61 results on '"D. Di Giuseppe"'
Search Results
2. Machine learning phenomics (MLP) combining deep learning with time-lapse-microscopy for monitoring colorectal adenocarcinoma cells gene expression and drug-response
- Author
-
M. D’Orazio, M. Murdocca, A. Mencattini, P. Casti, J. Filippi, G. Antonelli, D. Di Giuseppe, M. C. Comes, C. Di Natale, F. Sangiuolo, and E. Martinelli
- Subjects
Medicine ,Science - Abstract
Abstract High-throughput phenotyping is becoming increasingly available thanks to analytical and bioinformatics approaches that enable the use of very high-dimensional data and to the availability of dynamic models that link phenomena across levels: from genes to cells, from cells to organs, and through the whole organism. The combination of phenomics, deep learning, and machine learning represents a strong potential for the phenotypical investigation, leading the way to a more embracing approach, called machine learning phenomics (MLP). In particular, in this work we present a novel MLP platform for phenomics investigation of cancer-cells response to therapy, exploiting and combining the potential of time-lapse microscopy for cell behavior data acquisition and robust deep learning software architectures for the latent phenotypes extraction. A two-step proof of concepts is designed. First, we demonstrate a strict correlation among gene expression and cell phenotype with the aim to identify new biomarkers and targets for tailored therapy in human colorectal cancer onset and progression. Experiments were conducted on human colorectal adenocarcinoma cells (DLD-1) and their profile was compared with an isogenic line in which the expression of LOX-1 transcript was knocked down. In addition, we also evaluate the phenotypic impact of the administration of different doses of an antineoplastic drug over DLD-1 cells. Under the omics paradigm, proteomics results are used to confirm the findings of the experiments.
- Published
- 2022
- Full Text
- View/download PDF
3. Validity of clinical psoriatic arthritis diagnoses made by rheumatologists in the Swedish National Patient Register
- Author
-
JK Wallman, G-M Alenius, E Klingberg, V Sigurdardottir, S Wedrén, S Exarchou, U Lindström, D Di Giuseppe, J Askling, and LTH Jacobsson
- Subjects
Reumatologi och inflammation ,Rheumatology ,Immunology ,Immunology and Allergy ,General Medicine ,Rheumatology and Autoimmunity - Abstract
Objectives: Knowledge of the correspondence between clinical ICD diagnoses and classification criteria fulfilment is crucial to interpret studies identifying cases via ICD codes. We assessed the degree to which patients registered with ICD-10 diagnoses of psoriatic arthritis (PsA) in the Swedish National Patient Register (NPR) fulfil established PsA classification criteria. Method: Four hundred patients with at least one outpatient visit to one of five rheumatology or internal medicine departments (three university/two county departments across Sweden) in 2013–2015, with a main ICD-10 diagnosis of PsA (L40.5–M07.3), were randomly selected (80 cases/site). Through a structured medical record review, positive predictive values (PPVs) for fulfilment of the following classification criteria were assessed: CASPAR, Moll and Wright, Vasey and Espinoza, and modified ESSG criteria for PsA. A subset analysis regarding CASPAR fulfilment was also performed among cases with available rheumatoid factor and peripheral X-ray status (central CASPAR items; n = 227). Results: Of the 400 patients with a main ICD-10 diagnosis of PsA, 343 (86%) fulfilled at least one of the four PsA classification criteria. PPVs for the different criteria were: CASPAR 69% (82% in the subset analysis), Moll and Wright 51%, Vasey and Espinoza 76%, and modified ESSG 64%. Overall, only 6.5% of the 400 PsA diagnoses were judged as clearly incorrect by the medical record reviewers. Conclusion: The validity of rheumatologist-made, clinical ICD-10 diagnoses for PsA in the Swedish NPR is good, with PPVs of 69–82% for CASPAR fulfilment and 86% for meeting any established PsA classification criteria.
- Published
- 2022
- Full Text
- View/download PDF
4. Machine learning phenomics (MLP) combining deep learning with time-lapse-microscopy for monitoring colorectal adenocarcinoma cells gene expression and drug-response
- Author
-
M, D'Orazio, M, Murdocca, A, Mencattini, P, Casti, J, Filippi, G, Antonelli, D, Di Giuseppe, M C, Comes, C, Di Natale, F, Sangiuolo, and E, Martinelli
- Subjects
Machine Learning ,Microscopy ,Multidisciplinary ,Phenotype ,Deep Learning ,Gene Expression ,Humans ,Phenomics ,Adenocarcinoma ,Colorectal Neoplasms ,Time-Lapse Imaging ,Settore ING-INF/07 - Abstract
High-throughput phenotyping is becoming increasingly available thanks to analytical and bioinformatics approaches that enable the use of very high-dimensional data and to the availability of dynamic models that link phenomena across levels: from genes to cells, from cells to organs, and through the whole organism. The combination of phenomics, deep learning, and machine learning represents a strong potential for the phenotypical investigation, leading the way to a more embracing approach, called machine learning phenomics (MLP). In particular, in this work we present a novel MLP platform for phenomics investigation of cancer-cells response to therapy, exploiting and combining the potential of time-lapse microscopy for cell behavior data acquisition and robust deep learning software architectures for the latent phenotypes extraction. A two-step proof of concepts is designed. First, we demonstrate a strict correlation among gene expression and cell phenotype with the aim to identify new biomarkers and targets for tailored therapy in human colorectal cancer onset and progression. Experiments were conducted on human colorectal adenocarcinoma cells (DLD-1) and their profile was compared with an isogenic line in which the expression of LOX-1 transcript was knocked down. In addition, we also evaluate the phenotypic impact of the administration of different doses of an antineoplastic drug over DLD-1 cells. Under the omics paradigm, proteomics results are used to confirm the findings of the experiments.
- Published
- 2022
5. OP0064 FREQUENCY AND PREDICTORS OF MULTIPLE TREATMENT SWITCHING IN RHEUMATOID ARTHRITIS
- Author
-
E. Birgersson Wettersand, D. Di Giuseppe, J. Askling, and K. Chatzidionysiou
- Subjects
Rheumatology ,Immunology ,Immunology and Allergy ,General Biochemistry, Genetics and Molecular Biology - Abstract
BackgroundDespite the significant improvements in the field of rheumatoid arthritis (RA) treatment, a significant and troubling minority of patients remain refractory to multiple disease modifying antirheumatic drugs (DMARDs). The consequence of this ‘’difficult-to-treat’’ state is irreversible damage, risk of co-morbid conditions, and substantial loss of quality of life. Early, precise, and actionable identification of this challenging group of RA patients is crucial for optimal prevention and management.ObjectivesTo assess the frequency and to identify predictors of switching between multiple biological and targeted synthetic DMARDs (b/tsDMARDs) in RA patients in a large national register.MethodsObservational cohort study including RA patients starting a first-ever b/tsDMARD 2009-2018, based on data from the Swedish Quality Rheumatology register. Comorbidities were identified through linkage to the national Patient Register. Baseline (time of RA diagnosis) characteristics of the population were described. Three groups were investigated: A) Patients starting ≥3 treatment courses; B) Patients starting ≥4 treatment courses; and C) Patients starting ≥5 treatment courses. Predictors of multi-switching were explored using univariate and multivariable logistic regression analyses.Results23,908 RA patients were identified. Proportions of patients starting ≥3, ≥4 or ≥5 b/tsDMARDs treatment courses were 7%, 3.2% and 1.6%, during a mean (95% CI) of 3.6 (3.5-3.7), 4.3 (4.2-2.5) and 4.9 (4.7-5.2) years from RA diagnosis, respectively. In Table 1 baseline characteristics for each multi-switching group are summarized. For definition A, the following baseline univariate predictors were identified: female sex (OR=1.57, 95% CI=1.39-1.76), younger age (OR=0.96, 95% CI=0.95-0.96), positive RF (OR=1.36, 95% CI=1.22-1.53) and ACPA (OR=1.40, 95% CI=1.24-1.58), higher DAS28 (OR=1.21, 95% CI=1.15-1.26), HAQ (OR=1.46, 95%=1.33-1.61), pain (OR=1.014, 95% CI=1.012-1.017) and fatigue (OR=1.017, 95% CI=1.014-1.021). In the multivariable logistic regression model, female sex, younger age, higher HAQ, pain and fatigue at baseline were independent predictors of multiple treatment switching. Similar results were found for all three multi-switch definitions. Several comorbidities (i.e. heart failure, ischemic heart disease, malignancy, renal failure) were associated with a lower risk for multiple treatment switching, suggestive of medical contraindications for b/tsDMARDs.Table 1.Baseline (time of RA diagnosis) characteristicsAll patientsMulti-switching definitionA: ≥3 b/tsDMARDsB: ≥4 b/tsDMARDsC: ≥5 b/tsDMARDsN, %23 908 (100)1677 (7)755 (3.2)385 (1.6)Age (years), mean (SD)59.5 (15.2)50.3 (14.5)50.0 (14.6)47.6 (14.7)Sex (male) %30.7%22.5%22.4%21.6%CRP, mg/L (median, IQR)8.9 (4-22)10 (4-24)10 (4.2-25)10 (4-29)ESR (median, IQR)23 (12-40)23 (25-39)23 (11-37.5)22 (11.8-36.3)Patient global, VAS 0-100 (mean, SD)48.8 (26.8)58.4 (25.3)59.8 (24.5)60.8 (24.0)Patient pain, VAS 0-100 (mean, SD)50.2 (26.8)59.2 (25.4)59.8 (25.2)59.9 (25.2)Fatigue, VAS 0-100 (mean, SD)48.5 (28.8)61.2 (26.5)63.1 (25.9)64.6 (23.6)HAQ (mean, SD)0.97 (0.66)1.1 3(0.64)1.15 (0.64)1.13 (0.63)Swollen joint count (median, IQR)5 (2-9)6 (3-10)6 (3-10)6 (3-10)Tender joint count (median, IQR)5 (2-9)6 (3-12)6 (3-12)7 (3-11.25)Concomitant csDMARD, %52.5%50.4%51.7%51.4%Reumatoid factor positive, %63.1%69.6%69.8%67.8%ACPA positive, %67.0%73.4%74.3%72.7%DAS28-ESR (mean, SD)4.62 (1.51)5.00 (1.41)5.10 (1.37)5.12 (1.33)VAS general health physician (mean, SD)42.28 (23.02)45.30 (22.19)45.68 (20.69)51.40 (25.83)ConclusionIn this large national observational cohort, multiple treatment switching, indicative of difficult to treat RA, was observed in a significant proportion of patients, ranging between around 2 to 7% during the first 5 years from time of diagnosis. Risk factors include female gender, younger age, higher HAQ, pain and fatigue at the time of RA diagnosis, suggesting increased attention to this challenging group of patients.Disclosure of InterestsEmma Birgersson Wettersand: None declared, Daniela Di Giuseppe: None declared, Johan Askling Grant/research support from: Karolinska Institutet has entered into agreements between Karolinska Institutet (JA as principal investigator) with AbbVie, BMS, MSD, Eli Lilly, Pfizer, Roche, Samsung Bioepis, Sanofi and UCB, mainly regarding safety monitoring of anti-rheumatic therapies., Katerina Chatzidionysiou Consultant of: consultancy fees from Eli Lilly, AbbVie and Pfizer.
