI: 5.0–17.7; P<0.001). In the pooled study population, baseline serum albumin (adjusted HR 0.9; 95% CI: 0.9–0.9; P<0.001), proteinuria (adjusted HR: 1.3; 95% CI: 1.2–1.4; P<0.001), and low C3 levels (adjusted HR: 1.8; 95% CI: 1.3–2.5; P<0.001) were robust determinants of subsequent renal flare occurrence; similar associations were found in the belimumab and placebo subgroups. Furthermore, we observed an association between anti-dsDNA positivity and renal flare development in univariable models (HR: 2.1; 95% CI: 1.4–3.2; P<0.001 in the pooled population), which attenuated in multivariable models (Figure 1). Positive levels of anti-Sm antibodies were associated with renal flare occurrence in the placebo (adjusted HR: 2.9; 95% CI: 1.5–5.6; P=0.002) but not in the belimumab subgroup, whereas anti-ribosomal P antibodies were associated with renal flare development in belimumab-treated (HR: 2.8; 95% CI: 1.5–5.0; P<0.001) but not in placebo-treated patients. Finally, positive levels of aCL antibodies (any isotype) predicted renal flare development in the belimumab group (adjusted HR: 1.8; 95% CI: 1.1–2.8; P=0.020) but yielded a negative association in the placebo group (adjusted HR: 0.4; 95% CI: 0.2–0.9; P=0.028).

    Conclusion: Current or former renal involvement, high baseline proteinuria levels, hypoalbuminaemia, and C3 consumption were robust determinants of imminent renal flares. Beyond anti-dsDNA, anti-ribosomal P and aCL antibody positivity may prove valuable early signals of imminent renal flares in patients treated with belimumab. Anti-Sm positivity predicted imminent renal flares in patients treated with non-biological standard therapy, but not in belimumab-treated patients, assumably due to benefit conferred from belimumab in anti-Sm positive individuals, as shown previously [1].

    Reference: [1]Parodis I, et al. Int J Mol Sci. 2020; 21(10):3463

  • 18.
    Jónsdóttir, Thorunn
    et al.
    Department of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
    Gunnarsson, Iva
    Department of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
    Risselada, Anke
    Department of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
    Welin Henriksson, Elisabet
    Department of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
    Klareskog, Lars
    Department of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
    van Vollenhoven, Ronald F
    Department of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
    Treatment of refractory SLE with rituximab plus cyclophosphamide: clinical effects, serological changes, and predictors of response2008In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 67, no 3, p. 330-334Article in journal (Refereed)
    Abstract [en]

    OBJECTIVE: To evaluate efficacy, serological responses, and predictors of response in patients with severe and refractory systemic lupus erythematosus (SLE) treated with rituximab plus cyclophosphamide.

    METHODS: 16 patients entered a treatment protocol using rituximab plus cyclophosphamide. Disease activity was assessed by the SLE disease activity index (SLEDAI) and by the British Isles Lupus Assessment Group (BILAG) index.

    RESULTS: At six months follow up, mean SLEDAI values decreased significantly from (mean (SD)) 12.1 (2.2) to 4.7 (1.1). Clinical improvement (50% reduction in SLEDAI) occurred in all but three patients. All but one patient responded according to BILAG. Remission defined as SLEDAI <3 was achieved in nine of 16 patients. Isotype analysis of anti-dsDNA antibodies revealed preferential decreases of IgG and IgA, but not IgM. Higher absolute numbers of CD19+ cells at baseline were correlated with shorter depletion time (r = -0.6).

    CONCLUSIONS: The majority of patients improved following rituximab plus cyclophosphamide. The differential downregulation of anti-DNA of the IgG and IgA but not the IgM isotypes supports the hypothesis that cells producing pathogenic autoantibodies are preferentially targeted by the treatment. The fact that greater absolute numbers of CD19+ cells at baseline predict a less impressive clinical and serological response suggests that more flexible dosing could be advantageous.

  • 19.
    Jónsdóttir, Thórunn
    et al.
    Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
    Sundelin, Birgitta
    Department of Pathology and Cytology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Swede.
    Welin Henriksson, Elisabet
    Department of Neurobiology, Care Sciences and Society, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
    van Vollenhoven, Ronald F.
    Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
    Gunnarsson, Iva
    Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
    Rituximab-treated membranous lupus nephritis: clinical outcome and effects on electron dense deposits2011In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 70, no 6, p. 1172-3Article in journal (Refereed)
  • 20.
    Lee, S. Y.
    et al.
    King’s College Hospital, Rheumatology Department, London, United Kingdom.
    Holloway, A.
    King’s College Hospital, Rheumatology Department, London, United Kingdom.
    Nikiphorou, E.
    King’s College Hospital, Rheumatology Department, London, United Kingdom; King’s College London, Centre for Rheumatic Diseases, London, United Kingdom.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Sweden, Sweden; Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Ravichandran, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Day, J.
    The Royal Melbourne Hospital, Department of Rheumatology, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia; WEHI - Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.
    Joshi, M.
    Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospitals, Pune, India.
    Saha, S.
    Mymensingh Medical College, Mymensingh, Bangladesh.
    Jagtap, K.
    Seth Gordhandhas Sunderdas Medical College and King Edwards Memorial Hospital, Mumbai, Maharashtra, India.
    Katchamart, W.
    Faculty of Medicine Siriraj Hospital, Mahidol University, Division of Rheumatology, Department of Medicine, Bangkok, Thailand.
    Goo, P. A.
    Queen Savang Vadhana Memorial Hospital, Department of Medicine, Chonburi, Thailand.
    Vaidya, B.
    National Center for Rheumatic Diseases (NCRD), Ratopul, Kathmandu, Nepal.
    Velikova, T.
    Sofia University St. Kliment Ohridski, Medical Faculty, Sofia, Bulgaria.
    Sen, P.
    Maulana Azad Medical College, New Delhi, India.
    Katsuyuki Shinjo, S.
    Universidade de Sao Paulo, Division of Rheumatology, Faculdade de Medicina FMUSP, Sao Paulo, Brazil.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, Navi Mumbai, Maharashtra, India.
    Tan, A. L.
    Leeds Teaching Hospitals Trust, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom; University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom.
    Ziade, N.
    Saint-Joseph University, Rheumatology, Beirut, Lebanon; Hotel-Dieu de France Hospital, Rheumatology, Beirut, Lebanon.
    Milchert, M.
    Pomeranian Medical University in Szczecin, Department of Internal Medicine, Rheumatology, Diabetology, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Gracia-Ramos, A. E.
    National Medical Center “La Raza”, Instituto Mexicano del Seguro Social, Department of Internal Medicine, Mexico City, Mexico.
    Vinicio Caballero, C.
    Hospital Universidad del Norte, Department of Medicine, Barranquilla, Atlantico, Colombia.
    Chinoy, H.
    Manchester Academic Health Science Centre, The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester, United Kingdom; Manchester University NHS Foundation Trust, The University of Manchester, National Institute for Health Research Manchester Biomedical Research Centre, Manchester, United Kingdom; Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Department of Rheumatology, Salford, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pennsylvania, United States of America.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; Manchester Academic Health Science Centre, The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester, United Kingdom; Royal Wolverhampton Hospitals NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom; City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, United Kingdom.
    Wincup, C.
    Rayne Institute, University College London, Department of Rheumatology, Division of Medicine, London, United Kingdom; UCLH, GOSH, Centre for Adolescent Rheumatology Versus Arthritis at UCL, London, United Kingdom.
    IDENTIFYING DETERMINANTS OF FAVOURABLE AND POOR PHYSICAL FUNCTION IN SYSTEMIC LUPUS ERYTHEMATOSUS: RESULTS FROM AN INTERNATIONAL COLLABORATIVE STUDY2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 1110-1112, article id POS1506Article in journal (Other academic)
    Abstract [en]

    Background: Systemic lupus erythematosus (SLE) can result in impaired daily physical function through various mechanisms including active disease, chronic damage, and mental health symptoms that are common in the disease. However, the key drivers of reduced physical function are poorly understood, and no large-scale global studies investigating this have been conducted to date.

    Objectives: To investigate key factors that contribute to impaired physical function in SLE globally.

    Methods: SLE patients were identified from the COVAD 2 database, a global register of more than 20,000 respondents. Healthy controls (HC) were included to compare differences in physical function using the Patient Reported Outcome Measurement Information System (PROMIS) questionnaire. Demographics, medication, comorbidities, disease activity, Global Physical Health (GPH) and Global Mental Health (GMH) were collected. Multivariable regression analysis was used to identify contributing factors to favourable or poor physical function (measured by PROMIS Physical Function shortform PF-10a score).

    Results: 979 SLE patients and 3358 HCs were included in analysis. Patients with SLE had significantly lower PF-10a score as compared to HCs (median 42, IQR 36-47 vs median 49, IQR 45-50, p<0.0001). Determinants of physical function status in patients with SLE are summarised in Table 1. Briefly, factors associated with poor physical function included increasing age (-0.042, 95% CI -0.069 to -0.015, p=0.002) and methotrexate use (-0.928, 95% CI -1.844 to -0.012, p=0.047). Diabetes (-1.862, 95% CI -3.481 to -0.243, p=0.024) and interstitial lung disease (ILD) (-2.441, 95% CI -4.366 to -0.517, p=0.013), but not asthma or COPD, also contributed to lower PF-10a score. From a mental health perspective, anxiety (-0.970, 95% CI -1.853 to -0.087, p=0.031) but not depression contributed to a lower physical function score. Higher Pain Visual Analogue Scales (VAS) (-2.889, 95% CI -3.107 to -2.671, p<0.001) and Fatigue VAS (-1.459, 95% CI -1.974 to -0.945, p<0.001) also contributed to lower PF-10 scores. Hydroxychloroquine use (0.844, 95% CI 0.190 to 1.498, p=0.012) and higher GPH score (2.287, 95% CI 2.079 to 2.494, p<0.001) were associated with favourable physical function.