- Published
- 2022
- Full Text
- View/download PDF
6. POS0932 UPTAKE OF NEWER BIOLOGIC AND TARGETED SYNTHETIC DMARDs IN PSORIATIC ARTHRITIS, RESULTS FROM FOUR NORDIC BIOLOGIC REGISTRIES
- Author
-
B. Glintborg, D. DI Giuseppe, J. K. Wallman, D. Nordström, B. Gudbjornsson, M. L. Hetland, J. Askling, G. Gröndal, T. Sokka-Isler, S. Aarrestad Provan, and U. Lindström
- Subjects
Rheumatology ,Immunology ,Immunology and Allergy ,General Biochemistry, Genetics and Molecular Biology - Abstract
BackgroundThe treatment landscape in psoriatic arthritis (PsA) is changing, including newer biologic and targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs) with different modes of action becoming available. However, the most effective treatment strategy in routine care remains to be established.ObjectivesTo explore the uptake and treatment patterns of newer b/tsDMARDs, namely JAK-inhibitors (JAKi; baricitinib, tofacitinib, upadacitinib), IL-17-inhibitors (ixekizumab, secukinumab), abatacept, apremilast, and ustekinumab in PsA patients from the Nordic countries. Furthermore, to describe patient characteristics and extra-musculoskeletal manifestations at treatment start (=baseline).MethodsObservational cohort study, using prospectively collected routine care data from 4 Nordic rheumatology registries. Treatments (newer b/tsDMARDs with tumor-necrosis-factor inhibitors (TNFi) as the reference) initiated from January 2009 until December 2020 and corresponding baseline patient characteristics were identified. Linkage to national patient registries was used to identify previous extra-musculoskeletal manifestations (0-5 years). Country-level data were pooled for analyses. Uptake of each drug was explored as the cumulative number of treatment starts (a) overall, irrespective of previous b/tsDMARD experience, and (b) in b/tsDMARD-naïve patients. Each patient could contribute >1 treatment course.ResultsOverall, 13,364 unique patients contributing 24,325 treatment courses with either a newer b/tsDMARD (4,855, 20%) or a TNFi (19,470, 80%, whereof 10,897 were started year 2015-20) were identified. For the sub-group of 11,892 first b/tsDMARD treatment courses, 1,009 (8%) were a newer b/tsDMARD (10,883 were a TNFi, whereof 5,956 were started year 2015-20).Secukinumab dominated the newer b/tsDMARD uptake (1,848 new-starts, Figure 1). Ustekinumab-uptake increased over time both overall and in b/tsDMARD-naïve patients. In b/tsDMARD-naïve patients, apremilast had the fastest uptake (490 new-starts) (Figure 1). Use of JAKi was limited, especially in b/tsDMARD-naïve patients.Figure 1.Patients starting a newer b/tsDMARD tended to have longer disease duration and slightly higher disease activity at baseline (DAS28, patient-reported outcomes) than TNFi initiators (Table 1). Previous extra-musculoskeletal manifestations (uveitis, IBD) were rare, and with similar distributions across treatments (Table 1).Table 1.Baseline characteristics upon treatment startAbata-ceptApre-milastBari-citinibIxe-kizumabSecuki-numabTofa-citinibUpada-citinibUste-kinumabAny TNFiCumulative uptake, n3629351063421848494669119470Male gender, %334227384033333744Age54 (12)53 (12)55 (13)52 (13)51 (13)54 (13)52 (10)50 (12)49 (13)b/tsDMARD treatment number, %1952911149020562191512262518171925≥3723378746173836219Disease duration, yrs9 (8)8 (8)10 (8)10 (8)9 (9)11 (10)8 (8)8 (9)7 (8)Pain, VAS (0-100)63 (21)61 (23)64 (23)64 (25)63 (24)66 (23)75 (17)64 (23)59 (24)DAS284.73 (1.34)4.04 (1.35)3.95 (1.36)4.24 (1.19)4.13 (1.36)4.49 (1.33)4.74 (0.88)4.19 (1.32)4.07 (1.29)Uveitis, %*323123022IBD, %*113111-31Numbers are mean (SD) unless otherwise statedIBD: inflammatory bowel disease, bDMARD: biologic DMARD, ts: targeted synthetic*0-5 years previously, available all study period for Iceland, Sweden, Finland until 31Dec2018, not available for DenmarkConclusionIn this cross-country collaboration we were able to explore uptake of newer b/tsDMARDs. TNFi still dominates compared to newer b/tsDMARDs in routine care treatment of PsA. Newer b/tsDMARDs are mainly used in patients with several previous treatment failures, i.e. with longer disease duration and higher disease activity, indicating difficult to treat disease. Further studies are planned to explore real-world treatment patterns and outcomes.AcknowledgementsBG and DdiG contributed equally.Partly funded by NordForsk and Foreum grants. On behalf of the Danish DANBIO, Swedish SRQ, Norwegian NOR-DMARD, Finnish ROB-FIN and Icelandic ICEBIO registriesDisclosure of InterestsBente Glintborg Grant/research support from: Pfizer, AbbVie, BMS, Daniela Di Giuseppe: None declared, Johan K Wallman Consultant of: AbbVie, Amgen, Celgene, Eli Lilly, Novartis, Grant/research support from: AbbVie, Amgen, Eli Lilly, Novartis, Pfizer, Dan Nordström: None declared, Björn Gudbjornsson Speakers bureau: Novartis and Amgen, Merete Lund Hetland Grant/research support from: AbbVie, Biogen, BMS, Celltrion, Eli Lilly Denmark A/S, Janssen Biologics B.V, Lundbeck Fonden, MSD, Pfizer, Roche, Samsung Biopis, Sandoz, Novartis., Johan Askling Grant/research support from: Abbvie, Astra-Zeneca, BMS, Eli Lilly, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi, and UCB., Gerdur Gröndal: None declared, Tuulikki Sokka-Isler Grant/research support from: Abbvie, Amgen, BMS, Celgene, Eli Lilly, GSK, Medac, MSD, Novartis, Orion Pharma, Pfizer, Roche, Sandoz, UCB, Sella Aarrestad Provan: None declared, Ulf Lindström: None declared
- Published
- 2022
- Full Text
- View/download PDF
7. POS0001 CAN SINGLE IMPUTATION TECHNIQUES FOR BASDAI COMPONENTS RELIABLY CALCULATE THE COMPOSITE SCORE IN AXIAL SPONDYLOARTHRITIS PATIENTS?
- Author
-
S. Georgiadis, M. Riek, C. Polysopoulos, A. Scherer, D. DI Giuseppe, G. T. Jones, M. L. Hetland, M. Østergaard, S. H. Rasmussen, J. K. Wallman, B. Glintborg, A. G. Loft, K. Pavelka, J. Zavada, M. Birlik, A. Yazici, B. Michelsen, E. Kristianslund, A. Ciurea, M. J. Nissen, A. M. Rodrigues, M. J. Santos, G. Macfarlane, A. M. Hokkanen, H. Relas, C. Codreanu, C. Mogosan, Z. Rotar, M. Tomsic, B. Gudbjornsson, A. J. Geirsson, P. Hellamand, M. G. H. van de Sande, I. Castrejon, M. Pombo-Suarez, B. Frediani, F. Iannone, and L. Midtbøll Ørnbjerg
- Subjects
Rheumatology ,Immunology ,Immunology and Allergy ,General Biochemistry, Genetics and Molecular Biology - Abstract
BackgroundIn axial spondyloarthritis (axSpA), Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) is a key patient-reported outcome. However, one or more of its components may be missing when recorded in clinical practice.ObjectivesTo determine whether an individual patient’s BASDAI at a given timepoint can be reliably calculated with different single imputation techniques and to explore the impact of the number of missing components and/or differences between missingness of individual components.MethodsReal-life data from axSpA patients receiving tumour necrosis factor inhibitors (TNFi) from 13 countries in the European Spondyloarthritis (EuroSpA) Research Collaboration Network were utilized [1]. We studied missingness in BASDAI components based on simulations in a complete dataset, where we applied and expanded the approach of Ramiro et al. [2]. After introducing one or more missing components completely at random, BASDAI was calculated from the available components and with three different single imputation techniques: possible middle value (i.e. 50) of the component and mean and median of the available components. Differences between the observed (original) and calculated scores were assessed and correct classification of patients as having BASDAIResultsA total of 19,894 axSpA patients with at least one complete BASDAI registration at any timepoint were included. 59,126 complete BASDAI registrations were utilized for the analyses with a mean BASDAI of 38.5 (standard deviation 25.9). Calculating BASDAI from the available components and imputing with mean or median showed similar levels of agreement (Table 1). When allowing one missing component, >90% had a difference of ≤6.9 mm between the original and calculated scores and >95% were correctly classified as BASDAI90% (Figure 1, upper panels). As expected, it was observed that regardless of the BASDAI component set to missing and the imputation technique used, correct classification of patients as BASDAITable 1.Level of agreement between the original and calculated BASDAI and correct classification for BASDAILevel of agreement with Dif≤6.9 mm* (%)Correct classification for BASDAI1 missing componentAvailable93.996.9Value 5073.996.3Mean94.296.8Median93.196.82 missing componentsAvailable83.794.8Value 5040.792.8Mean83.594.8Median82.894.73 missing componentsAvailable71.992.6Value 5028.187.3Mean72.292.6Median69.792.2* The levels of agreement with a difference (Dif) of ≤6.9 mm between the original and calculated scores were based on the half of the smallest detectable change. Agreement of >90% was considered as acceptable. ** Correct classification of >95% was considered as acceptable.Figure 1.Level of agreement between the original and calculated BASDAI and correct classification for BASDAIConclusionBASDAI calculation with available components gave similar results to single imputation of missing components with mean or median. Only when missing one of BASDAI components 5 or 6, single imputation techniques can reliably calculate individual BASDAI scores. However, missing any single component value results in misclassification of patients with original BASDAI scores close to 40.References[1]Ørnbjerg et al. (2019). Ann Rheum Dis, 78(11), 1536-1544.[2]Ramiro et al. (2014). Rheumatology, 53(2), 374-376.AcknowledgementsNovartis Pharma AG and IQVIA for supporting the EuroSpA collaboration.Disclosure of InterestsStylianos Georgiadis Grant/research support from: Novartis, Myriam Riek Grant/research support from: Novartis, Christos Polysopoulos Grant/research support from: Novartis, Almut Scherer Grant/research support from: Novartis, Daniela Di Giuseppe: None declared, Gareth T. Jones Speakers bureau: Janssen, Grant/research support from: AbbVie, Pfizer, UCB, Amgen, GSK, Merete Lund Hetland Grant/research support from: Abbvie, Biogen, BMS, Celltrion, Eli Lilly, Janssen Biologics B.V, Lundbeck Fonden, MSD, Medac, Pfizer, Roche, Samsung Biopies, Sandoz, Novartis, Mikkel Østergaard Speakers bureau: Abbvie, BMS, Boehringer-Ingelheim, Celgene, Eli-Lilly, Hospira, Janssen, Merck, Novartis, Novo, Orion, Pfizer, Regeneron, Roche, Sandoz, Sanofi, UCB, Consultant of: Abbvie, BMS, Boehringer-Ingelheim, Celgene, Eli-Lilly, Hospira, Janssen, Merck, Novartis, Novo, Orion, Pfizer, Regeneron, Roche, Sandoz, Sanofi, UCB, Grant/research support from: Abbvie, BMS, Merck, Celgene, Novartis, Simon Horskjær Rasmussen Grant/research support from: Novartis, Johan K Wallman Consultant of: AbbVie, Amgen, Celgene, Eli Lilly, Novartis, Bente Glintborg Grant/research support from: Pfizer, Abbvie, BMS, Anne Gitte Loft Speakers bureau: AbbVie, Janssen, Lilly, MSD, Novartis, Pfizer, Roche, UCB, Consultant of: AbbVie, Janssen, Lilly, MSD, Novartis, Pfizer, Roche, UCB, Karel Pavelka Speakers bureau: Pfizer, MSD, BMS, UCB, Amgen, Egis, Roche, AbbVie, Consultant of: Pfizer, MSD, BMS, UCB, Amgen, Egis, Roche, AbbVie, Jakub Zavada Speakers bureau: Abbvie, Elli-Lilly, Sandoz, Novartis, Egis, UCB, Consultant of: Abbvie, Elli-Lilly, Sandoz, Novartis, Egis, UCB, Merih Birlik: None declared, Ayten Yazici Grant/research support from: Roche, Brigitte Michelsen Grant/research support from: Novartis, Eirik kristianslund: None declared, Adrian Ciurea Speakers bureau: AbbVie, Eli Lilly, Merck Sharp & Dohme, Novartis, Pfizer, Consultant of: AbbVie, Eli Lilly, Merck Sharp & Dohme, Novartis, Pfizer, Michael J. Nissen Speakers bureau: AbbVie, Eli Lilly, Janssens, Novartis, Pfizer, Consultant of: AbbVie, Eli Lilly, Janssens, Novartis, Pfizer, Ana Maria Rodrigues Speakers bureau: Abbvie, Amgen, Consultant of: Abbvie, Amgen, Grant/research support from: Novartis, Pfizer, Amgen, Maria Jose Santos Speakers bureau: Abbvie, AstraZeneca, Lilly, Novartis, Pfizer, Gary Macfarlane Grant/research support from: GSK, Anna-Mari Hokkanen Grant/research support from: MSD, Heikki Relas Speakers bureau: Abbvie, Celgene, Pfizer, UCB, Viatris, Consultant of: Abbvie, Celgene, Pfizer, UCB, Viatris, Catalin Codreanu Speakers bureau: AbbVie, Amgen, Boehringer Ingelheim, Ewopharma, Lilly, Novartis, Pfizer, Consultant of: AbbVie, Amgen, Boehringer Ingelheim, Ewopharma, Lilly, Novartis, Pfizer, Corina Mogosan: None declared, Ziga Rotar Speakers bureau: Abbvie, Novartis, MSD, Medis, Biogen, Eli Lilly, Pfizer, Sanofi, Lek, Janssen, Consultant of: Abbvie, Novartis, MSD, Medis, Biogen, Eli Lilly, Pfizer, Sanofi, Lek, Janssen, Matija Tomsic Speakers bureau: Abbvie, Amgen, Biogen, Eli Lilly, Janssen, Medis, MSD, Novartis, Pfizer, Sanofi, Sandoz-Lek, Consultant of: Abbvie, Amgen, Biogen, Eli Lilly, Janssen, Medis, MSD, Novartis, Pfizer, Sanofi, Sandoz-Lek, Björn Gudbjornsson Speakers bureau: Amgen, Novartis, Consultant of: Amgen, Novartis, Arni Jon Geirsson: None declared, Pasoon Hellamand Grant/research support from: Novartis, Marleen G.H. van de Sande Speakers bureau: Eli Lilly, Novartis, UCB, Janssen, Abbvie, Consultant of: Eli Lilly, Novartis, UCB, Janssen, Abbvie, Grant/research support from: Eli Lilly, Novartis, UCB, Janssen, Abbvie, Isabel Castrejon: None declared, Manuel Pombo-Suarez Consultant of: Abbvie, MSD, Roche, Bruno Frediani: None declared, Florenzo Iannone Speakers bureau: Abbvie, Amgen, AstraZeneca, BMS, Galapagos, Janssen, Lilly, MSD, Novartis, Pfizer, Roche, UCB, Consultant of: Abbvie, Amgen, AstraZeneca, BMS, Galapagos, Janssen, Lilly, MSD, Novartis, Pfizer, Roche, UCB, Lykke Midtbøll Ørnbjerg Grant/research support from: Novartis
- Published
- 2022
- Full Text
- View/download PDF
8. POS0061 THE RISK OF LUNG CANCER IN RHEUMATOID ARTHRITIS AND IN RELATION TO AUTOANTIBODY POSITIVITY AND SMOKING
- Author
-
K. Chatzidionysiou, D. Di Giuseppe, J. Söderling, A. Catrina, and J. Askling
- Subjects
Rheumatology ,Immunology ,Immunology and Allergy ,General Biochemistry, Genetics and Molecular Biology - Abstract
BackgroundLung cancer is a common malignancy in rheumatoid arthritis (RA)1,2. Since smoking is a risk factor for both (seropositive) RA and lung cancer, it remains unclear whether RA, in itself, increases lung cancer risk.ObjectivesThe aim of this study was to examine whether and to what extent the increased risk of lung cancer in RA may (or may not) be attributable to smoking, and to examine this association, both in terms of absolute and relative risks, specifically in relation to RA serostatus.MethodsWe performed a population-based cohort study of RA patients and individually matched general population reference individuals identified in Swedish registers and from the EIRA early RA study, prospectively followed for lung cancer occurrence 1995 through 2018. We calculated incidence rates and performed Cox regression to estimate hazard ratios (HR) including 95% confidence intervals (CI) of lung cancer, taking smoking and sero-status into account.ResultsOverall, we included 44,101 RA patients (590 incident lung cancers, 56 per 100,000), and 216,495 matched general population individuals (1,691 incident lung cancers, 33 per 100,000), corresponding to a crude HR (95% CI) of 1.76 (1.60-1.93). In subset analyses this increased risk remained after adjustment for smoking (HR=1.77, 95% CI 1.06-2.97). Compared to general population subjects who were never smokers, RA patients who were ever smokers had almost 7 times higher risk of lung cancer.Positive autoantibody status was associated with an at least doubled risk of lung cancer in ACPA positive patients (vs. ACPA negative patients) and double seropositive (vs. double seronegative) patients after adjusting for comorbidities and smoking (Table 1).Table 1.Number of events, person-years of follow-up, number of events per 100,000 person-years, and relative risk of lung cancer according to autoantibody status in the EIRA sub-cohort. Five Hazard ratios are presented: a) crude; b) adjusted for age, sex, index year, county of residency (model A); c) age, sex, index year, county of residency and comorbidities (renal failure, heart failure, ischemic heart disease, COPD, respiratory infections, hospitalization) (model B) c) all the above plus smoking (model C) and d) as model C with packet-years instead of smoking ever vs. never.No of events (person years of follow-up; No of events/100 000 person years)Crude Hazard ratio (95% CI)Model A Hazard ratio* (95% CI)Model BHazard ratio** (95% CI)Model CHazard ratio** (95% CI)Model D with smoking as pack-years instead of ever/neverPositiveNegativeRF (N=2060)30(49,440; 60.7)6(49,440; 12.1)2.78 (1.16-6.69)3.01 (1.25-7.26)2.82 (1.17-6.82)2.44 (1.01-5.89)2.16 (0.88-5.28)ACPA (N=2060)30(49,440; 60.7)6(49,440; 12.1)3.13 (1.30-7.51)3.43 (1.42-8.25)3.22 (1.33-7.77)2.88 (1.19-6.95)3.29 (1.26-8.58)RF and/or ACPA (N=2060)34(49,440; 68.8)2(49,440; 4.0)6.38 (1.53-26.56)7.62 (1.83-31.83)7.20 (1.72-30.11)6.29 (1.51-26.30)5.76 (1.37-24.21)RF and ACPA (positive vs. double negative)(N=1608)26(38,592; 67.4)2(38,592; 5.2)6.67 (1.58-28.08)7.92 (1.87-33.50)7.08 (1.67-29.98)6.21 (1.47-26.33)5.86 (1.37-25.01)The average absolute five-year risk of lung cancer counting from RA diagnosis was 1.3% in ever-smoking seropositive RA. At 20 years the risk was almost 3% in RA overall, and over 4% for patients who were ever smokers and had at least one autoantibody.ConclusionRA seropositivity is a strong and at least seemingly independent risk factor for lung cancer in RA. The absolute risks point to the potential for regular lung cancer screening, at least in seropositive RA.References[1]Simon TA, Thompson A, Gandhi KK, et al. Incidence of malignancy in adult patients with rheumatoid arthritis: A meta-analysis. Arthritis Res Ther Published Online First: 2015.[2]Khurana R, Wolf R, Berney S, et al. Risk of development of lung cancer is increased in patients with rheumatoid arthritis: A large case control study in US veterans. J Rheumatol 2008.Disclosure of InterestsKaterina Chatzidionysiou Consultant of: consultancy fees from Eli Lilly, AbbVie and Pfizer, Daniela Di Giuseppe: None declared, Jonas Söderling: None declared, Anca Catrina: None declared, Johan Askling Grant/research support from: Karolinska Institutet has entered into agreements between Karolinska Institutet (JA as principal investigator) with AbbVie, BMS, MSD, Eli Lilly, Pfizer, Roche, Samsung Bioepis, Sanofi and UCB, mainly regarding safety monitoring of anti-rheumatic therapies
- Published
- 2022
- Full Text
- View/download PDF
9. POS0073 COMPARISON OF TIME FROM METHOTREXATE INITIATION TO START OF A b/tsDMARD IN PSORIATIC ARTHRITIS VERSUS RHEUMATOID ARTHRITIS. A NATIONWIDE REGISTER-BASED STUDY
- Author
-
U. Lindström, D. Di Giuseppe, S. Exarchou, G. M. Alenius, T. Olofsson, E. Klingberg, L. T. H. Jacobsson, J. Askling, and J. K. Wallman
- Subjects
Rheumatology ,Immunology ,Immunology and Allergy ,General Biochemistry, Genetics and Molecular Biology - Abstract
BackgroundIn Sweden, methotrexate (MTX) is recommended as first-line DMARD for both psoriatic arthritis (PsA) and rheumatoid arthritis (RA). Whereas in RA the use of MTX monotherapy is supported by efficacy data from randomized trials, in PsA no clear efficacy has been demonstrated in placebo-controlled trials. Therefore, the use of MTX in PsA is mostly based on clinical experience and results from studies not primarily designed to evaluate MTX efficacy.ObjectivesTo compare time from MTX initiation until start of a biological or targeted synthetic DMARD (b/tsDMARD), as marker of insufficient MTX response, in previously DMARD-naïve, incident cases with PsA and matched, corresponding, reference subjects with RA.MethodsPatients with PsA, having collected a prescription of MTX as their first ever DMARD any time between 2011 through 2018, and with a first ever PsA diagnosis in the Swedish National Patient Register within two years prior to this date, were included. Each individual was required to have a visit in rheumatology, but no visit in dermatology, within 6 weeks prior to MTX initiation, to ensure that PsA rather than psoriasis was the main reason for MTX treatment. For each individual with PsA, a corresponding individual with incident RA was identified, matched on sex, age, and year of MTX initiation. Only PsA cases with an identified RA comparator were included. All individuals with a diagnosis indicating axial spondyloarthritis prior to MTX start were excluded. The data were enriched through linkage to other national registers.Follow-up was defined as the time from MTX initiation until start of any b/tsDMARD. Censoring was performed at the first of death, migration or 31 Dec 2020. Time until start of a b/tsDMARD was compared for PsA and RA through crude survival curves and conditional Cox-regression, crude and adjusted for comorbidity, level of education and patient global health.Results3098 patients with PsA, and their individually matched RA comparators were included. At initiation of MTX, PsA cases had a mean 28-joint disease activity score (DAS28) of 4.0 and RA-controls of 4.6, while patient-reported global health was 51 (of 100) for both groups and number of swollen joints (28-joint count) 4.0 for PsA and 6.8 for RA, Table 1. The comorbidity burden was similar at baseline.Table 1.Characteristics of PsA cases and matched RA controls at start of MTX.Psoriatic arthritis N=3098Rheumatoid arthritis N=3098Age, mean (sd)55 (14)55 (14)Sex, male49%49%Length of education, yrs 20%24% 10-1251%49% >1228%26%Diabetes1, 27.4%6.7%Myocardial infarction11.3%1.8%Malignancy14.7%5.0%Congestive heart disease10.2%0.2%Chronic lung disease11.6%2.2%Use of anti-depressive drugs1, 315.4%11.4%DAS28-CRP, mean (sd)44.0 (1.1)4.6 (1.2)Patient global, mean (sd) 451 (23)51 (23)CRP, median (IQR) 47 (14)10 (21)Tender joint count (28), mean (sd) 45.2 (4.9)7.0 (5.7)Swollen joint count (28), mean (sd) 44.0 (4.1)6.8 (5.2)1) Diagnosis within 5 years. 2) Also identified by collecting ≥1 prescription of anti-diabetics in 1 year. 3) Identified by collecting ≥1 prescription within 1 year before methotrexate start. 4) Data on disease activity variables available for 28-36% of PsA and 56-62% of RA.During a mean follow-up of 4.5 and 4.4 years, 34% and 33% of PsA and RA patients, respectively, started a b/tsDMARD, of whom 63% and 84% had also used ≥1 non-MTX conventional synthetic DMARD before the b/tsDMARD initiation. The crude survival curves for time from MTX initiation until start of a b/tsDMARD were identical for PsA and RA, Figure 1A. The adjusted HR for starting a b/tsDMARD in PsA compared with RA was 0.99 (95% CI 0.90-1.09). No calendar time trends were observed (Figure 1B and C).ConclusionIn this study, the risk of escalating treatment from MTX, by adding or switching to a b/tsDMARD, was identical in PsA cases and matched RA controls. This supports a good response to MTX in PsA, similar to that in RA. Due to the matching, neither the results from the PsA nor the RA populations may be fully generalizable.Disclosure of InterestsUlf Lindström: None declared, Daniela Di Giuseppe: None declared, Sofia Exarchou Consultant of: AbbVie, Amgen, Janssen, Novartis, Grant/research support from: AbbVie, Amgen, Eli Lilly, Novartis, Pfizer, Gerd-Marie Alenius: None declared, Tor Olofsson Consultant of: Merck Sharp & Dohme, Eva Klingberg: None declared, Lennart T.H. Jacobsson Speakers bureau: Janssen, Eli Lilly, Novartis, Consultant of: Janssen, Eli Lilly, Novartis, Johan Askling Grant/research support from: Abbvie, Astra-Zeneca, BMS, Eli Lilly, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi, and UCB., Johan K Wallman Consultant of: AbbVie, Amgen, Celgene, Eli Lilly, Novartis, Grant/research support from: AbbVie, Amgen, Eli Lilly, Novartis, Pfizer
- Published
- 2022
- Full Text
- View/download PDF
10. POS1069 COVERAGE OF THE SWEDISH RHEUMATOLOGY QUALITY REGISTER: TO WHAT DEGREE ARE b/tsDMARD TREATMENTS FOR PSORIATIC ARTHRITIS RECORDED?