    Conclusion: Patients with SLE show significantly reduced physical function compared with HCs. Key contributors to poor physical function include intercurrent diabetes and ILD. Screening for, and aggressive early treatment of these conditions may confer improved long-term function. As expected, higher levels of pain and fatigue were associated with poor physical function. Methotrexate use was also identified as a contributing factor to reduced function, which could represent its use in articular manifestations that limit physical function. Importantly, use of hydroxychloroquine was associated with favourable physical function, adding to the well-recognised benefits of this drug in SLE.

  • 21.
    Lindblom, J.
    et al.
    Karolinska Institutet, Department of Medicine Solna, Stockholm, Sweden.
    Toro-Dominguez, D.
    Centre for Genomics and Oncological Research: Pfizer, GENYO, Granada, Spain.
    Borghi, M. O.
    Università degli Studi di Milano and Istituto Auxologico Italiano, Department of Clinical Sciences and Community Health, Milano, Italy.
    Iacobaeus, E.
    Karolinska Institutet, Department of Clinical Neuroscience, Neuroimmunology Unit, Stockholm, Sweden.
    Enman, Y.
    Karolinska Institutet, Department of Medicine Solna, Stockholm, Sweden.
    Repsilber, Dirk
    Örebro University, School of Medical Sciences.
    Mohan, C.
    University of Houston, Department Biomedical Engineering, Houston, United States of America.
    Alarcon-Riquelme, M.
    Centre for Genomics and Oncological Research: Pfizer, GENYO, Granada, Spain.
    Barturen, G.
    Centre for Genomics and Oncological Research: Pfizer, GENYO, Granada, Spain.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    TRANSCRIPTOME PROFILING AND AUTOIMMUNITY-RELATED SEROLOGICAL MARKERS IDENTIFY TP53 AND C3AR AS DRUG TARGETS IN NEUROPSYCHIATRIC SYSTEMIC LUPUS ERYTHEMATOSUS2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 326-326, article id POS0188Article in journal (Other academic)
  • 22. MacFarlane, G.J.
    et al.
    Pallewatte, N.
    Paudyal, P.
    Blyth, F. M.
    Coggon, D.
    Crombez, G.
    Linton, Steven J.
    Örebro University, School of Law, Psychology and Social Work.
    Leino-Arjas, P.
    Silman, A. J.
    Smeets, R. J.
    van der Windt, D.
    Evaluation of work-related psychosocial factors and regional musculoskeletal pain: results from a EULAR Task Force2009In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 68, no 6, p. 885-891Article in journal (Refereed)
    Abstract [en]

    Objectives: To establish whether review articles provide consistent conclusions on associations between workplace psychosocial factors and musculoskeletal pain and, if differences exist, to explore whether this is related to the methods used.

    Methods: Reviews, reported up to February 2007, that included consideration of workplace psychosocial factors and upper limb, back or knee pain were identified through searches of multiple databases. The specific work-related psychosocial factors considered were job demands, support, job autonomy and job satisfaction. The conclusions of each review on one or more of the psychosocial/musculoskeletal pain associations were extracted.

    Results: 15 review articles were identified that considered one or more of the regional pain syndromes included in the study. For back pain, the most consistent conclusions (four reviews positive out of six) were with high job demands and low job satisfaction. The studies of upper limb pain were exclusively related to shoulder and/or neck pain, and the most consistent positive conclusions were with high and low job demands (four reviews positive out of six and two reviews positive out of three, respectively). For knee pain, only a single review was identified. For individual reviews of back and upper limb pain, there were marked differences in the number of associations concluded to be positive between reviews.

    Conclusions: The reasons for reviews coming to different conclusions included that they were often evaluating different bodies of evidence (according to their search criteria, the year when the review was conducted, the role that quality assessment played in whether studies contributed to evidence, and the combination of risk factors addressed in individual studies), but more important was whether the review specified explicit criteria for making conclusions on strength of evidence. These conclusions emphasise the importance of developing standardised methods for conducting such evaluations of existing evidence and the importance of new longitudinal studies for clarifying the temporal relationship between psychosocial factors and musculoskeletal pain in the workplace.

  • 23.
    Olofsson, T.
    et al.
    Department of clinical sciences Lund, Rheumatology, Lund University, Lund, Sweden.
    Mogård, E.
    Department of clinical sciences Lund, Rheumatology, Lund University, Lund, Sweden.
    Andreasson, K.
    Department of clinical sciences Lund, Rheumatology, Lund University, Lund, Sweden.
    Marsal, J.
    Department of clinical sciences Lund, Gastroenterology, Lund University, Lund, Sweden.
    Geijer, Mats
    Örebro University, School of Medical Sciences. Örebro University Hospital. Department of Radiology, Örebro University Hospital, Örebro, Sweden.
    Kristensen, L. -E
    Dept Rheumatol, Parker Inst, Copenhagen Univ Hosp, Copenhagen, Denmark; Dept Clin Sci Lund, Rheumatol, Lund Univ, Lund, Sweden.
    Lindqvist, E.
    Department of clinical sciences Lund, Rheumatology, Lund University, Lund, Sweden.
    Wallman, J. K.
    Department of clinical sciences Lund, Rheumatology, Lund University, Lund, Sweden.
    Faecal Calprotectin, But Not Anti-Saccharomyces Cerevisiae Antibodies, Is Linked To Worse Disease Status In Axial Spondyloarthritis Patients Without Inflammatory Bowel Disease: Results From The Spartakus Cohort2017In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 76, no Suppl. 2, p. 655-655Article in journal (Other academic)
  • 24.
    Olsson, E. Kihlgren
    et al.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Ravichandran, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Rheumatology Department, London, United Kingdom.
    Lindblom, J.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Saha, S.
    Mymensingh Medical College, Mymensingh Medical College, Mymensingh, Bangladesh.
    Shaharir, S. S.
    Universiti Kebangsaan Malaysia, Faculty of Medicine, Cheras, Kuala Lumpur, Malaysia.
    Katchamart, W.
    Faculty of Medicine Siriraj Hospital, Mahidol University, Division of Rheumatology, Department of Medicine, Bangkok, Thailand.
    Goo, P. A.
    Queen Savang Vadhana Memorial Hospital, Department of Medicine, Chonburi, Thailand.
    Traboco, L.
    St. Luke’s Medical Center-Global City, Department of Medicine, Section of Rheumatology, Taguig, Philippines.
    Chen, Y. M.
    Taichung Veterans General Hospital, Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung City, Taiwan, Republic of China; Taichung Veterans General Hospital, Department of Medical Research, Taichung, Taiwan, Republic of China.
    Jagtap, K.
    Seth Gordhandhas Sunderdas Medical College and King Edwards Memorial Hospital, Seth Gordhandhas Sunderdas Medical College and King Edwards Memorial Hospital, Mumbai, Maharashtra, India.
    Lilleker, J. B.
    The University of Manchester, Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester, United Kingdom; Salford Royal NHS Foundation Trust, Manchester Centre for Clinical Neurosciences, Salford, United Kingdom.
    Nune, A.
    Southport and Ormskirk Hospital NHS Trust, Southport and Ormskirk Hospital NHS Trust, Southport, United Kingdom.
    Pauling, J.
    Bristol Medical School Translational Health Sciences, Bristol Medical School Translational Health Sciences, Bristol, United Kingdom; North Bristol NHS Trust, Department of Rheumatology, Bristol, United Kingdom.
    Wincup, C.
    Rayne Institute, University College London, Department of Rheumatology, Division of Medicine, London, United Kingdom; Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, United Kingdom.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, Mahatma Gandhi Mission Medical College, Navi Mumbai, Maharashtra, India.
    Dzifa, D.
    University of Ghana Medical School, College of Health Sciences, Korle-Bu, Rheumatology Unit, Department of Medicine and Therapeutics, Accra, Ghana.
    Toro Gutierrez, C. E.
    Pontificia Universidad Javeriana Cali, General Director, Reference Center for Osteoporosis, Rheumatology and Dermatology, Cali, Colombia.
    Vinicio Caballero, C.
    Hospital Universidad del Norte, Department of Medicine, Barranquilla, Atlantico, Colombia.
    Chinoy, H.
    The University of Manchester, Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester, United Kingdom; Manchester University NHS Foundation Trust, The University of Manchester, National Institute for Health Research Manchester Biomedical Research Centre, Manchester, United Kingdom; Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Department of Rheumatology, Salford, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, Pennsylvania, United States of America.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; The University of Manchester, Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester, United Kingdom; Royal Wolverhampton Hospitals NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom; Sandwell and West Birmingham Hospitals NHS Trust, City Hospital, Birmingham, United Kingdom.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    BREAKTHROUGH SARS-COV-2 INFECTION IN FULLY VACCINATED PATIENTS WITH SYSTEMIC LUPUS ERYTHEMATOSUS: RESULTS FROM THE COVID-19 VACCINATION IN AUTOIMMUNE DISEASE (COVAD) STUDY2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 540-541, article id POS0549Article in journal (Other academic)
    Abstract [en]

    Background: Although many studies have been conducted on COVID-19 in recent years, there are still unanswered questions regarding breakthrough infections (BTIs), particularly in patients with systemic lupus erythematosus (SLE).

    Objectives: This study aimed to determine the occurrence of breakthrough COVID-19 infections in patients with SLE versus other autoimmune rheumatic diseases (AIRDs), non-rheumatic autoimmune diseases (nrAIDs), and healthy controls (HCs).

    Methods: The study was based on data from the COVAD questionnaire which amassed a total of 10,783 complete responses from patients with SLE, AIRD, or nrAIRD, and HCs. After exclusion of individuals who were unvaccinated, those who received one vaccine dose only, and those with uncertain responses regarding the vaccine doses, a total of 9,595 patients formed the study population of the present investigation. If a COVID-19 infection occurred after the initial two vaccine doses and at least one booster dose (at least three doses in total, herein termed full vaccination), it was considered a BTI. Data were analysed using multivariable regression models. Statistically significant results were denoted by p values <0.05.