- Author
-
D. DI Giuseppe, U. Lindström, J. K. Wallman, L. Ljung, and J. Askling
- Subjects
Rheumatology ,Immunology ,Immunology and Allergy ,General Biochemistry, Genetics and Molecular Biology - Abstract
BackgroundIn a national context, the Swedish Rheumatology Quality register (SRQ) is a major source of information on clinical data for patients treated with biological and targeted synthetic disease-modifying anti-rheumatic drug (b/tsDMARD) for rheumatic diseases. Data from SRQ are fundamental for research on drug effectiveness and safety.ObjectivesTo understand to what degree patients with psoriatic arthritis (PsA) and their dispensed b/tsDMARD treatments are recorded in the SRQ.MethodsWe identified all dispensed oral or subcutaneously administered b/tsDMARDs, approved for use in PsA, from the Swedish Prescribed Drug register (PDR, with 100% coverage) in 2018-2019. We required the patients who received the dispensation to have at least one ICD code for PsA (L405, M070, M071, M072, M073) as main diagnosis from a visit to a rheumatology or internal medicine (IM) unit in the National Patient Register (NPR) before dispensation, but no main diagnosis of rheumatoid arthritis (ICD code: M05, M06 (excluding M06.1 and M06.4), M12.3). Furthermore, to limit the assessment to patients with contemporary contact with the specialized rheumatology care, we also required at least one visit with a PsA main diagnosis from rheumatology/IM during 2017-2019. We then checked if the patients and their treatments were registered in SRQ. In a sensitivity analysis, we excluded patients with a visit in dermatology within 6 weeks prior to the first prescription of each b/tsDMARD, in order to exclude patients being prescribed the drug for cutaneous psoriasis.ResultsIn 2018-2019, a total of 7922 unique b/tsDMARD prescriptions had been dispensed to 6311 patients with PsA, having contemporary contact with the specialized rheumatology care. Of them, 5687 patients were registered in SRQ (90.1%), of which 94.4% with a PsA diagnosis and 95.5% with at least one registration of a b/tsDMARD. The coverage of the single drugs in SRQ, as compared to dispensations in PDR, ranged between 53.5% to 93.3% (69.3% to 95.8% considering only patients also in SRQ), with the tumor necrosis factor inhibitors (TNFi) having the best coverage (79.4-93.3%) (Table 1). In a sensitivity analysis, among the 5290 patients without a main diagnosis of psoriasis before start of treatment, 4919 (93%) were registered in SRQ, of which 94% with a PsA diagnosis and 96% with at least one registration of a b/tsDMARD.Table 1.coverage of the single b/tsDMARD approved for use in PsA patients in the SRQ as compared to the prescribed drug register, 2018-2019ATCDrug*Total patients in PDRPatients with the same drug in SRQPercentagePercentage only considering patients included in SRQL04AB01Etanercept2753234885.3%92.5%L04AB04Adalimumab2436193379.4%88.8%L04AB05Certolizumab pegol24321387.7%89.9%L04AB06Golimumab41839093.3%95.8%L04AA24Abatacept947377.7%83.9%L04AA32Apremilast54729453.7%69.3%L04AC10Secukinumab83365678.8%84.8%L04AC13Ixekizumab1187160.2%74%L04AC05Ustekinumab21913863%76.2%L04AA29Tofacitinib26119474.3%77%* In Sweden intravenously administered drugs are not prescribed and therefore not included in the PDR. Also, some hospitals administer ustekinumab in hospital, due to its high cost. However, all b/tsDMARDs, irrespectively of administration route, can be registered in SRQ.ConclusionThe coverage of b/tsDMARD-treated PsA patients in SRQ as compared to the PDR in Sweden is ranging between 90 and 93%, and SRQ is performing well in the registration of the single drugs, especially for TNFi. These results suggest that research studies based on data from the SRQ are representative of the Swedish b/tsDMARD-treated PsA population.Disclosure of InterestsDaniela Di Giuseppe: None declared, Ulf Lindström: None declared, Johan K Wallman Consultant of: AbbVie, Amgen, Celgene, Eli Lilly, Novartis, Grant/research support from: AbbVie, Amgen, Eli Lilly, Novartis, Pfizer, Lotta Ljung: None declared, Johan Askling Grant/research support from: Abbvie, Astra-Zeneca, BMS, Eli Lilly, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi, and UCB
- Published
- 2022
- Full Text
- View/download PDF
11. POS0637 SAFETY OF b/tsDMARDs FOR RA AS USED IN CLINICAL PRACTICE - RESULTS FROM THE LAST DECADE OF THE ARTIS PROGRAM
- Author
-
T. Frisell, H. Bower, E. Baecklund, D. Di Giuseppe, B. Delcoigne, N. Feltelius, H. Forsblad-D’elia, E. Lindqvist, U. Lindström, and J. Askling
- Subjects
Rheumatology ,Immunology ,Immunology and Allergy ,General Biochemistry, Genetics and Molecular Biology - Abstract
BackgroundWhile the relative efficacy of treatments can be demonstrated in relatively small studies with limited follow-up, most safety concerns are infrequent, requiring longer follow-up and larger populations. This is recognized by the regulatory framework, where data from pivotal randomized controlled trials are usually considered sufficient for demonstrating efficacy and non-toxicity, but post-approval safety studies are required for many years to fully evaluate drug-associated risks. Though such regulatory safety-studies often focus on one drug (vs. all others), clinical decision-making requires data across all available treatment options. Long-standing longitudinal clinical registries, like the Anti-Rheumatic Therapies in Sweden (ARTIS) database, thus have a key role in assessing the relative safety of b/tsDMARDs, allowing simultaneous comparison of all drugs used in clinical practice, with consistent definitions of treatment cohorts, follow-up, and outcomes.ObjectivesTo assess incidence rates of critical safety endpoints for individual b/tsDMARDs used to treat RA, updating previously published reports and including more recently introduced treatments.MethodsNationwide register-based cohort study including all RA patients in Sweden registered as starting any b/tsDMARD between Jan 1st 2010 and Dec 31st 2019, and followed until Dec 31st 2020. The incidence rates of selected outcomes, identified through national healthcare registers, were compared between individual b/tsDMARDs while adjusting for a range of potential confounders (covering demographics, RA-related characteristics and disease activity, and comorbidity) using Inverse Probability of Treatment Weighting. Probabilities were predicted by multinomial logistic regression, regressing all covariates on treatment status. Exposure time was counted from treatment start until stop (+90 days’ lag time), censored at emigration and death.ResultsThere were clear differences between patients starting individual b/tsDMARDs, in particular with TNF inhibitors more often used as a first line b/tsDMARD; sarilumab, baricitinib, and tofacitinib predominantly used later in the treatment course; rituximab used more often for older patients, and non-TNFi generally used more frequently for patients with higher disease activity or comorbidity. Expectedly, these differences translated into differences in the crude rate of safety endpoints.Several differences remained after confounder-adjustment (Table 1), including a higher rate of treatment discontinuation due to adverse events on baricitinib, tofacitinib, and sarilumab. Rituximab was associated with higher rates of several outcomes, but the confounder-adjustment markedly reduced risks and residual confounding likely explain part of the remaining increase. Baricitinib and tofacitinib were associated with higher rates of hospitalised herpes zoster, but not with similarly elevated rates of other serious infections. There were no clear differences in the rate of cardiovascular events or severe depression. Low number of events limit the comparison, in particular for sarilumab and tofacitinib.Table 1.Weighted incidence rate per 1,000 person-years of selected safety outcomes.DMARDNDiscont. due to. adverse eventACSStrokeLiver diseaseHosp. infectionHosp. Herpes zosterHosp. depressionAny hosp.All-cause mortalityETA8244456.24.51.4322.92.315610.8ADA5069465.95.61.1363.51.51669.5INF2832508.25.83.1433.22.019712.7CER2072546.47.02.5343.61.717211.0GOL1796515.96.8-322.8-15411.5ABA3254567.34.71.9362.31.617213.9RTX3990318.46.22.2413.32.419415.1TCZ2619305.75.02.1312.91.616315.7SAR271100---18--298-BARI1665693.04.21.4378.82.617316.7TOFA39282---3212.9-129-Note: Rates based on ConclusionWe found large differences in the rate of treatment discontinuations due to adverse events across b/tsDMARDs, which were not generally mirrored by corresponding differences in the rates for specific serious adverse events.ReferencesN/AAcknowledgementsARTIS has been or is currently supported by agreements with Abbvie, BMS, Eli Lilly, MSD, Pfizer, Roche, Samsung Bioepis, and Sanofi.Disclosure of InterestsThomas Frisell: None declared, Hannah Bower: None declared, Eva Baecklund: None declared, Daniela Di Giuseppe: None declared, Bénédicte Delcoigne: None declared, Nils Feltelius Employee of: NF is employed by the Medical Products Agency (MPA), which is a governmental body. The views in this abstract may not represent the views of the MPA, Helena Forsblad-d’Elia: None declared, Elisabet Lindqvist: None declared, Ulf Lindström: None declared, Johan Askling Grant/research support from: Karolinska Institutet has entered into agreements with the following companies, with JA as PI: Abbvie,BMS, Eli Lilly, Galapagos, Janssen, Pfizer, Roche, Samsung Bioepis and Sanofi.
- Published
- 2022
- Full Text
- View/download PDF
12. Incidence of Clinically Diagnosed Psoriatic Arthritis in Sweden.
- Author
-
Exarchou S, Di Giuseppe D, Klingberg E, Sigurdardottir V, Wedrén S, Lindström U, Turesson C, Jacobsson LTH, Askling J, and Wallman JK
- Subjects
- Humans, Sweden epidemiology, Incidence, Female, Male, Middle Aged, Adult, Aged, Young Adult, Adolescent, Arthritis, Psoriatic epidemiology, Arthritis, Psoriatic drug therapy, Arthritis, Psoriatic diagnosis, Registries, Antirheumatic Agents therapeutic use
- Abstract
Objective: Prior incidence estimates of psoriatic arthritis (PsA) vary considerably. We aimed to assess the annual incidence of clinically diagnosed PsA among adults in Sweden in 2014-2016, overall and stratified by age/sex/education/geography, and to investigate potential time trends in incidence in 2006-2018. Use of disease-modifying antirheumatic drugs (DMARDs) during the 2 years after diagnosis was also examined., Methods: Patients (aged ≥ 18 years) with incident clinically diagnosed PsA in Sweden were identified from the National Patient Register (NPR) and/or the Swedish Rheumatology Quality Register (SRQ). Population statistics, stratification variables, and DMARD information were retrieved from other nationwide registers. Incidence was estimated according to a base case (BC) definition (ie, ≥ 1 main International Classification of Diseases, 10th revision, diagnosis of PsA [L40.5/M07.0-M07.3] from rheumatology/internal medicine in NPR, or a PsA diagnosis in SRQ during the relevant year, and no prior such diagnoses) and 4 different sensitivity analysis case definitions., Results: The mean annual incidence of clinically diagnosed PsA among adults in Sweden in 2014-2016 was estimated at 21.77 per 100,000 person-years (PYs) at risk, according to the BC definition; 17.41 per 100,000 PYs at risk after accounting for diagnostic misclassification; and 15.78 to 28.83 per 100,000 PYs at risk across all sensitivity analyses. Incidence was slightly higher in female individuals, was lower in those with higher education (aged > 12 years), and peaked during the ages of 50 to 59 years. No apparent increasing or decreasing time trend was observed in 2006-2018. Within 2 years of diagnosis, 71.03% of patients had received DMARD therapy (22.37% biologic or targeted synthetic DMARDs)., Conclusion: From 2014 to 2016, the annual incidence of clinically diagnosed PsA in the adult Swedish population was approximately 20 per 100,000 PYs at risk. Two years after diagnosis, almost three-quarters of patients had received DMARD therapy., (Copyright © 2024 by the Journal of Rheumatology.)
- Published
- 2025
- Full Text
- View/download PDF
13. Exposure to antibiotics and risk of latent autoimmune diabetes in adults and type 2 diabetes: results from a Swedish case-control study (ESTRID) and the Norwegian HUNT study.
- Author
-
Edstorp J, Rossides M, Ahlqvist E, Alfredsson L, Askling J, Di Giuseppe D, Grill V, Sorgjerd EP, Tuomi T, Åsvold BO, and Carlsson S
- Subjects
- Humans, Case-Control Studies, Sweden epidemiology, Norway epidemiology, Female, Male, Adult, Middle Aged, Aged, Risk Factors, Diabetes Mellitus, Type 1 epidemiology, Diabetes Mellitus, Type 1 chemically induced, Diabetes Mellitus, Type 1 drug therapy, Diabetes Mellitus, Type 2 epidemiology, Diabetes Mellitus, Type 2 drug therapy, Anti-Bacterial Agents adverse effects, Anti-Bacterial Agents therapeutic use, Latent Autoimmune Diabetes in Adults epidemiology
- Abstract
Aims/hypothesis: Some studies find an increased risk of type 1 diabetes in children exposed to antibiotics. We investigated if exposure to antibiotics increases the risk of latent autoimmune diabetes in adults (LADA) and type 2 diabetes., Methods: We used data from a Swedish case-control study (Epidemiological Study of Risk Factors for LADA and Type 2 Diabetes [ESTRID]: LADA, n=597; type 2 diabetes, n=2065; control participants matched on participation time, n=2386) and a case-control study nested within the Norwegian Trøndelag Health Study (HUNT) (n=82/1279/2050). Anatomical Therapeutic Chemical (ATC) codes indicating antibiotic dispensations were retrieved from the Swedish National Prescribed Drug Register and Norwegian Prescription Database. Multivariable adjusted ORs with 95% CIs were estimated by conditional logistic regression and pooled using fixed-effects inverse-variance weighting., Results: We observed no increased risk of LADA with exposure to antibiotics up to 1 year (OR
pooled 1.15, 95% CI 0.93, 1.41) or 1-5 years (ORpooled 0.98, 95% CI 0.80, 1.20) prior to diagnosis/matching for one or more vs no dispensation of any type of antibiotic. An increased risk was observed for one or more vs no dispensations of narrow-spectrum antibiotics, but not broad-spectrum antibiotics, 6-10 years prior to LADA diagnosis (ORpooled 1.39, 95% CI 1.01, 1.91), which was driven by the Swedish data. There was little evidence of an increased risk of type 2 diabetes associated with antibiotic exposure 1-10 years prior to diagnosis., Conclusions/interpretation: We found no evidence that exposure to broad-spectrum antibiotics up to 10 years prior to diagnosis increases the risk of LADA. There was some indication of increased LADA risk with exposure to narrow-spectrum antibiotics, which warrants further investigation., Competing Interests: Acknowledgements: We thank the participants in ESTRID and HUNT, as well as the administrative personnel, nurses and research team members. Data availability: The de-identified datasets generated and analysed in the current study are available from the corresponding author on reasonable request (ESTRID) and with permission of the HUNT study by applying to the HUNT study data access committee. Funding: The ESTRID study was funded by grants from the Swedish Research Council (2018-03035), Research Council for Health, Working Life and Welfare (FORTE, 2018-00337), Novo Nordisk Foundation (NNF19OC0057274) and Swedish Diabetes Foundation. The ANDIS study was financed by Swedish governmental funding of clinical research (ALF), the Faculty of Medicine at Lund University, the Swedish Research Council (project grant no. 2020-02191 and strategic research area grant no. 2009-1039 [EXODIAB]), the Swedish Foundation for Strategic Research (IRC15-0067 [LUDC-IRC]) and Vinnova Swelife. EA was also funded by the Novo Nordisk Foundation (grant no. NNF21OC0070457), the Crafoord Foundation, the Bo & Kerstin Hjelt Diabetes Foundation and the Albert Påhlsson Foundation. The Trøndelag Health Study (HUNT) is a collaboration between the HUNT Research Centre (Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology), Trøndelag County Council, the Central Norway Regional Health Authority and the Norwegian Institute of Public Health. Authors’ relationships and activities: The authors declare that there are no relationships or activities that might bias, or be perceived to bias, their work. Contribution statement: SC, MR and JE contributed to the conception and design of the work. EA, LA, JA, DDG, VG, EPS, TT and BOA contributed to the acquisition of data. JE performed the analyses. All authors contributed to the interpretation of data. JE drafted the article and all authors edited, reviewed and approved the final version to be published. JE is responsible for the integrity of this work as a whole., (© 2024. The Author(s).)- Published
- 2025
- Full Text
- View/download PDF
14. Toxicity of size separated chrysotile fibres: The relevance of the macrophage-endothelial axis crosstalk.
- Author
-
Mirata S, Almonti V, Passalacqua M, Vernazza S, Bassi AM, Di Giuseppe D, Gualtieri AF, and Scarfì S
- Abstract
Asbestos minerals have been widely exploited due to their physical-chemical properties, and chrysotile asbestos has accounted for about 95% of all asbestos commercially employed worldwide. The exposure to chrysotile, classified like other five amphibole asbestos species as carcinogenic to humans, represents a serious occupational and environmental hazard. Nevertheless, this mineral is still largely employed in about 65% of the countries worldwide, which still allow its "safe use". The complex mechanisms through which the mineral fibres induce toxicity are not yet completely understood. In this regard, the morphometric parameters of asbestos fibres (e.g., length, width, aspect ratio) are known for their fundamental role in determining the degree of pathogenicity. In this context, the potential toxicity of short chrysotile fibres remains widely debated due to the contradictory results from countless studies. Thus, the present study investigated the different toxicity mechanisms of two representative batches of short (length ≤5 µm) and long (length >5 µm) chrysotile fibres obtained by cryogenic milling. The fibre doses were based upon equal mass and size, since due to chrysotile ability to form bundles, it was not possible to calculate the number of fibers applied per cell. The cytotoxic, genotoxic, and pro-inflammatory potential of the two size-separated chrysotile fractions was investigated on human THP-1-derived macrophages and HECV endothelial cells, both separately and in a co-culture setup, mimicking the alveolar pro-inflammatory microenvironment, in time course experiments up to 1 week. Both chrysotile fractions displayed cytotoxic, genotoxic, and pro-inflammatory effects, with results comparable to the well-known damaging effects of crocidolite asbestos, or higher, as in the case of the longer chrysotile fraction. Furthermore, in presence of HECV, fibre-treated macrophages showed prolonged inflammation, indicating an interesting crosstalk between these cells able to sustain a low-grade chronic inflammation in the lung. In conclusion, these results help to shed light on some important open questions on the mechanisms of toxicity of chrysotile asbestos fibres., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Recording of non-musculoskeletal manifestations, comorbidities and safety outcomes in European spondyloarthritis registries: a survey.
- Author
-
Ahmadzay ZF, Heberg J, Jørgensen JB, Ørnbjerg LM, Østergaard M, Møller-Bisgaard S, Michelsen B, Loft AG, Jones GT, Hellamand P, Scherer A, Nissen MJ, Pavelka K, Závada J, Laas K, Vorobjov S, Nordström D, Sokka-Isler T, Regierer AC, Reich A, Gudbjornsson B, Thorarinsdottir K, Iannone F, Favalli EG, van de Sande M, Provan SA, Kvien TK, Rodrigues AM, Gonçalves CF, Codreanu C, Mogosan C, Rotar Z, Prikmajer KP, Castrejon I, Otero-Varela L, Di Giuseppe D, Wallman JK, Ciurea A, Möller B, Kenar-Artın G, Yıldırım TD, Macfarlane GJ, Rotariu O, Glintborg B, and Hetland ML
- Abstract
Objectives: Real-world evidence is needed to inform treatment strategies for patients with PsA and axial SpA (axSpA) who have non-musculoskeletal manifestations (NMMs), various risk factors and comorbidities. International collaboration is required to ensure statistical power and to enhance generalizability. The first step forward is identifying which data are currently being collected. Across 17 registries participating in the European Spondyloarthritis Research Collaboration (EuroSpA), we aimed to map recording practices for NMMs, comorbidities and safety outcomes in patients with PsA and axSpA., Methods: Through a survey with 4,420 questionnaire items, we explored the recording practices of 58 pre-defined conditions (i.e. NMMs, comorbidities and safety outcomes) covering 10 disease areas. In all registries we mapped for each condition whether it was recorded, the recording procedure and the potential to identify it through linkage to other national registries., Results: Conditions were generally recorded at entry into the registry and clinical follow-up visits using a pre-specified list or a coding system. Most registries recorded conditions within the following disease areas: NMMs (number of registries, n = 15-16), cardiovascular diseases ( n = 10-14), gastrointestinal diseases ( n = 12-13), infections ( n = 10-13) and death ( n = 14). Nordic countries had the potential for data linkage and generally had limited recording of conditions in their registry, while other countries had comprehensive recording practices., Conclusion: A wide range of conditions were consistently recorded across the registries. The recording practices of many conditions and disease areas were comparable across the registries. Our findings support the potential for future collaborative research., (© The Author(s) 2024. Published by Oxford University Press on behalf of the British Society for Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
16. Comparing DAPSA, DAPSA28, and DAS28-CRP in Patients With Psoriatic Arthritis Initiating a First Tumor Necrosis Factor Inhibitor Across Nine European Countries.