    Results: A total of 7,016/9,595 (73.1%) individuals were fully vaccinated. Among those, 1,002 (14.2%) reported at least one BTI, and 166 (2.3%) reported at least two BTIs. Among SLE patients, 867/1,218 (71.2%) were fully vaccinated. Among fully vaccinated SLE patients, 137 (15.8%) reported at least one BTI while 28 (3.2%) reported at least two BTIs. BTI frequencies in fully vaccinated SLE patients were comparable to those of other AIRDs (OR: 1.0; 95% CI: 0.8–1.3; p=0.447) and nrAIDS (OR: 0.9; 95% CI: 0.6–1.3; p=0.856) but higher compared with HCs (OR: 1.2; 95% CI: 1.0–1.6; p=0.022).

    For SLE patients with three vaccine doses, 113/137 (82.5%) reported at least one BTI while the corresponding number for four vaccine doses was 24/137 (17.5%). Compared with HCs (OR: 10.6; 95% CI: 1.2–93.0; p=0.032) and other AIRDs (OR: 3.5; 95% CI: 1.08–11.5; p=0.036), SLE patients showed higher frequencies of hospitalisation.

    AID multimorbidity was associated with a 15-fold increased risk for a need of advanced treatment for COVID-19 (OR: 15.3; 95% CI: 2.6–88.2; p=0.002).

    Conclusion: COVID-19 BTIs occurred in nearly 1 every 6th fully vaccinated patient with SLE, and 20% more frequently in this patient population compared with fully vaccinated HCs. Moreover, BTIs in SLE patients were more severe compared with BTIs in HCs or patients with AIRDs other than SLE, resulting in a greater need for hospitalisation. AID multimorbidity contributed to a more severe COVID-19 BTI requiring advanced management. These insights call for greater attention to vaccination in the vulnerable group of SLE patients, with appropriate risk stratification towards optimised vaccination strategies.

  • 25.
    Palazzo, L.
    et al.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Lindblom, J.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Cetrez, N.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Ala, H.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    PREDICTORS OF NEUROPSYCHIATRIC FLARES IN PATIENTS WITH SYSTEMIC LUPUS ERYTHEMATOSUS: RESULTS FROM FIVE PHASE III CLINICAL TRIALS OF BELIMUMAB2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 1088-1088, article id POS1467Article in journal (Other academic)
    Abstract [en]

    Background: Neuropsychiatric (NP) flares in systemic lupus erythematosus (SLE) constitute a challenging issue for the clinician on many levels as they are rather unpredictable and include severe and potentially life-threatening manifestations [1]. The identification of clinical and serological profiles predictive of NP flares would be instrumental for early detection of NP involvement, thus allowing early treatment, and hopefully resulting in improved outcomes. The B-cell depleting agent belimumab has widely shown ability to reduce rates of flare in SLE (2), but little is known on its potential benefits in the treatment of NPSLE.

    Objectives: To evaluate predictors of NP flares in patients with SLE under standard therapy (ST) with or without add-on belimumab, given for active disease yet not ongoing severe NP involvement.

    Methods: Data from five clinical trials of belimumab in SLE (BLISS-52 (NCT00424476), BLISS-76 (NCT00410384), BLISS-NEA (NCT01345253), BLISS-SC (NCT01484496), and EMBRACE (NCT01632241)) were used (N=3638). Flares were defined using the British Isles Lupus Assessment Group (BILAG) activity index as development of any new NP BILAG score A in patients with baseline NP BILAG score B, and development of any new NP BILAG score A or B in patients with baseline NP BILAG score C–E during a 52-week follow-up. Predictors of NP flare were investigated using univariable and multivariable Cox regression analysis. A subgroup analysis was performed to determine predictors of de novo NP flare in patients with no NP history (baseline NP BILAG E). Organ damage was assessed using the SLICC/ACR Damage Index (SDI). p values <0.05 were considered statistically significant.

    Results: In total, 105 (2.9%) NP flares were documented. Of these, NP flare occurred in 52 (1.6%) patients with baseline NP BILAG E. In multivariable analysis adjusting for age, sex, ethnicity, and variables that were significant in univariable analysis, male sex (HR: 2.37; 95% CI: 1.31–4.28; p=0.004) and NP BILAG score B–D at baseline (HR: 5.91; 95% CI: 3.86–9.06; p<0.001) were highly associated with NP flare development in the pooled study population. Belimumab use displayed no clear protection at any dose or administration form. In the subgroup analysis of the baseline NP BILAG E population, male sex (HR: 3.26; 95% CI: 1.51–7.04; p=0.003) was the strongest predictor of de novo NP flare. In a separate analysis of SDI domains, the NP domain (HR: 3.25; 95% CI: 2.72–3.88; p<0.001) was the strongest predictor of NP flare, with cognitive impairment (HR: 14.29; 95% CI: 9.22–22.14; p<0.001), transverse myelitis (HR: 21.89; 95% CI: 5.40–88.72; p<0.001), and neuropathy (HR: 8.87; 95% CI: 5.59–14.09; p<0.001) mainly driving this association.

    Conclusion: Current or past NP activity and established organ damage in the NP domain at baseline were the strongest predictors of NP flare in SLE patients treated for active disease yet not ongoing severe NP involvement. Whether belimumab treatment protects against NP events remains unclear and warrants future investigation.

    References:

    [1]Schwartz N, Stock AD, Putterman C. Neuropsychiatric lupus: new mechanistic insights and future treatment directions. Nat Rev Rheumatol. 2019;15(3):137-52.

    [2]Singh JA, Shah NP, Mudano AS. Belimumab for systemic lupus erythematosus. Cochrane Database Syst Rev. 2021;2(2):CD010668.

  • 26.
    Parodis, Ioannis
    et al.
    Örebro University, School of Medical Sciences. Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden; Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
    Girard-Guyonvarc'h, Charlotte
    Division of Rheumatology, Department of Medicine, University Hospital of Geneva, Geneve, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland.
    Arnaud, Laurent
    Department of Rheumatology, Hôpitaux Universitaires de Strasbourg, Centre National de Référence (RESO), INSERM UMR-S 1109, Strasbourg, France.
    Distler, Oliver
    Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
    Domján, Andrea
    Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
    Van den Ende, Cornelia H. M.
    Department of Research, Sint Maartenskliniek, Nijmegen, The Netherlands; Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
    Fligelstone, Kim
    Federation of European Scleroderma Associations (FESCA), Brussels, Belgium.
    Kocher, Agnes
    Institute of Nursing Science, Faculty of Medicine, University of Basel, Basel, Switzerland.
    Larosa, Maddalena
    Rheumatology Unit, Department of Medical Specialties, ASL3, Genoa, Italy.
    Lau, Martin
    Arthritis Action, London, UK.
    Mitropoulos, Alexandros
    Department of Nursing and Midwifery, College of Health Wellbeing and Life Science, Sheffield Hallam University, Sheffield, UK.
    Ndosi, Mwidimi
    School of Health and Social Wellbeing, University of the West of England, Bristol, UK.
    Poole, Janet L.
    Occupational Therapy Graduate Program, School of Medicine, University of New Mexico, Albuquerque, New Mexico, USA.
    Redmond, Anthony
    Leeds Institute for Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
    Ritschl, Valentin
    Section for Outcomes Research, Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria.
    Alexanderson, Helene
    Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Occupational Therapy and Physiotherapy, Karolinska University Hospital, Stockholm, Sweden.
    Sjöberg, Yvonne
    Swedish Rheumatism Association, Stockholm, Sweden.
    von Perner, Gunilla
    Swedish Rheumatism Association, Stockholm, Sweden.
    Uhlig, Till
    Center for Treatment of Rheumatic and Musculoskeletal Diseases, Diakonhjemmet Hospital, Oslo, Norway.
    Varju, Cecilia
    Department of Rheumatology and Immunology, Medical School of University of Pécs, Pécs, Hungary.
    Vriezekolk, Johanna E.
    Department of Research, Sint Maartenskliniek, Nijmegen, The Netherlands.
    Welin, Elisabet
    Örebro University, School of Health Sciences.
    Westhovens, René
    Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.
    Stamm, Tanja A.
    Section for Outcomes Research, Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria.
    Boström, Carina
    Department of Occupational Therapy and Physiotherapy, Karolinska University Hospital, Stockholm, Sweden; Division of Physiotherapy, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
    EULAR recommendations for the non-pharmacological management of systemic lupus erythematosus and systemic sclerosis2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, article id ard-2023-224416Article in journal (Refereed)
    Abstract [en]

    OBJECTIVE: To develop evidence-based recommendations for the non-pharmacological management of systemic lupus erythematosus (SLE) and systemic sclerosis (SSc).

    METHODS: A task force comprising 7 rheumatologists, 15 other healthcare professionals and 3 patients was established. Following a systematic literature review performed to inform the recommendations, statements were formulated, discussed during online meetings and graded based on risk of bias assessment, level of evidence (LoE) and strength of recommendation (SoR; scale A-D, A comprising consistent LoE 1 studies, D comprising LoE 4 or inconsistent studies), following the European Alliance of Associations for Rheumatology standard operating procedure. Level of agreement (LoA; scale 0-10, 0 denoting complete disagreement, 10 denoting complete agreement) was determined for each statement through online voting.

    RESULTS: Four overarching principles and 12 recommendations were developed. These concerned common and disease-specific aspects of non-pharmacological management. SoR ranged from A to D. The mean LoA with the overarching principles and recommendations ranged from 8.4 to 9.7. Briefly, non-pharmacological management of SLE and SSc should be tailored, person-centred and participatory. It is not intended to preclude but rather complement pharmacotherapy. Patients should be offered education and support for physical exercise, smoking cessation and avoidance of cold exposure. Photoprotection and psychosocial interventions are important for SLE patients, while mouth and hand exercises are important in SSc.

    CONCLUSIONS: The recommendations will guide healthcare professionals and patients towards a holistic and personalised management of SLE and SSc. Research and educational agendas were developed to address needs towards a higher evidence level, enhancement of clinician-patient communication and improved outcomes.

  • 27.
    Parodis, Ioannis
    et al.
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Gomez, A.
    Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Tsoi, A.
    Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Chow, J. Weng
    Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Pezzella, D.
    Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Girard, C.
    University Hospital of Geneva and University of Geneva, Division of Rheumatology, Department of Medicine, Geneva, Switzerland.
    Stamm, T.
    Medical University of Vienna, Section for Outcomes Research, Center for Medical Statistics, Informatics and Intelligent Systems, Vienna, Austria; Ludwig Boltzmann Institute, Cluster Arthritis and Rehabilitation, Vienna, Austria.
    Boström, C.
    Karolinska Institutet, Division of Physiotherapy, Department of Neurobiology, Care sciences and Society, Stockholm, Sweden.
    SYSTEMATIC LITERATURE REVIEW TO INFORM THE EULAR TASK FORCE FOR RECOMMENDATIONS/POINTS TO CONSIDER FOR THE NON-PHARMACOLOGICAL MANAGEMENT OF SYSTEMIC LUPUS ERYTHEMATOSUS AND SYSTEMIC SCLEROSIS2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 1098-1099, article id POS1506-HPArticle in journal (Other academic)
  • 28.
    Parodis, Ioannis
    et al.
    Örebro University, School of Medical Sciences. Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden; Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
    Houssiau, Frederic A.
    Pôle de Pathologies Rhumatismales Inflammatoires et Systémiques, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium; Rheumatology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
    From sequential to combination and personalised therapy in lupus nephritis: moving towards a paradigm shift?2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no 1, p. 15-19Article in journal (Refereed)
    Abstract [en]

    The current treatment paradigm in lupus nephritis consists of an initial phase aimed at inducing remission and a subsequent remission maintenance phase. With this so-called sequential treatment approach, complete renal response is achieved in a disappointing proportion of 20-30% of the patients within 6-12 months, and 5-20% develop end-stage kidney disease within 10 years. Treat-to-target approaches are detained owing to uncertainty as to whether the target should be determined based on clinical, histopathological, or immunopathological features. Until reliable non-invasive biomarkers exist, tissue-based evaluation remains the gold standard, necessitating repeat kidney biopsies for treatment evaluation and therapeutic decision-making. In this viewpoint, we discuss the pros and cons of voclosporin and belimumab as add-on agents to standard therapy, the first drugs to be licenced for lupus nephritis after recent successful randomised phase III clinical trials. We also discuss the prospect of obinutuzumab and anifrolumab, also on top of standard immunosuppression, currently tested in phase III trials after initial auspicious signals. Undoubtably, the treatment landscape in lupus nephritis is changing, with combination treatment regimens challenging the sequential concept. Meanwhile, the enrichment of the treatment armamentarium shifts the need from lack of therapies to the challenge of how to select the right treatment for the right patient. This has to be addressed in biomarker surveys along with tissue-level mapping of inflammatory phenotypes, which will ultimately lead to person-centred therapeutic approaches. After many years of trial failures, we may now anticipate a heartening future for patients with lupus nephritis.

  • 29.
    Parodis, Ioannis
    et al.
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Lindblom, J.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Cetrez, N.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Palazzo, L.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Ala, H.
    Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Houssiau, F.
    Université Catholique de Louvain and Service de Rhumatologie, Cliniques Universitaires Saint-Luc, Pôle de Pathologies Rhumatismales Inflammatoires et Systémiques, Institut de Recherche Expérimentale et Clinique, Brussels, Belgium.
    Sjowall, C.
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection/Rheumatology, Linköping, Sweden.
    Rovin, B. H.
    The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, United States of America.
    PREDICTORS OF DE NOVO RENAL FLARES IN SYSTEMIC LUPUS ERYTHEMATOSUS - TIME TO REVISIT BELIMUMAB DOSE FOR EXTRA-RENAL DISEASE?: RESULTS FROM FIVE PHASE III CLINICAL TRIALS OF BELIMUMAB2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 909-910, article id POS1156Article in journal (Other academic)
    Abstract [en]

    Background: Each lupus nephritis (LN) flare causes nephron loss that equals a decade or more of reduction in renal function lifespan, making prompt initiation of therapy imperative and prevention of flares even more desirable. Identification of readily available signals of imminent flare is therefore expected to improve prognosis.

    Objectives: In light of observed cases of de novo LN during belimumab treatment (1), we evaluated predictors of de novo renal flare occurrence in patients with systemic lupus erythematosus (SLE) and no prior history of renal disease undergoing standard therapy (ST) with or without add-on belimumab in clinical trial settings.

    Methods: Data from five clinical trials of belimumab in SLE (BLISS-52 NCT00424476; BLISS-76 NCT00410384; BLISS-NEA NCT01345253; BLISS-SC NCT01484496; EMBRACE NCT01632241) were utilised. The study population comprised 1932 patients with a baseline renal British Isles Lupus Assessment Group (BILAG) score E. De novo renal flares were defined as a change from renal BILAG E to A or B within a 52-week follow-up. Comparisons of baseline data were made using the Mann-Whitney U test, Pearson’s chi-squared (χ2) test or Fisher’s exact test as appropriate. Predictors of renal flare occurrence were investigated using univariable and multivariable Cox regression analysis. p values <0.05 were considered statistically significant.

    Results: De novo renal flares were documented in 146 (7.6%) patients. Among patients who developed at least one renal flare, greater proportions were Asians (30.8% versus 20.2%; p<0.003), had positive baseline anti-dsDNA levels (74.0% versus 61.3%; p=0.003), and had low baseline levels of C3 (51.4% versus 38.2%; p=0.002) and C4 (45.2% versus 35.8%; p=0.030) compared with patients who did not flare. In univariable Cox regression analysis, azathioprine use was protective against renal flares (HR: 0.70; 95% CI: 0.49–0.99; p=0.047), while anti-Sm positivity at baseline showed a trend towards an association with imminent renal flare (HR: 1.68; 95% CI: 0.99–2.85; p=0.057). In multivariable Cox regression analysis adjusting for age, sex, ethnicity, serum creatinine, and variables that differed significantly in univariable analysis, Asian ancestry (HR: 1.60; 95% CI: 1.03–2.49; p=0.036), high mean prednisone dose from baseline until renal flare occurrence or throughout the follow-up (HR: 1.03; 95% CI: 1.02–1.05; p<0.001), and baseline serum creatinine (HR: 1.02; 95% CI: 1.01–1.03; p=0.001) were associated with imminent de novo renal flare, while extra-renal clinical SLE Disease Activity Index 2000 (cSLEDAI) showed a negative association (HR: 0.92; 95% CI: 0.86–0.98; p=0.007). Notably, use of belimumab 1 mg/kg by intravenous (IV) infusion yielded a nearly 3 times decreased hazard of renal flare (HR: 0.37; 95% CI: 0.20–0.68; p=0.001), whereas IV belimumab 10 mg/kg and belimumab 200 mg administered subcutaneously (SC) displayed no clear protection.

    Conclusion: Asian patients appeared particularly susceptible to new-onset renal involvement, corroborating the substantial vulnerability of Asian SLE populations to renal affliction. Add-on low-dose IV belimumab on top of ST appeared protective against renal flares in SLE patients with no prior history of nephritis, while addition of the approved 10 mg/kg IV belimumab dose and SC belimumab yielded no clear protection. Discrepant results between low and high/approved belimumab doses warrant in-depth mechanistic exploration of underlying reasons e.g., potential effects of belimumab on B cell subsets that acquire regulatory properties.

    Reference: [1]Parodis I, Vital EM, et al. Rheumatology (Oxford). 2021;60(9):4348-54.

  • 30.
    Parodis, Ioannis
    et al.
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Lindblom, J.
    Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Toro-Dominguez, D.
    University of Granada, GENYO, Centre for Genomics and Oncological Research: Pfizer, Granada, Spain.
    Borghi, M. O.
    Università degli Studi di Milano and Istituto Auxologico Italiano, Department of Clinical Sciences and Community Health, Milan, Italy.
    Enman, Y.
    Karolinska Institutet, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden.
    Repsilber, Dirk
    Örebro University, School of Medical Sciences.
    Mohan, C.
    University of Houston, Department Biomedical Engineering, Houston, United States of America.
    Alarcon-Riquelme, M.
    University of Granada, GENYO, Centre for Genomics and Oncological Research: Pfizer, Granada, Spain.
    Barturen, G.
    University of Granada, GENYO, Centre for Genomics and Oncological Research: Pfizer, Granada, Spain.
    DRUG REPURPOSING FOR TREATING LUPUS NEPHRITIS BASED ON TRANSCRIPTOME PROFILING AND AUTOIMMUNITY-RELATED SEROLOGICAL MARKERS2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 326-326, article id POS0187Article in journal (Other academic)
  • 31.
    Parodis, Ioannis
    et al.
    Örebro University, School of Medical Sciences. Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden ; Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden; Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
    Lindblom, Julius
    Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
    Barturen, Guillermo
    GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada / Andalusian Regional Government, Granada, Spain, Medical Genomics, Granada, Spain; Department of Genetics, Faculty of Sciences, University of Granada, Granada, Spain.
    Ortega-Castro, Rafaela
    Servicio Andaluz de Salud, Hospital Universitario Reina Sofía, Cordoba, Spain.
    Cervera, Ricard
    Department of Autoimmune Diseases, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Catalonia, Spain.
    Pers, Jacques-Olivier
    Centre Hospitalier Universitaire de Brest, Hopital de la Cavale Blanche, Brest, France.
    Genre, Fernanda
    Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, Spain.
    Hiepe, Falk
    Charité Universitätsmedizin Berlin, Berlin, Germany.
    Gerosa, Maria
    Università degli studi di Milano, Milan, Italy.
    Kovács, László
    University of Szeged, Szeged, Hungary.
    De Langhe, Ellen
    Katholieke Universiteit Leuven and Universitair Ziekenhuis Leuven, Leuven, Belgium.
    Piantoni, Silvia
    Rheumatology and Clinical Immunology Unit, Department of Clinical and Experimental Sciences, Azienda Socio Sanitaria Territoriale Spedali Civili and University of Brescia, Brescia, Italy.
    Stummvoll, Georg
    Medical University of Vienna, Vienna, Austria.
    Vasconcelos, Carlos
    Centro Hospitalar do Porto, Porto, Portugal.
    Vigone, Barbara
    Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
    Witte, Torsten
    Hannover Medical School, Hannover, Germany.
    Alarcón-Riquelme, Marta E.
    GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada / Andalusian Regional Government, Granada, Spain, Medical Genomics, Granada, Spain; Department of Environmental Medicine, Karolinska Institute, Stockholm, Sweden.
    Beretta, Lorenzo
    Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
    Molecular characterisation of lupus low disease activity state (LLDAS) and DORIS remission by whole-blood transcriptome-based pathways in a pan-European systemic lupus erythematosus cohort2024In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, article id ard-2023-224795Article in journal (Refereed)
    Abstract [en]

    OBJECTIVES: To unveil biological milieus underlying low disease activity (LDA) and remission versus active systemic lupus erythematosus (SLE).