- Author
-
Linde L, Georgiadis S, Ørnbjerg LM, Rasmussen SH, Michelsen B, Askling J, Di Giuseppe D, Wallman JK, Závada J, Pavelka K, Bernardes M, Matos CO, Glintborg B, Loft AG, Nordström D, Kuusalo L, Möller B, Nissen MJ, Codreanu C, Mogosan C, Gudbjornsson B, Love TJ, Akleylek C, Iannone F, Kvien TK, Rotar Z, Castrejon I, Macfarlane GJ, Hetland ML, and Østergaard M
- Subjects
- Humans, Male, Female, Middle Aged, Europe, Adult, Prospective Studies, Treatment Outcome, Antirheumatic Agents therapeutic use, Tumor Necrosis Factor Inhibitors therapeutic use, Aged, Tumor Necrosis Factor-alpha antagonists & inhibitors, Biomarkers blood, Arthritis, Psoriatic drug therapy, Arthritis, Psoriatic blood, Severity of Illness Index, C-Reactive Protein analysis, Remission Induction
- Abstract
Objective: Because 66/68 joint counts are not always performed in routine care, we aimed to determine which of the modified 28-joint disease activity index for psoriatic arthritis (DAPSA28) or 28-joint disease activity score with C-reactive protein (DAS28-CRP) should be preferred for monitoring disease activity in psoriatic arthritis (PsA) when the original DAPSA (66/68 joints) is not available., Methods: Prospectively collected real-world data of European bionaive patients with PsA initiating a first tumor necrosis factor inhibitor were pooled. Remission and response status were evaluated at 6 months by remission (DAPSA ≤ 4, DAPSA28 ≤ 4, and DAS28-CRP < 2.6), response (75% improvement for DAPSA and DAPSA28), and combined EULAR good/moderate responses for DAS28-CRP. Logistic regression analyses on multiple imputed data were used to identify baseline predictors., Results: Remission and response cohorts included 3,159 and 1,866 patients, respectively. The 6-month proportions achieving remission/response were DAPSA (27%/44%), DAPSA28 (28%/44%), and DAS28-CRP (59%/80%). Of 14 possible baseline predictors, 11 predicted both DAPSA and DAPSA28 remission (8 of which also predicted their response, indicated by "*"): longer disease duration*, male sex*, and higher CRP* were positive, whereas older age*, higher body mass index*, patient fatigue*, and global, physician global, health assessment questionnaire score*, and tender and swollen* joint counts were negative predictors. Eight and five of these predicted DAS28-CRP remission and response, respectively., Conclusion: In patients with PsA, DAPSA28 should be preferred over DAS28-CRP as a substitute for DAPSA when 66/68 joint counts are not available because of the large overlap in remission and response status and in predictors between DAPSA and DAPSA28., (© 2024 The Author(s). Arthritis Care & Research published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
17. Interstitial Lung Disease in Patients With Rheumatoid Arthritis or Psoriatic Arthritis Initiating Biologics and Controls: Data From 5 Nordic Registries.
- Author
-
Provan SA, Ljung L, Kristianslund EK, Michelsen B, Uhlig T, Jonmundsson T, Sexton J, Gudbjornsson B, Di Giuseppe D, Hetland ML, Reynisdottir GB, Glintborg B, Relas H, Aaltonen K, Kvien TK, and Askling J
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Incidence, Scandinavian and Nordic Countries epidemiology, Follow-Up Studies, Lung Diseases, Interstitial epidemiology, Arthritis, Psoriatic drug therapy, Arthritis, Psoriatic epidemiology, Arthritis, Rheumatoid drug therapy, Arthritis, Rheumatoid epidemiology, Arthritis, Rheumatoid complications, Registries, Antirheumatic Agents therapeutic use, Antirheumatic Agents adverse effects, Methotrexate therapeutic use, Biological Products therapeutic use, Biological Products adverse effects
- Abstract
Objective: Interstitial lung disease (ILD) is one of the most common pulmonary manifestations of rheumatoid arthritis (RA), but its prevalence has not been investigated in psoriatic arthritis (PsA). The role of methotrexate (MTX) in ILD development remains under debate. This study (1) compares the incidences of ILD in patients with RA or PsA initiating a first biologic disease-modifying antirheumatic drug (bDMARD) to that in the general population, and (2) investigates the role of MTX comedication on ILD incidence., Methods: Patients were identified in 5 rheumatology registries. Demographics, MTX use, and disease activity were retrieved. Matched subjects from the general population were available from 4 countries. Incidence of ILD during follow-up of up to 5 years was assessed through national patient registries. Subjects with prior ILD were excluded. Adjusted hazard ratios (HRs) were calculated for ILD incidence in patients vs the general population, and for MTX users vs nonusers., Results: During follow-up of 29,478 patients with RA and 10,919 patients with PsA initiating a first bDMARD and 362,087 population subjects, 225, 23, and 251 cases of ILD were identified, respectively. HRs for ILD (vs population subjects) were 9.7 (95% CI 7.97-11.91) in RA and 4.4 (95% CI 2.83-6.97) in PsA. HRs for ILD with MTX comedication (vs nonuse) were 1.0 (95% CI 0.72-1.25) in RA and 0.9 (95% CI 0.38-2.05) in PsA., Conclusion: Among patients with RA and PsA initiating a bDMARD, the risk of ILD was higher than in the general population, and was highest in RA. MTX comedication was not a risk determinant for ILD., (Copyright © 2024 by the Journal of Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
18. Effectiveness of JAK Inhibitors Compared With Biologic Disease-Modifying Antirheumatic Drugs on Pain Reduction in Rheumatoid Arthritis: Results From a Nationwide Swedish Cohort Study.
- Author
-
Eberhard A, Di Giuseppe D, Askling J, Bergman S, Bower H, Chatzidionysiou K, Forsblad-d'Elia H, Kastbom A, Olofsson T, Frisell T, and Turesson C
- Abstract
Objective: To compare the effectiveness of JAK inhibitors (JAKis) and biologic disease-modifying antirheumatic drugs (bDMARDs) on pain in patients with rheumatoid arthritis., Methods: In this retrospective study, we investigated patients with a diagnosis of rheumatoid arthritis, starting treatment with a JAKi (n = 1,827), a tumor necrosis factor inhibitor (TNFi; n = 6,422), an interleukin-6 inhibitor (n = 887), abatacept (n = 1,102), or rituximab (n = 1,149) in 2017 to 2019, using data from several linked Swedish national registers. Differences in change in pain, assessed with a visual analogue scale (0-100 mm), from baseline to 3 months, as well as proportions of patients remaining on initial treatment with low pain (visual analogue scale pain <20) at 12 months, were compared between treatments. Comparisons of treatment responses between JAKis and bDMARDs were evaluated using multivariable linear regression, adjusted for patient characteristics, comorbidities, current comedication, and previous treatment., Results: JAKi treatment was associated with a greater decrease in pain at 3 months compared with TNFi treatment (adjusted mean additional decrease 4.0 mm; 95% confidence interval 1.6-6.3), with similar trends in comparisons with non-TNFi bDMARDs. More patients achieved low pain at 12 months on JAKis compared with TNFis, in particular among those previously treated with at least two bDMARDs (adjusted change contrast 5.3 percentage points; 95% confidence interval 1.0-9.6)., Conclusion: JAKis had a slightly better effect on pain outcomes at 3 and 12 months compared with TNFis, with significantly greater differences in patients previously treated with at least two bDMARDs. The effect of JAKis on pain reduction was at least similar to that of non-TNFi bDMARDs., (© 2024 The Author(s). Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
19. Toxicity and inflammatory potential of mineral fibres: The contribute of released soluble metals versus cell contact direct effects.
- Author
-
Almonti V, Vernazza S, Mirata S, Tirendi S, Passalacqua M, Gualtieri AF, Di Giuseppe D, Scarfì S, and Bassi AM
- Subjects
- Humans, Oxidative Stress drug effects, Apoptosis drug effects, Endothelial Cells drug effects, Endothelial Cells metabolism, DNA Damage drug effects, Asbestos, Serpentine toxicity, THP-1 Cells, Cytokines metabolism, Inflammation chemically induced, Monocytes drug effects, Monocytes metabolism, Metals, Heavy toxicity, NF-kappa B metabolism, Cell Line, Asbestos, Crocidolite toxicity, Zeolites, Mineral Fibers toxicity
- Abstract
Asbestos fibres have been considered an environmental hazard for decades. However, little is known about the attempts of circulating immune cells to counteract their toxicity. We addressed the early effects of fibre-released soluble factors (i.e. heavy metals) in naïve immune cells, circulating immediately below the alveolar/endothelial cell layer. By comparison, the direct fibre effects on endotheliocytes were also studied since these cells are known to sustain inflammatory processes. The three mineral fibres analysed showed that mainly chrysotile (CHR) and erionite (ERI) were able to release toxic metals in extracellular media respect to crocidolite (CRO), during the first 24 h. Nevertheless, all three fibres were able to induce oxidative stress and genotoxic damage in indirectly challenged naïve THP-1 monocytes (separated by a membrane). Conversely, only CHR-released metal ions induced apoptosis, NF-κB activation, cytokines and CD163 gene overexpression, indicating a differentiation towards the M0 macrophage phenotype. On the other hand, all three mineral fibres in direct contact with HECV endothelial cells showed cytotoxic, genotoxic and apoptotic effects, cytokines and ICAM-I overexpression, indicating the ability of these cells to promote an inflammatory environment in the lung independently from the type of inhaled fibre. Our study highlights the different cellular responses to mineral fibres resulting from both the nature of the cells and their function, but also from the chemical-physical characteristics of the fibres. In conclusion, CHR represented the main pro-inflammatory trigger, able to recruit and activate circulating naïve monocytes, through its released metals, already in the first 24 h after inhalation., (© 2024 John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
20. 3D Cell Migration Chip (3DCM-Chip): A New Tool toward the Modeling of 3D Cellular Complex Systems.
- Author
-
Buonvino S, Di Giuseppe D, Filippi J, Martinelli E, Seliktar D, and Melino S
- Subjects
- Humans, Cell Line, Tumor, Coculture Techniques methods, Cell Culture Techniques, Three Dimensional methods, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Fibroblasts cytology, Fibroblasts metabolism, Models, Biological, Cell Movement physiology, Lab-On-A-Chip Devices, Hydrogels chemistry, Human Umbilical Vein Endothelial Cells metabolism
- Abstract
3D hydrogel-based cell cultures provide models for studying cell behavior and can efficiently replicate the physiologic environment. Hydrogels can be tailored to mimic mechanical and biochemical properties of specific tissues and allow to produce gel-in-gel models. In this system, microspheres encapsulating cells are embedded in an outer hydrogel matrix, where cells are able to migrate. To enhance the efficiency of such studies, a lab-on-a-chip named 3D cell migration-chip (3DCM-chip) is designed, which offers substantial advantages over traditional methods. 3DCM-chip facilitates the analysis of biochemical and physical stimuli effects on cell migration/invasion in different cell types, including stem, normal, and tumor cells. 3DCM-chip provides a smart platform for developing more complex cell co-cultures systems. Herein the impact of human fibroblasts on MDA-MB 231 breast cancer cells' invasiveness is investigated. Moreover, how the presence of different cellular lines, including mesenchymal stem cells, normal human dermal fibroblasts, and human umbilical vein endothelial cells, affects the invasive behavior of cancer cells is investigated using 3DCM-chip. Therefore, predictive tumoroid models with a more complex network of interactions between cells and microenvironment are here produced. 3DCM-chip moves closer to the creation of in vitro systems that can potentially replicate key aspects of the physiological tumor microenvironment., (© 2024 Wiley‐VCH GmbH.)
- Published
- 2024
- Full Text
- View/download PDF
21. Effectiveness of secukinumab in radiographic and non-radiographic axial spondyloarthritis: a European routine-care observational study.