    METHODS: We determined differentially expressed pathways (DEPs) in SLE patients from the PRECISESADS project (NTC02890121) stratified into patients fulfilling and not fulfilling the criteria of (1) Lupus LDA State (LLDAS), (2) Definitions of Remission in SLE remission, and (3) LLDAS exclusive of remission.

    RESULTS: We analysed data from 321 patients; 40.8% were in LLDAS, and 17.4% in DORIS remission. After exclusion of patients in remission, 28.3% were in LLDAS. Overall, 604 pathways differed significantly in LLDAS versus non-LLDAS patients with an false-discovery rate-corrected p (q)<0.05 and a robust effect size (dr)≥0.36. Accordingly, 288 pathways differed significantly between DORIS remitters and non-remitters (q<0.05 and dr≥0.36). DEPs yielded distinct molecular clusters characterised by differential serological, musculoskeletal, and renal activity. Analysis of partially overlapping samples showed no DEPs between LLDAS and DORIS remission. Drug repurposing potentiality for treating SLE was unveiled, as were important pathways underlying active SLE whose modulation could aid attainment of LLDAS/remission, including toll-like receptor (TLR) cascades, Bruton tyrosine kinase (BTK) activity, the cytotoxic T lymphocyte antigen 4 (CTLA-4)-related inhibitory signalling, and the nucleotide-binding oligomerization domain leucine-rich repeat-containing protein 3 (NLRP3) inflammasome pathway.

    CONCLUSIONS: We demonstrated for the first time molecular signalling pathways distinguishing LLDAS/remission from active SLE. LLDAS/remission was associated with reversal of biological processes related to SLE pathogenesis and specific clinical manifestations. DEP clustering by remission better grouped patients compared with LLDAS, substantiating remission as the ultimate treatment goal in SLE; however, the lack of substantial pathway differentiation between the two states justifies LLDAS as an acceptable goal from a biological perspective.

  • 32.
    Ravichandran, N.
    et al.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Clinical Immunology and Rheumatology, Lucknow, India.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institute, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Gupta, L.
    The Royal Wolverhampton NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom; The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre The University of Manchester, Manchester, United Kingdom.
    Katsuyuki Shinjo, S.
    University of São Paulo, Division of Rheumatology, Faculdade de Medicina FMUSP, Sao Paulo, Brazil.
    Ziade, N.
    Saint Joseph University of Beirut, Rheumatology Department, Bayrut, Lebanon; Hôtel-Dieu de France, Rheumatology Department, Bayrut, Lebanon.
    Milchert, M.
    Pomeranian Medical University, Department of Internal Medicine, Rheumatology, Diabetology, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Cavagna, L.
    The University of Pavia, Rheumatology Unit, Dipartimento di Medicine Interna e Terapia Medica, Pavia, Italy.
    Tan, A. L.
    University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom; Trust Headquarters, Leeds Teaching Hospitals NHS Trust, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom.
    Lilleker, J. B.
    The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre The University of Manchester, Manchester, United Kingdom; Salford Royal, Manchester Centre for Clinical Neurosciences, Salford, United Kingdom.
    Nune, A.
    Southport & Ormskirk Hospital NHS Trust, Southport, United Kingdom.
    Pauling, J.
    University of Bristol, Bristol Medical School Translational Health Sciences, Bristol, United Kingdom; North Bristol NHS Trust, Department of Rheumatology, Bristol, United Kingdom.
    Wincup, C.
    Rayne Building, UCL, Department of Rheumatology, Division of Medicine, London, United Kingdom; University College London, Centre for Adolescent Rheumatology Versus Arthritis, London, United Kingdom.
    Katchamart, W.
    Faculty of Medicine Siriraj Hospital, Mahidol University, Division of Rheumatology, Department of Medicine, Bangkok, Thailand.
    Goo, P. A.
    Queen Savang Vadhana Memorial Hospital, Department of Medicine, Tambon Si Racha, Thailand.
    Study, C.
    Karolinska Institute, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; The Royal Wolverhampton NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom.
    Chinoy, H.
    The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre The University of Manchester, Manchester, United Kingdom; Manchester University NHS Foundation Trust, National Institute for Health Research Manchester Biomedical Research Centre, Manchester, United Kingdom; Salford Royal, Department of Rheumatology, Salford, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, United States of America.
    Agarwal, V.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Clinical Immunology and Rheumatology, Lucknow, India.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Rheumatology Department, London, United Kingdom.
    PREVALENCE, CHARACTERISTICS, AND PREDICTORS OF BREAKTHROUGH COVID-19 INFECTIONS IN PATIENTS WITH RHEUMATOID ARTHRITIS: DATA FROM THE COVID-19 VACCINATION IN AUTOIMMUNE DISEASE (COVAD) STUDY2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 56-56, article id OP0081Article in journal (Other academic)
    Abstract [en]

    Background: Global data on COVID-19 breakthrough infections (BI) following COVID-19 vaccination among autoimmune rheumatic diseases (AIRDs) and especially rheumatoid arthritis (RA) is scarce.

    Objectives: This study aimed to examine the characteristics of COVID-19 BI among patients with RA and compare them with AIRDs and healthy controls (HCs).

    Methods: A global e-survey, January-May 2022, collected data on COVID-19 vaccination, and BI in patients with RA, AIRDs, non-rheumatic autoimmune disease (nrAIDs), and HCs. BI was defined as infection after both primary or booster vaccine doses. Severe BI was defined as the need for hospitalization, including intensive unit care, oxygen therapy, or advanced treatment in the form of monoclonal antibodies.

    Results: Of the 9595 vaccinated respondents of the e-survey, 3224 (33.6%) reported COVID-19. One BI was reported in 323/1802 (17.9%) patients with RA, 584/3869 (15.0%) patients with other AIRDs, and 467/3435 (13.5%) HCs. Similarly, second BI was reported by 280 (8.6%); 42 (2.3%) among RA, 90 (2.3%) among other AIRDs, and 124 (3.6%) among HCs.

    The prevalence of first BI in patients with RA was higher than that in those with AIRDs (OR=1.2; 95%CI=1.1-1.4; p=0.001) and HCs (OR=1.4; 95%CI=1.2-1.6; p<0.001), but similar to nrAIDs (p=0.783). The prevalence of second BI was lower in patients with RA than in HCs (OR=0.6; 95%CI=0.4-0.9; p=0.012) and nrAIDs (OR=0.4; 95%CI=0.2-0.7; p=0.004), but similar to AIRDs (p=0.991). When compared with HCs, patients with RA reported significantly higher joint pain, hospitalizations, and need for advanced treatment at first BI. Patients with RA from very high HDI countries had lower hazard of first BI than those from high HDI countries (HR=0.026; 95%CI=0.001-0.6; p=0.027). Rituximab use predicted more frequent hospitalization (OR=3.4; 95%CI=1.3-11.4; p=0.045) and severe BI (OR=3.0; 95%CI=1.2-7.3; p=0.014).

    Conclusion: Nearly one in five patients with RA reported BI. BI prevalence was higher in patients with RA and of higher severity than in HCs. Country HDI was an important determinant of outcomes, suggesting potential impact of environmental dynamics, local vaccination policy, and syndemic constructs that merit further exploration. Rituximab use predicted more frequent hospitalizations and more severe BI.

  • 33.
    Reitsema, R.
    et al.
    Örebro University, School of Medical Sciences. Orebro Univ, Fac Med & Hlth, Orebro, Sweden..
    Hesselink, B. C.
    University Medical Center Groningen, Rheumatology and Clinical Immunology, Groningen, Netherlands.
    Van der Geest, K.
    University Medical Center Groningen, Rheumatology and Clinical Immunology, Groningen, Netherlands.
    Abdulahad, W.
    University Medical Center Groningen, Rheumatology and Clinical Immunology, Groningen, Netherlands.
    Boots, A.
    University Medical Center Groningen, Rheumatology and Clinical Immunology, Groningen, Netherlands.
    Brouwer, E.
    University Medical Center Groningen, Rheumatology and Clinical Immunology, Groningen, Netherlands.
    Heeringa, P.
    University Medical Center Groningen, Rheumatology and Clinical Immunology, Groningen, Netherlands.
    Van Sleen, Y.
    University Medical Center Groningen, Rheumatology and Clinical Immunology, Groningen, Netherlands.
    ABERRANT PHENOTYPE OF CIRCULATING ANTIGEN PRESENTING CELLS IN GIANT CELL ARTERITIS AND POLYMYLAGIA RHEUMATICA2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 1071-1071, article id POS1434Article in journal (Other academic)
    Abstract [en]

    Background: Giant Cell Arteritis (GCA) and Polymyalgia Rheumatica (PMR) are overlapping diseases occurring exclusively in people older than 50 years. Antigen-presenting cells (APCs), including monocytes and dendritic cells (DCs), are main contributors to the immunopathology of GCA and PMR. In GCA tissues, DCs may be prone to activation, leading to chemokine production and recruitment of CD4+ T-cells and monocytes to the arterial wall. However, little is known about APC phenotypes in the peripheral blood at the time of GCA/PMR diagnosis.

    Objectives: We aimed to investigate the phenotype of the circulating monocytes and DCs in GCA and PMR patients.