- Author
-
Christiansen SN, Horskjær Rasmussen S, Ostergaard M, Pons M, Michelsen B, Pavelka K, Codreanu C, Ciurea A, Glintborg B, Santos MJ, Sari I, Rotar Z, Gudbjornsson B, Macfarlane GJ, Relas H, Iannone F, Laas K, Wallman JK, van de Sande M, Provan SA, Castrejon I, Zavada J, Mogosan C, Nissen MJ, Loft AG, Barcelos A, Erez Y, Pirkmajer KP, Grondal G, Jones GT, Hokkanen AM, Chimenti MS, Vorobjov S, Di Giuseppe D, Kvien TK, Otero-Varela L, van der Horst-Bruinsma I, Hetland ML, and Ørnbjerg LM
- Subjects
- Humans, Male, Female, Adult, Treatment Outcome, Europe, Middle Aged, Antirheumatic Agents therapeutic use, Prospective Studies, Severity of Illness Index, Patient Reported Outcome Measures, Radiography, Remission Induction, Antibodies, Monoclonal, Humanized therapeutic use, Axial Spondyloarthritis drug therapy, Axial Spondyloarthritis etiology
- Abstract
Objectives: To compare the treatment effectiveness of secukinumab in radiographic (r) versus non-radiographic (nr) axial spondyloarthritis (axSpA) patients treated in routine care across Europe., Methods: Prospectively collected data on secukinumab-treated axSpA patients with known radiographic status were pooled from nine countries.Remission rates based on patient-reported outcomes (PROs; Numeric Rating Scale (0-10), for example, pain ≤2/Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) ≤2 and Ankylosing Spondylitis Disease Activity Score (ASDAS) inactive disease (ID) <1.3 after 6/12/24 months of secukinumab treatment were calculated.Remission and drug retention rates in r-axSpA versus nr-axSpA patients were compared by logistic and Cox regression models (unadjusted/adjusted for age+sex/adjusted for multiple confounders)., Results: Overall, 1161 secukinumab-treated patients were included (r-axSpA/nr-axSpA: 922/239). At baseline, r-axSpA patients had longer disease duration and higher C reactive protein, were more often male and HLA-B27 positive and had received fewer prior biological or targeted synthetic disease-modifying antirheumatic drugs compared with nr-axSpA patients, whereas PROs were largely similar.During follow-up, crude PRO remission rates were significantly higher in r-axSpA compared with nr-axSpA patients (6 months: pain≤2: 40%/28%, OR=1.7; BASDAI≤2: 37%/25%, OR=1.8), as were drug retention rates (24 months: 66%/58%, HR 0.73 (ref: r-axSpA)). Proportions of patients achieving ASDAS ID were low for both groups, particularly nr-axSpA (6 months: 11%/8%).However, when adjusting for age+sex, these differences diminished, and after adjusting for multiple confounders, no significant between-group differences remained for either remission or drug retention rates., Conclusion: Crude remission/drug retention rates in European secukinumab-treated patients were higher in r-axSpA compared with nr-axSpA patients. In adjusted analyses, secukinumab effectiveness was similar in both groups, suggesting that observed differences were related to factors other than radiographic status., Competing Interests: Competing interests: SNC: Speaker fees BMS and GE, Research grant from Novartis (paid to the employer). SHR: Research grant from Novartis (paid to the employer). MO: Speaker and/or consultancy fees from AbbVie, BMS, Boehringer-Ingelheim, Celgene, Eli-Lilly, Hospira, Janssen, Merck, Novartis, Novo, Orion, Pfizer, Regeneron, Roche, Sandoz, Sanofi, UCB. Research grants from AbbVie, BMS, Merck, Novartis and UCB. MP: Research grant from Novartis (paid to the employer). Speaker fees: Sandoz. Brigitte Michelsen: Consulting fees from Novartis. Research grant from Novartis (paid to employer). KP: Consultancy fees: AbbVie, UCB, Pfizer, Eli Lilly, Celltrion, MSD and Novartis. Catalin Codreanu: Speaker and consultancy fees from AbbVie, Amgen, Boehringer Ingelheim, Ewopharma, Lilly, Novartis, Pfizer. Adrian Ciurea: None. Bente Glintborg: Research grants from Pfizer, Abbvie, BMS, Sandoz. MJS: Speaker fees from AbbVie, AstraZeneca, Janssen, Lilly, Medac, Novartis, Pfizer. Ismail Sari: None. ZR: Speaker and consultancy fees from Abbvie, Novartis, Eli Lilly, Pfizer, Janssen, SOBI, Swixx BioPharma, AstraZeneca, Amgen, MSD, Medis, Biogen, Eli Lilly, Sanofi, Lek. BG: Speaker and consultancy fees from Novartis and Nordic-Pharma. GJM: Research grant from GSK. HR: Consulting and/or speaking fees from AbbVie, Celgene, Pfizer, UCB, Viatris. FI: None. Karin Laas: Speakers fees from AbbVie, Johnson and Johnson, Novartis, Pfizer. JKW: Speaker fees from AbbVie, Amgen. Research support from AbbVie, Amgen, Eli Lilly, Novartis, Pfizer. MvdS: Consultant for Novartis, AbbVie, Eli Lilly UCB, Speakers fee: Novartis, UCB, Janssen, Grant/research support: UCB, Janssen, Novartis, Eli Lilly. Sella Aarrestad Provan: Consultancy fees and Research grants from Boehringer Ingelheim. IC: Speaker and/or consultancy fees from BMS, Eli-Lilly, Galapagos, Gilead, Janssen, Novartis, MSD, Pfizer, GSK. JZ: Speakers fees from AbbVie, Elli-Lilly, Sandoz, Novartis, Egis, UCB, Sanofi, Astra Zeneca, Sobi. CM: None. MJN: Speaker and/or consultancy fees from AbbVie, Amgen, Eli Lilly, Janssens, Novartis, Pfizer. Research grants from Novartis and Pfizer. AGL: Speaking and/or consulting fees from AbbVie, Janssen, Lilly, MSD, Novartis, Pfizer, UCB. Research grant from Novartis. AB: Speaker and/or consultancy fees from AbbVie, Lilly, Janssen and Novartis. YE: None. Katja Perdan Pirkmajer: Speaker and/or consultancy fees from AbbVie, Novartis, MSD, Medis, Eli Lilly, Pfizer, Lek, Janssen and Boehringer Ingelheim. GG: None. GTJ: Speaker fee from Janssen. Research grants (paid to employer) from AbbVie, Pfizer, UCB, Amgen, GSK. A-MH: Grant/research support from MSD. MSC: None. SV: None. DdG: None. TKK: Speaker and/or consultancy fees from AbbVie, Amgen, Celltrion, Gilead, Novartis, Pfizer, Sandoz, UCB and Grünenthal. LO-V: None. IvdH-B: Speaker and/or consultancy fees from AbbVie, UCB, MSD, Novartis, Lilly. Unrestricted Grants received for investigator initiated studies from MSD, Pfizer, AbbVie, UCB. Fees received for Lectures from BMS, AbbVie, Pfizer, MSD, UCB. MLH: Advisory Board AbbVie (No personal income, paid to institution). Prev. chaired the steering committee of the Danish Rheumatology Quality Registry (DANBIO, DRQ), which receives public funding from the hospital owners and funding from pharmaceutical companies. Speaker for Pfizer, Medac, Sandoz (no personal income, institution). Research grants (institution) from AbbVie, Biogen, BMS, Celltrion, Eli Lilly, Janssen Biologics B.V, Lundbeck Fonden, MSD, Medac, Pfizer, Roche, Samsung Biopies, Sandoz, Novartis, Nordforsk. LMO: Research grant from Novartis (paid to the employer)., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
22. Cut-Offs for Disease Activity States in Axial Spondyloarthritis With Ankylosing Spondylitis Disease Activity Score (ASDAS) Based on C-Reactive Protein and ASDAS Based on Erythrocyte Sedimentation Rate: Are They Interchangeable?
- Author
-
Georgiadis S, Ørnbjerg LM, Michelsen B, Kvien TK, Di Giuseppe D, Wallman JK, Závada J, Provan SA, Kristianslund EK, Rodrigues AM, Santos MJ, Rotar Ž, Pirkmajer KP, Nordström D, Macfarlane GJ, Jones GT, van der Horst-Bruinsma I, Hellamand P, Østergaard M, and Hetland ML
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Registries, C-Reactive Protein analysis, Blood Sedimentation, Spondylitis, Ankylosing blood, Spondylitis, Ankylosing diagnosis, Severity of Illness Index, Axial Spondyloarthritis blood, Axial Spondyloarthritis diagnosis
- Abstract
Objective: Ankylosing Spondylitis Disease Activity Score based on C-reactive protein (ASDAS-CRP) is recommended over ASDAS based on erythrocyte sedimentation rate (ASDAS-ESR) to assess disease activity in axial spondyloarthritis (axSpA). Although ASDAS-CRP and ASDAS-ESR are not interchangeable, the same disease activity cut-offs are used for both. We aimed to estimate optimal ASDAS-ESR values corresponding to the established ASDAS-CRP cut-offs (1.3, 2.1, and 3.5) and investigate the potential improvement of level of agreement between ASDAS-ESR and ASDAS-CRP disease activity states when applying these estimated cut-offs., Methods: We used data from patients with axSpA from 9 European registries initiating a tumor necrosis factor inhibitor. ASDAS-ESR cut-offs were estimated using the Youden index. The level of agreement between ASDAS-ESR and ASDAS-CRP disease activity states was compared against each other., Results: In 3664 patients, mean ASDAS-CRP was higher than ASDAS-ESR at both baseline (3.6 and 3.4, respectively) and aggregated follow-up at 6, 12, or 24 months (1.9 and 1.8, respectively). The estimated ASDAS-ESR values corresponding to the established ASDAS-CRP cut-offs were 1.4, 1.9, and 3.3. By applying these cut-offs, the proportion of discordance between disease activity states according to ASDAS-ESR and ASDAS-CRP decreased from 22.93% to 19.81% in baseline data but increased from 27.17% to 28.94% in follow-up data., Conclusion: We estimated the optimal ASDAS-ESR values corresponding to the established ASDAS-CRP cut-off values. However, applying the estimated cut-offs did not increase the level of agreement between ASDAS-ESR and ASDAS-CRP disease activity states to a relevant degree. Our findings did not provide evidence to reject the established cut-off values for ASDAS-ESR., (Copyright © 2024 by the Journal of Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
23. Second and third TNF inhibitors in European patients with axial spondyloarthritis: effectiveness and impact of the reason for switching.
- Author
-
Linde L, Ørnbjerg LM, Heegaard Brahe C, Wallman JK, Di Giuseppe D, Závada J, Castrejon I, Díaz-Gonzalez F, Rotar Z, Tomšič M, Glintborg B, Gudbjornsson B, Geirsson AJ, Michelsen B, Kristianslund EK, Santos MJ, Barcelos A, Nordström D, Eklund KK, Ciurea A, Nissen M, Akar S, Hejl Hyldstrup L, Krogh NS, Hetland ML, and Østergaard M
- Subjects
- Humans, Female, Male, Adult, Europe, Middle Aged, Treatment Outcome, Antirheumatic Agents therapeutic use, Tumor Necrosis Factor Inhibitors therapeutic use, Prospective Studies, Registries, Remission Induction, Tumor Necrosis Factor-alpha antagonists & inhibitors, Drug Substitution, Axial Spondyloarthritis drug therapy
- Abstract
Objective: To investigate real-world effectiveness of tumor necrosis factor inhibitors (TNFi) in patients with axial spondyloarthritis (axSpA) and the association with (i) treatment line (second and third TNFi-series) and (ii) reason for withdrawal from the preceding TNFi [lack of efficacy (LOE) vs adverse events (AE)]., Methods: Prospectively collected routine care data from 12 European registries were pooled. Rates for 12-month drug retention and 6-month remission [Ankylosing Spondylitis Disease Activity Score C-reactive protein inactive disease (ASDAS-ID)] were assessed in second and third TNFi-series and stratified by withdrawal reason., Results: We included 8254 s and 2939 third TNFi-series; 12-month drug retention rates were similar (71%). Six-month ASDAS-ID rates were higher for the second (23%) than third TNFi (16%). Twelve-month drug retention rates for patients withdrawing from the preceding TNFi due to AE vs LOE were similar for the second (68% and 67%) and third TNFi (both 68%), while for the second TNFi, rates were lower in primary than secondary non-responders (LOE <26 vs ≥26 weeks) (58% vs 71%, P < 0.001). Six-month ASDAS-ID rates for the second TNFi were higher if the withdrawal reason was AE (27%) vs LOE (17%), P < 0.001, while similar for the third TNFi (19% vs 13%, P = 0.20)., Conclusion: A similar proportion of axSpA patients remained on a second and third TNFi after one year, but with low remission rates for the third TNFi. Remission rates on the second TNFi (but not the third) were higher if the withdrawal reason from the preceding TNFi was AE vs LOE., (© The Author(s) 2023. Published by Oxford University Press on behalf of the British Society for Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