    Methods: APCs among peripheral blood mononuclear cells (PBMCs) of treatment-naive GCA and PMR patients were compared to those in age- and sex-matched healthy controls (HCs) using flow cytometry (n=15 in each group). Using a 13-colour panel, we identified three monocyte subsets: classical (CD14+CD16-), intermediate (CD14+CD16+), and non-classical (CD14lowCD16+) monocytes. DC subsets were subdivided in CD303+CD11c- plasmacytoid DCs (pDCs), CD11c+CD141+ conventional DCs (cDC1) and CD11c+CD1c+ conventional DCs (cDC2). Each of these subsets was analysed for expression of pattern recognition receptors (Toll-like receptor 4 (TLR4), TLR2) and activation markers (CD86, Programmed Death- Ligand 1 (PD-L1), CD40, HLA-DR, CD11c) by assessing the mean-fluorescence intensity of these markers. Data were analysed by conventional gating strategies and by unsupervised tSNE.

    Results: GCA and PMR patients displayed a monocytosis, which was due to increases in classical and intermediate monocyte counts, whereas the proportion of non-classical monocytes was reduced. Intermediate monocytes of GCA patients had significantly higher TLR2 expression, a similar trend was observed in non-classical monocytes of GCA and PMR patients. A divergent pattern was observed in the expression of activation markers on classical versus non-classical monocytes: classical monocytes of GCA/PMR patients appeared to be less activated, whereas non-classical monocytes showed an increased marker expression compared to HCs (Figure 1). Even though no differences were observed in DC counts in the peripheral blood, cDC2 counts correlated negatively with CRP levels (r=-0.60 for GCA, r=-0.55 for PMR).

    Conclusion: Circulating non-classical monocytes, but not DCs, display an activated phenotype in GCA and PMR patients at diagnosis. These cells are thought to be pro-inflammatory, representing the end stage of monocyte maturation in the blood. In contrast, classical monocytes show reduced expression of activation markers in GCA and PMR patients, potentially signalling either an immature or exhausted phenotype. Shown is the mean fluorescence intensity (MFI) of CD11c on the surface of monocyte subsets and CD1c+ conventional dendritic cells (cDC2). Data are shown for patients with giant cell arteritis (GCA) or polymyalgia rheumatica (PMR) and age-matched healthy controls (HCs), n=15 for each group. Statistics by Mann Whitney U. CD11c expression data for pDCs (no CD11c expression) and cDC1 (too small population) are not shown.

  • 34.
    Sen, P.
    et al.
    Maulana Azad Medical College, Maulana Azad Medical College, New Delhi, India.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; Royal Wolverhampton Hospitals NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom.
    POST COVID-19 SYNDROME IN PATIENTS WITH AUTOIMMUNE RHEUMATIC DISEASES: RESULTS FROM THE COVAD STUDY2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 375-376, article id POS0273Article in journal (Other academic)
    Abstract [en]

    Background: Post COVID-19 syndrome (PCS) is an emerging cause of morbidity and poor quality of life in COVID-19 survivors [1, 2].

    Objectives: We aimed to assess the prevalence, risk factors, outcomes, and association with disease flares of PCS in patients with autoimmune rheumatic diseases (AIRDs) and non-rheumatic autoimmune diseases (nrAIDs), both vulnerable groups understudied in the current literature using data from the 2nd COVID-19 Vaccination in Autoimmune Diseases (COVAD) global multicentre patient self-reported e-survey.

    Methods: The survey was circulated from February to July 2022 by the international COVAD Study Group (157 collaborators from 106 countries), and demographics, comorbidities, AIRD/nrAID status, COVID-19 history, vaccination details, and PROMIS physical and mental function were recorded. PCS was defined as symptom resolution time >90 days following acute COVID-19. Predictors of PCS were analysed using regression models for the different groups.

    Results: 7666 total respondents completed the survey. Of these, 2650 respondents with complete responses had positive COVID-19 infection, and 1677 (45.0% AIRDs, 12.5% nrAIDs, 42.5% HCs) completed the survey >90 days post acute COVID-19. Of these, 136 (8.1%) had PCS. Prevalence of PCS was higher in AIRDs (10.8%) than healthy controls HCs (5.3%) (OR: 2.1; 95%CI: 1.4-3.1, p=0.002). Across the entire cohort, a higher risk of PCS was seen in women (OR: 2.9; 95%CI: 1.1-7.7, p=0.037), patients with long duration of AIRDs/nrAIDs (OR 1.01; 95%CI: 1.0-1.02, p=0.016), those with comorbidities (OR: 2.8; 95%CI: 1.4-5.7, p=0.005), and patients requiring oxygen supplementation for severe acute COVID-19 (OR: 3.8; 95%CI: 1.1-13.6, p=0.039). Among patients with AIRDs, comorbidities (OR 2.0; 95%CI: 1.08-3.6, p=0.026), and advanced treatment (OR: 1.9; 95%CI: 1.08-3.3, p=0.024), or intensive care (OR: 3.8; 95%CI: 1.01-14.4, p=0.047) for severe COVID-19 were risk factors for PCS. Notably, patients who developed PCS had poorer PROMIS global physical [15 (12-17) vs 12 (9-15)] and mental health [14 (11-16) vs 11 (8-14)] scores than those without PCS.

    Conclusion: Individuals with AIRDs have a greater risk of PCS than HCs. Associated comorbid conditions, and advanced treatment or intensive care unit admission for severe COVID-19 confer a higher risk of PCS. It is imperative to identify risk factors for PCS for immediate multidisciplinary management in anticipation of poor physical and mental health.

  • 35.
    Sen, P.
    et al.
    Maulana Azad Medical College, Undergraduate, New Delhi, India.
    R, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Nune, A.
    Southport and Ormskirk Hospital NHS Trust, Southport, United Kingdom.
    Lilleker, J. B.
    School of Biological Sciences, The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Manchester, United Kingdom; Manchester Centre for Clinical Neurosciences, Neurology, Salford, United Kingdom.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, Navi Mumbai, India.
    Kardes, S.
    Istanbul University Faculty of Medicine, Department of Medical Ecology and Hydroclimatology, Istanbul, Turkey.
    Kim, M.
    University of Illinois College of Medicine at Peoria, Department of Internal Medicine, Peoria, United States of America.
    Day, J.
    Royal Melbourne Hospital, Department of Rheumatology, Parkville, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia.
    Milchert, M.
    Pomeranian Medical University in Szczecin, Department of Internal Medicine, Rheumatology, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Gheita, T. A.
    Kasr Al Ainy School of Medicine, Cairo University, Department of Rheumatology, Cairo, Egypt.
    Salim, B.
    Fauji Foundation Hospital, Department of Rheumatology, Rawalpindi, Pakistan.
    Velikova, T.
    University Hospital “Lozenetz”, Sofia University, Department of Clinical Immunology, Sofia, Bulgaria.
    Gracia-Ramos, A. E.
    General Hospital, National Medical Center “La Raza”, Instituto Mexicano del Seguro Social, Department of Internal Medicine, Mexico City, Mexico.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Stockholm, Sweden; Örebro University Faculty of Medicine and Health, Department of Rheumatology, Örebro, Sweden.
    Selva-O'callaghan, A.
    Vall D’hebron General Hospital, Universitat Autonoma de Barcelona, Department of Internal Medicine, Barcelona, Spain.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Department of Rheumatology, London, United Kingdom.
    Chatterjee, T.
    University of Illinois College of Medicine at Peoria, Department of Internal Medicine, Peoria, United States of America.
    Tan, A. L.
    Leeds Teaching Hospitals Trust, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom; University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom.
    Cavagna, L.
    Fondazione I.R.C.C.S. Policlinico San Matteo, Department of Rheumatology, Pavia, Italy; The University of Pavia, Department of Internal Medicine and Medical Therapeutics, Pavia, Italy.
    Saavedra, M. A.
    Hospital de Especialidades Dr. Antonio Fraga Mouret, Centro Médico Nacional La Raza, IMSS, Department of Rheumatology, Mexico City, Mexico.
    Shinjo, S. Katsuyuki
    Universidade de Sao Paulo Faculdade de Medicina FMUSP, Division of Rheumatology, Sao Paulo, Brazil.
    Ziade, N.
    Saint-Joseph University, Department of Rheumatology, Beirut, Lebanon; Hotel-Dieu de France Hospital, Department of Rheumatology, Beirut, Lebanon.
    Knitza, J.
    Friedrich-Alexander-Universität Erlangen- Nürnberg, Department of Rheumatology and Immunology, Erlangen, Germany.
    Kuwana, M.
    Nippon Medical School Graduate School of Medicine, Department of Allergy and Rheumatology, Tokyo, Japan.
    Distler, O.
    University Hospital Zurich, University of Zurich, Department of Rheumatology, Zurich, Switzerland.
    Chinoy, H.
    School of Biological Sciences The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Manchester, United Kingdom; National Institute for Health Research Manchester Biomedical Research Centre, Manchester, United Kingdom; Manchester Academic Health Science Centre, Department of Rheumatology, Salford, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, United States of America.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; Royal Wolverhampton Hospitals NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom.
    COVID-19 VACCINATION-RELATED ADVERSE EVENTS AMONG AUTOIMMUNE DISEASE PATIENTS: RESULTS FROM THE COVID-19 VACCINATION IN AUTOIMMUNE DISEASES (COVAD) STUDY2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 966-967, article id POS1260Article in journal (Other academic)
  • 36.
    Walhelm, T.
    et al.
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection/Rheumatology, Linköping, Sweden.
    Wirestam, L.
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection/Rheumatology, Linköping, Sweden.
    Enman, Y.
    Karolinska Institutet and Karolinska University Hospital, Department of Medicine Solna, Division of Rheumatology, Stockholm, Sweden.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Department of Medicine Solna, Division of Rheumatology, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Sjowall, C.
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection/Rheumatology, Linköping, Sweden.
    FACTORS ASSOCIATED WITH SURVIVAL AND DISCONTINUATION OF ANTIMALARIAL AGENTS IN SYSTEMIC LUPUS ERYTHEMATOSUS: RESULTS FROM A SWEDISH LONGITUDINAL REGISTRY2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 902-903, article id POS1145Article in journal (Other academic)
    Abstract [en]

    Background: Hydroxychloroquine (HCQ) and chloroquine, referred to as antimalarial agents (AMA), are cornerstone drugs in systemic lupus erythematosus (SLE), which inhibit type I interferon release via toll-like receptor binding and increasing the pH in plasmacytoid dendritic cell lysosomes [1]. AMA use has established benefits in SLE, such as improved prognosis and decelerated accrual of organ damage. Use of HCQ is safe for most patients and serious side-effects are uncommon, even during pregnancy. Medical therapy to prevent repeated disease flares is of essential weight in the treatment of SLE. However, it is well-known that non-adherence to prescription of AMA is a considerable problem [2].

    Objectives: The aim of this cross-sectional study was to investigate the frequency of AMA prescription, and evaluate factors associated with ongoing use and discontinuation of AMA in a Swedish SLE population.

    Methods: We retrieved data from the Clinical Lupus Register in North-Eastern Gothia (Swedish acronym: KLURING), a longitudinal research and quality registry, including all prevalent and incident cases of SLE in the Östergötland County from 2008 onwards. All included subjects fulfilled the validated 1982 American College of Rheumatology (ACR) and/or the 2012 Systemic Lupus International Collaborating Clinics (SLICC) classification criteria and had been diagnosed from 1963 onwards. Factors associated with ongoing use and discontinuation of AMA were investigated using logistic regression analysis, Mann-Whitney U test and chi-square test.

    Results: A total of 328 subjects were included in the analysis. The mean age at diagnosis was 40.0 years (range: 3–85; standard deviation [SD]: 17.7) and 85.7% were females. The mean SLICC/ACR damage index (SDI) score at last visit was 2.0 (range: 0—11; SD: 2.5). In total, 92.4% had used AMA at some point during their disease course (“ever” users; Table 1). Data from the last available visit indicated that 73.2% were currently prescribed AMA, exclusively HCQ, yielding a daily mean HCQ dose of 228.0 mg (range: 100—400; SD: 71.0). Among individuals who had discontinued AMA, 25.9% had developed a contraindication, mostly on ophthalmological basis (33.3%). Less common reasons were cardiac conditions (19.0%) and renal failure (9.5%). Subjective side-effects were also common; the most frequently reported were gastrointestinal symptoms (n=20/37). Most common patient-related factor associated with discontinuation was intentional non-adherence (e.g., low motivation; 8/11). Patients who had discontinued AMA showed a higher SDI score at the last visit (mean: 2.9; SD: 2.8; mean follow-up: 20.0 years) compared with patients on AMA (mean: 1.4; SD: 1.8; p=0.001; mean follow-up: 15.3 years). Those who fulfilled the immunological disorder ACR criterion (ACR-10) were more likely to continue on AMA (p=0.003). No significant differences were found regarding gender or smoking status. 

    Conclusion: The vast majority of patients in KLURING had been exposed to AMA, and approximately 25% discontinued AMA therapy during follow-up. The main reason for discontinuation were therapy-related factors, such as contraindications and experience of side-effects. Above 50% of the reported side-effects that led to discontinuation were gastrointestinal symptoms. The group of discontinued AMA users accrued more damage over time.

    References:

    [1]Crow MK, Rönnblom L. Type I interferons in host defence and inflammatory diseases. Lupus Sci Med 2019;6:e000336

    [2]Costedoat-Chalumeau N, Houssiau F, Izmirly P, et al. A Prospective International Study on Adherence to Treatment in 305 Patients With Flaring SLE: Assessment by Drug Levels and Self-Administered Questionnaires. Clin Pharmacol Ther 2018;103:1074-1082

  • 37.
    Weiss, R. J.
    et al.
    Department of Orthopaedic Surgery, Karolinska University Hospital, Stockholm, Sweden.
    Stark, A.
    Department of Orthopaedic Surgery, Karolinska University Hospital, Stockholm, Sweden.
    Wick, M. C.
    Department of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
    Ehlin, A.
    Department of Medicine, Unit of Clinical Epidemiology, Karolinska University Hospital, Stockholm, Sweden.
    Palmblad, K.
    Department of Rheumatology, Astrid Lindgren Children’s Hospital, Stockholm, Sweden.
    Wretenberg, Per
    Örebro University, School of Medical Sciences. Department of Orthopaedic Surgery, Karolinska University Hospital, Stockholm, Sweden.
    Orthopaedic surgery of the lower limbs in 49,802 rheumatoid arthritis patients: results from the Swedish National Inpatient Registry during 1987 to 20012006In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 65, no 3, p. 335-341Article in journal (Refereed)
    Abstract [en]

    Objectives: To analyse changes in the rates of hospital admission and use of orthopaedic surgery to the lower limbs in Swedish patients with rheumatoid arthritis between 1987 and 2001.

    Methods: Data for all rheumatoid patients admitted to hospital between 1987 and 2001 were abstracted from the Swedish National Hospital Discharge Register (SNHDR). The data in the register are collected prospectively, recording all inpatient admissions throughout Sweden. The SNHDR uses the codes for diagnoses at discharge and surgical procedures according to the Swedish version of the International Classification of Diseases (ICD).

    Results: In all, 49,802 individual patients with rheumatoid arthritis were identified, accounting for 159,888 inpatient visits. Hospital admissions for rheumatoid arthritis decreased by 42% (p<0.001) during the period 1987 to 2001. Twelve per cent of all admissions were for a rheumatoid arthritis related surgical procedure to the lower limbs; those admissions decreased markedly (by 16%) between 1987 and 1996, and by 12% between 1997 and 2001, as did the overall number of rheumatoid arthritis related surgical procedures to the lower limbs during both time periods. Between 1997 and 2001, 47% of all rheumatoid arthritis related surgical procedures were total joint arthroplasties. There was an overall trend towards reduced length of hospital stay after orthopaedic surgery to the lower limbs during the study period.

    Conclusions: Rates of hospital admission and rheumatoid arthritis related surgical procedures to the lower limbs in Swedish patients with rheumatoid arthritis decreased between 1987 and 2001. This may reflect trends in disease severity, management, and health outcomes of this disease in Sweden.

  • 38.
    Yoshida, A.
    et al.
    Nippon Medical School Graduate School of Medicine, Department of Allergy and Rheumatology, Tokyo, Japan.
    Kim, M.
    University of Illinois College of Medicine Peoria, Center for Outcomes Research, Department of Internal Medicine, Illinois, United States of America.
    Kuwana, M.
    Nippon Medical School Graduate School of Medicine, Department of Allergy and Rheumatology, Tokyo, Japan.
    R, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Lilleker, J. B.
    The University of Manchester, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester, United Kingdom; Northern Care Alliance NHS Foundation Trust, Manchester Centre for Clinical Neurosciences, Salford, United Kingdom.
    Sen, P.
    Maulana Azad Medical College (MAMC), New Delhi, India.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, Navi Mumbai, India.
    Kardes, S.
    Istanbul University, Department of Medical Ecology and Hydroclimatology, Istanbul Faculty of Medicine, Istanbul, Turkey.
    Day, J.
    Royal Melbourne Hospital, Department of Rheumatology, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.
    Makol, A.
    Mayo Clinic, Division of Rheumatology, Rochester, Minnesota, United States of America.
    Milchert, M.
    Pomeranian Medical University in Szczecin, Department of Rheumatology, Internal Medicine, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Gheita, T. A.
    Cairo University, Rheumatology Department, Kasr Al Ainy School of Medicine, Cairo, Egypt.
    Salim, B.
    Fauji Foundation Hospital, Rheumatology Department, Rawalpindi, Pakistan.
    Velikova, T.
    University Hospital “Lozenetz”, Department of Clinical Immunology, Medical Faculty, Sofia, Bulgaria.
    Gracia-Ramos, A. E.
    National Medical Center “La Raza”, Department of Internal Medicine, General Hospital, Mexico City, Mexico.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Selva-O'callaghan, A.
    Vall D’hebron General Hospital, Universitat Autonoma de Barcelona, Systemic Autoimmune Diseases Unit, Internal Medicine Department, Barcelona, Spain.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Rheumatology Department, London, United Kingdom.
    Chatterjee, T.
    University of Illinois College of Medicine at Peoria, Department of Internal Medicine, Illinois, United States of America.
    Tan, A. L.
    Leeds Teaching Hospitals Trust, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom; University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom.
    Nune, A.
    Southport and Ormskirk Hospital NHS Trust, Department of Rheumatology, Southport, United Kingdom.
    Cavagna, L.
    Università degli studi di Pavia, Rheumatology Unit, Dipartimento di Medicine Interna e Terapia Medica, Pavia, Lombardy, Italy.
    Saavedra, M. A.
    Centro Médico Nacional La Raza, Departamento de Reumatología Hospital de Especialidades Dr. Antonio Fraga Mouret, Mexico City, Mexico.
    Katsuyuki Shinjo, S.
    Universidade de Sao Paulo, Division of Rheumatology, Faculdade de Medicina FMUSP, Sao Paulo, Brazil.
    Ziade, N.
    Saint-Joseph University, Rheumatology Department, Beirut, Lebanon; Hotel-Dieu de France Hospital, Rheumatology Department, Beirut, Lebanon.
    Knitza, J.
    Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen- Nürnberg, Medizinische Klinik 3 - Rheumatologie und Immunologie, Erlangen, Germany.
    Distler, O.
    University Hospital Zurich, University of Zurich, Department of Rheumatology, Zurich, Switzerland.
    Chinoy, H.
    The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Manchester, United Kingdom; Manchester University NHS Foundation Trust, The University of Manchester, National Institute for Health Research Manchester Biomedical Research Centre, Manchester, United Kingdom; Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Department of Rheumatology, Salford, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, Pennsylvania, United States of America.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; Royal Wolverhampton Hospitals NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom.
    IMPAIRED PROMIS PHYSICAL FUNCTION IN IDIOPATHIC INFLAMMATORY MYOPATHY PATIENTS: RESULTS FROM THE MULTICENTER COVAD PATIENT REPORTED E-SURVEY2022In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 81, no Suppl. 1, p. 720-722, article id POS0855Article in journal (Other academic)
  • 39.
    Yoshida, A.
    et al.
    Nippon Medical School Graduate School of Medicine, Department of Allergy and Rheumatology, Tokyo, Japan.
    Li, Y.
    School of Public Health, University of Texas Health Science Center at Houston, Department of Biostatistics and Data Science, Houston TX, United States of America.
    Maroufy, V.
    School of Public Health, University of Texas Health Science Center at Houston, Department of Biostatistics and Data Science, Houston TX, United States of America.
    Kuwana, M.
    Nippon Medical School Graduate School of Medicine, Department of Allergy and Rheumatology, Tokyo, Japan.
    Ravichandran, N.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India.
    Makol, A.
    Mayo Clinic, Division of Rheumatology, Rochester Minnesota, United States of America.
    Sen, P.
    Maulana Azad Medical College, New Delhi, India.
    Lilleker, J. B.
    The University of Manchester, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester, United Kingdom; Northern Care Alliance NHS Foundation Trust, Neurology, Manchester Centre for Clinical Neurosciences, Salford, United Kingdom.
    Agarwal, V.
    Mahatma Gandhi Mission Medical College, Navi Mumbai, Maharashtra, India.
    Kardes, S.
    Istanbul University, Department of Medical Ecology and Hydroclimatology, Istanbul Faculty of Medicine, Istanbul, Turkey.
    Day, J.
    Royal Melbourne Hospital, Department of Rheumatology, Parkville, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; University of Melbourne, Department of Medical Biology, Parkville, Australia.
    Milchert, M.
    Pomeranian Medical University in Szczecin, Department of Internal Medicine, Rheumatology, Diabetology, Geriatrics and Clinical Immunology, Szczecin, Poland.
    Joshi, M.
    Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospitals, Pune, India.
    Gheita, T. A.
    Cairo University, Rheumatology Department, Kasr Al Ainy School of Medicine, Cairo, Egypt.
    Salim, B.
    Fauji Foundation Hospital, Rheumatology Department, Rawalpindi, Pakistan.
    Velikova, T.
    University Hospital “Lozenetz”, Sofia University St. Kliment Ohridski, Department of Clinical Immunology, Medical Faculty, Sofia, Bulgaria.
    Gracia-Ramos, A. E.
    General Hospital, National Medical Center “La Raza”, Instituto Mexicano del Seguro Social, Department of Internal Medicine, Mexico City, Mexico.
    Parodis, Ioannis
    Örebro University, School of Medical Sciences. Örebro University Hospital. Karolinska Institutet and Karolinska University Hospital, Division of Rheumatology, Department of Medicine Solna, Stockholm, Sweden; Örebro University, Department of Rheumatology, Faculty of Medicine and Health, Örebro, Sweden.
    Nikiphorou, E.
    King’s College London, Centre for Rheumatic Diseases, London, United Kingdom; King’s College Hospital, Rheumatology Department, London, United Kingdom.
    Tan, A. L.
    Leeds Teaching Hospitals Trust, NIHR Leeds Biomedical Research Centre, Leeds, United Kingdom; University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, United Kingdom.
    Nune, A.
    Southport and Ormskirk Hospital NHS Trust, Southport, United Kingdom.
    Cavagna, L.
    Fondazione I.R.C.C.S. Policlinico San Matteo, Department of Rheumatology, Pavia, Italy; Università degli studi di Pavia, Rheumatology Unit, Dipartimento di Medicine Interna e Terapia Medica, Pavia, Italy.
    Saavedra, M. A.
    Centro Médico Nacional La Raza, IMSS, Departamento de Reumatología Hospital de Especialidades Dr. Antonio Fraga Mouret, Mexico City, Mexico.
    Katsuyuki Shinjo, S.
    Universidade de Sao Paulo, Division of Rheumatology, Faculdade de Medicina FMUSP, Sao Paulo, Brazil.
    Ziade, N.
    Saint- Joseph University, Rheumatology Department, Beirut, Lebanon; Hotel- Dieu de France Hospital, Rheumatology Department, Beirut, Lebanon.
    Knitza, J.
    Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen- Nürnberg, Medizinische Klinik 3 - Rheumatologie und Immunologie, Erlangen, Germany.
    Distler, O.
    University Hospital Zurich, University of Zurich, Department of Rheumatology, Zurich, Switzerland.
    Chinoy, H.
    The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Manchester, United Kingdom; The University of Manchester, National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Department of Rheumatology, Salford, United Kingdom.
    Aggarwal, R.
    University of Pittsburgh School of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, Pennsylvania, United States of America.
    Gupta, L.
    Sanjay Gandhi Postgraduate Institute of Medical Sciences, Department of Clinical Immunology and Rheumatology, Lucknow, India; The University of Manchester, Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Manchester, United Kingdom; Royal Wolverhampton Hospitals NHS Trust, Department of Rheumatology, Wolverhampton, United Kingdom; City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, United Kingdom.
    IMPAIRED HEALTH-RELATED QUALITY OF LIFE IN PATIENTS WITH IDIOPATHIC INFLAMMATORY MYOPATHIES: A CROSS-SECTIONAL ANALYSIS FROM AN INTERNATIONAL SURVEY2023In: Annals of the Rheumatic Diseases, ISSN 0003-4967, E-ISSN 1468-2060, Vol. 82, no Suppl. 1, p. 952-953, article id POS1231Article in journal (Other academic)
    Abstract [en]

    Background: Comprehensive and large-scale assessment of health-related quality of life in patients with idiopathic inflammatory myopathies (IIMs) worldwide is lacking. The second COVID-19 vaccination in autoimmune disease (COVAD-2) study [1] is an international, multicentre, self-reported e-survey assessing several aspects of COVID-19 infection and vaccination as well as validated patient-reported outcome measures (PROMs) to outline patient experience in various autoimmune diseases (AIDs), with a particular focus on IIMs.

    Objectives: To investigate physical and mental health in a global cohort of IIM patients compared to those with non-IIM autoimmune inflammatory rheumatic diseases (AIRDs), non-rheumatic AIDs (NRAIDs), and those without AIDs (controls), using Patient-Reported Outcome Measurement Information System (PROMIS) global health data obtained from the COVAD-2 survey.

    Methods: Demographics, AID diagnoses, comorbidities, disease activity, treatments, and PROMs were extracted from the COVAD-2 database. The primary outcomes were PROMIS Global Physical Health (GPH) and Global Mental Health (GMH) scores. Secondary outcomes included PROMIS physical function short form-10a (PROMIS PF-10a), pain visual analogue scale (VAS), and PROMIS Fatigue-4a scores. Each outcome was compared between IIMs, non-IIM AIRDs, NRAIDs, and controls. Factors affecting GPH and GMH scores in IIMs were identified using multivariable regression analysis.

    Results: A total of 10,502 complete responses from 1582 IIMs, 4700 non-IIM AIRDs, 545 NRAIDs, and 3675 controls, which accrued as of May 2022, were analysed. Patients with IIMs were older [59±14 (IIMs) vs. 48±14 (non-IIM AIRDs) vs. 45±14 (NRAIDs) vs. 40±14 (controls) years, p<0.001] and more likely to be Caucasian [82.7% (IIMs) vs. 53.2% (non-IIM AIRDs) vs. 62.4% (NRAIDs) vs. 34.5% (controls), p<0.001]. Among IIMs, dermatomyositis (DM) and juvenile DM were the most common (31.4%), followed by inclusion body myositis (IBM) (24.9%). Patients with IIMs were more likely to have comorbidities [68.1% (IIMs) vs. 45.7% (non-IIM AIRDs) vs. 45.1% (NRAIDs) vs. 26.3% (controls), p<0.001] including mental disorders [33.4% (IIMs) vs. 28.2% (non-IIM AIRDs) vs. 28.4% (NRAIDs) vs. 17.9% (controls), p<0.001].

    GPH median scores were lower in IIMs compared to NRAIDs or controls [13 (interquartile range 10–15) IIMs vs. 13 (11–15) non-IIM AIRDs vs. 15 (13–17) NRAIDs vs. 17 (15–18) controls, p<0.001] and PROMIS PF-10a median scores were the lowest in IIMs [34 (25–43) IIMs vs. 40 (34–46) non-IIM AIRDs vs. 47 (40–50) NRAIDs vs. 49 (45–50) controls, p<0.001]. GMH median scores were lower in AIDs including IIMs compared to controls [13 (10–15) IIMs vs. 13 (10–15) non-IIM AIRDs vs. 13 (11–16) NRAIDs vs. 15 (13–17) controls, p<0.001]. Pain VAS median scores were higher in AIDs compared to controls [3 (1–5) IIMs vs. 4 (2–6) non-IIM AIRDs vs. 2 (0–4) NRAIDs vs. 0 (0–2) controls, p<0.001]. Of note, PROMIS Fatigue-4a median scores were the highest in IIMs [11 (8–14) IIMs vs. 8 (10–14) non-IIM AIRDs vs. 9 (7–13) NRAIDs vs. 7 (4–10) controls, p<0.001].

    Multivariable regression analysis in IIMs identified older age, male sex, IBM, comorbidities including hypertension and diabetes, active disease, glucocorticoid use, increased pain and fatigue as the independent factors for lower GPH scores, whereas coexistence of interstitial lung disease, mental disorders including anxiety disorder and depression, active disease, increased pain and fatigue were the independent factors for lower GMH scores.

    Conclusion: Both physical and mental health are significantly impaired in patients with IIMs compared to those with non-IIM AIDs or those without AIDs. Our results call for greater attention to patient-reported experience and comorbidities including mental disorders to provide targeted approaches and optimise global well-being in patients with IIMs.

1 - 39 of 39
CiteExportLink to result list
Permanent link
Cite
Citation style
  • apa
  • ieee
  • modern-language-association-8th-edition
  • vancouver
  • Other style
More styles
Language
  • de-DE
  • en-GB
  • en-US
  • fi-FI
  • nn-NO
  • nn-NB
  • sv-SE
  • Other locale
More languages
Output format
  • html
  • text
  • asciidoc
  • rtf