24. Mortality in patients with psoriatic arthritis in Sweden: a nationwide, population-based cohort study.
- Author
-
Exarchou S, Di Giuseppe D, Klingberg E, Sigurdardottir V, Wedrén S, Lindström U, Turesson C, Jacobsson LTH, Askling J, and Wallman JK
- Subjects
- Adult, Humans, Female, Cohort Studies, Sweden epidemiology, Comorbidity, Incidence, Arthritis, Psoriatic epidemiology, Cardiovascular Diseases epidemiology
- Abstract
Objectives: To compare all-cause mortality and causes of death between patients with psoriatic arthritis (PsA) and the general population in Sweden., Methods: Adults with at least one main PsA diagnosis (International Classification of Diseases-10: L40.5/M07.0-M07.3) from outpatient rheumatology/internal medicine departments 2001-2017 were identified from the National Patient Register. Each case was matched to five population comparator-subjects on sex/county/age at the case's first arthritis diagnosis. Follow-up ran from 1 January 2007, or from first PsA diagnosis thereafter, until death, emigration or 31 December 2018. Mortality was assessed overall, and stratified by sex and duration since diagnosis (diagnosis before/after 1 January 2007), using matched Cox proportional hazard regression (excluding/including adjustments for comorbidity) or Breslow test, as appropriate. Incidence rate ratios (IRR) of death, overall and stratified by sex/duration since diagnosis/age, as well as causes of death in PsA cases and comparator-subjects were also described., Results: All-cause mortality was elevated in PsA (HR: 1.11 (95% CI: 1.07 to 1.16); IRR: 1.18 (95% CI: 1.13 to 1.22)), mainly driven by increased risks in women (HR: 1.23 (95% CI: 1.16 to 1.30)) and cases with longer time since diagnosis (HR: 1.18 (95% CI: 1.12 to 1.25)). IRR of death were significantly increased for all ages except below 40 years, with the numerically highest point-estimates for ages 40-59 years. When adjusted for comorbidity, however, the elevated mortality risk in PsA disappeared. Causes of death were similar among PsA cases/comparator-subjects, with cardiovascular disease and malignancy as the leading causes., Conclusions: Mortality risk in PsA in Sweden was about 10% higher than in the general population, driven by excess comorbidity and with increased risks mainly in women and patients with longer disease duration., Competing Interests: Competing interests: SE: Grant/research support from AbbVie, Amgen, Eli Lilly, Novartis and Pfizer (as detailed under Funding statement); Consulting fees from Amgen, Janssen, Novartis and UCB Pharma. DDG: None declared. EK: Speaker’s bureau fees from Amgen, Janssen, Pfizer and UCB Pharma. VS: Speaker’s bureau fees from AbbVie, AstraZeneca and Novartis; Advisory board participation for AbbVie, Galapagos, Janssen, Novartis, Sanofi and UCB Pharma. SW: None declared. UL: None declared. CT: Grant/research support from BMS; Speaker’s bureau fees from AbbVie, BMS, Nordic Drugs, Pfizer and Roche; Advisory board participation for Roche. LTHJ: Speaker’s bureau fees from AbbVie, Janssen and Novartis; Advisory board participation for AbbVie, Eli Lilly, Janssen, Novartis and Pfizer. JA: Grant/research support for the Swedish biologics register ARTIS from AbbVie, BMS, Eli Lilly, Galapagos, Merck, Pfizer, Roche, Samsung Bioepis, Sanofi and UCB Pharma. JKW: Grant/research support from AbbVie, Amgen, Eli Lilly, Novartis and Pfizer (as detailed under Funding statement); Speaker′s bureau fees from AbbVie and Amgen., (© Author(s) (or their employer(s)) 2024. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
25. Predictors of DAPSA28 remission in patients with psoriatic arthritis initiating a first TNF inhibitor: results from 13 European registries.
- Author
-
Linde L, Ørnbjerg LM, Georgiadis S, H Rasmussen S, Lindström U, Askling J, Michelsen B, Di Giuseppe D, Wallman JK, Gudbjornsson B, Love TJ, Nordström DC, Yli-Kerttula T, Nekvindová L, Vencovský J, Iannone F, Cauli A, Loft AG, Glintborg B, Laas K, Rotar Z, Tomšič M, Macfarlane GJ, Möller B, van de Sande M, Codreanu C, Nissen MJ, Birlik M, Erten S, Santos MJ, Vieira-Sousa E, Hetland ML, and Østergaard M
- Subjects
- Male, Humans, Female, Tumor Necrosis Factor Inhibitors therapeutic use, Fatigue, Immunotherapy, Registries, Arthritis, Psoriatic drug therapy
- Abstract
Objectives: In bio-naïve patients with PsA initiating a TNF inhibitor (TNFi), we aimed to identify baseline predictors of Disease Activity index for PsA in 28 joints (DAPSA28) remission (primary objective) and DAPSA28 moderate response at 6 months, as well as drug retention at 12 months across 13 European registries., Methods: Baseline demographic and clinical characteristics were retrieved and the three outcomes investigated per registry and in pooled data, using logistic regression analyses on multiply imputed data. In the pooled cohort, selected predictors that were either consistently positive or negative across all three outcomes were defined as common predictors., Results: In the pooled cohort (n = 13 369), 6-month proportions of remission, moderate response and 12-month drug retention were 25%, 34% and 63% in patients with available data (n = 6954, n = 5275 and n = 13 369, respectively). Five common baseline predictors of remission, moderate response and 12-month drug retention were identified across all three outcomes. The odds ratios (95% CIs) for DAPSA28 remission were: age, per year: 0.97 (0.96-0.98); disease duration, years (<2 years as reference): 2-3 years: 1.20 (0.89-1.60), 4-9 years: 1.42 (1.09-1.84), ≥10 years: 1.66 (1.26-2.20); men vs women: 1.85 (1.54-2.23); CRP of >10 vs ≤10 mg/l: 1.52 (1.22-1.89) and 1 mm increase in patient fatigue score: 0.99 (0.98-0.99)., Conclusion: Baseline predictors of remission, response and adherence to TNFi therapy were identified, of which five were common for all three outcomes, indicating that the predictors emerging from our pooled cohort may be considered generalizable from country level to disease level., (© The Author(s) 2023. Published by Oxford University Press on behalf of the British Society for Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
26. Impact of Risk Factors on COVID-19 Outcomes in Unvaccinated People With Rheumatic Diseases: A Comparative Analysis of Pandemic Epochs Using the COVID-19 Global Rheumatology Alliance Registry.
- Author
-
Yazdany J, Ware A, Wallace ZS, Bhana S, Grainger R, Hachulla E, Richez C, Cacoub P, Hausmann JS, Liew JW, Sirotich E, Jacobsohn L, Strangfeld A, Mateus EF, Hyrich KL, Gossec L, Carmona L, Lawson-Tovey S, Kearsley-Fleet L, Schaefer M, Ribeiro SLE, Al-Emadi S, Hasseli R, Müller-Ladner U, Specker C, Schulze-Koops H, Bernardes M, Fraga VM, Rodrigues AM, Sparks JA, Ljung L, Di Giuseppe D, Tidblad L, Wise L, Duarte-García A, Ugarte-Gil MF, Colunga-Pedraza IJ, Martínez-Martínez MU, Alpizar-Rodriguez D, Xavier RM, Isnardi CA, Pera M, Pons-Estel G, Izadi Z, Gianfrancesco MA, Carrara G, Scirè CA, Zanetti A, and Machado PM
- Subjects
- Humans, Male, Pandemics, COVID-19 Vaccines therapeutic use, COVID-19 Testing, Risk Factors, Registries, Rheumatology, COVID-19 epidemiology, Rheumatic Diseases diagnosis, Rheumatic Diseases drug therapy, Rheumatic Diseases epidemiology
- Abstract
Objective: Approximately one third of individuals worldwide have not received a COVID-19 vaccine. Although studies have investigated risk factors linked to severe COVID-19 among unvaccinated people with rheumatic diseases (RDs), we know less about whether these factors changed as the pandemic progressed. We aimed to identify risk factors associated with severe COVID-19 in unvaccinated individuals in different pandemic epochs corresponding to major variants of concern., Methods: Patients with RDs and COVID-19 were entered into the COVID-19 Global Rheumatology Alliance Registry between March 2020 and June 2022. An ordinal logistic regression model (not hospitalized, hospitalized, and death) was used with date of COVID-19 diagnosis, age, sex, race and/or ethnicity, comorbidities, RD activity, medications, and the human development index (HDI) as covariates. The main analysis included all unvaccinated patients across COVID-19 pandemic epochs; subanalyses stratified patients according to RD types., Results: Among 19,256 unvaccinated people with RDs and COVID-19, those who were older, male, had more comorbidities, used glucocorticoids, had higher disease activity, or lived in lower HDI regions had worse outcomes across epochs. For those with rheumatoid arthritis, sulfasalazine and B-cell-depleting therapy were associated with worse outcomes, and tumor necrosis factor inhibitors were associated with improved outcomes. In those with connective tissue disease or vasculitis, B-cell-depleting therapy was associated with worse outcomes., Conclusion: Risk factors for severe COVID-19 outcomes were similar throughout pandemic epochs in unvaccinated people with RDs. Ongoing efforts, including vaccination, are needed to reduce COVID-19 severity in this population, particularly in those with medical and social vulnerabilities identified in this study., (© 2023 American College of Rheumatology.)
- Published
- 2024
- Full Text
- View/download PDF
27. Are JAKis more effective among elderly patients with RA, smokers and those with higher cardiovascular risk? A comparative effectiveness study of b/tsDMARDs in Sweden.
- Author
-
Bower H, Frisell T, di Giuseppe D, Delcoigne B, Lindström U, Turesson C, Chatzidionysiou K, Lindqvist E, Knight A, Forsblad-d'Elia H, and Askling J
- Subjects
- Humans, Aged, Sweden epidemiology, Smokers, Risk Factors, Tumor Necrosis Factor-alpha, Tumor Necrosis Factor Inhibitors, Heart Disease Risk Factors, Janus Kinase Inhibitors, Cardiovascular Diseases epidemiology, Cardiovascular Diseases etiology
- Abstract
Objectives: To investigate whether the relative effectiveness of janus kinase inhibitors (JAKis) versus tumour necrosis factor inhibitors (TNFi) or other biological disease-modifying antirheumatic drugs in rheumatoid arthritis differ by the presence or absence of risk factors for cardiovascular (CV) disease, age, sex and smoking., Methods: Through Swedish registers, we identified 13 493 individuals with 3166 JAKi, 5575 non-TNFi and 11 286 TNFi treatment initiations 2016-2022. All lines of therapy were included, with the majority in second line or higher. Treatment response was defined as the proportion reaching European Alliance of Associations for Rheumatology (EULAR) good response and Clinical Disease Activity Index (CDAI) remission, respectively, within 6 months. Crude percentage point differences in these proportions (JAKis, and non-TNFis, vs TNFis) overall and by risk factors were observed, and adjusted for confounders using linear regression models. Predicted probabilities of response and remission were estimated from adjusted Poisson models, and presented across CV risk and age., Results: Overall, adjusted percentage point differences indicated higher response (+5.0%, 95% CI 2.2% to 7.9%) and remission (+5.8%, 95% CI 3.2% to 8.5%) with JAKis versus TNFis. The adjusted percentage point differences for response in those above 65, at elevated CV risk, and smokers were +5.9% (95% CI 2.7% to 9.0%), +8.3% (95% CI 5.3% to 11.4%) and +6.0% (95% CI 3.3% to 8.7%), respectively. The corresponding estimates for remission were +8.0% (95% CI 5.3% to 10.8%), +5.6% (95% CI 3.0% to 8.2%) and +7.6% (95% CI 5.5% to 9.7%)., Conclusions: As used in clinical practice, response and remission at 6 months with JAKis are higher than with TNFi. Among patients with risk factors of concern, effectiveness is similar or numerically further increased. For individualised benefit-to-risk ratios to guide treatment choice, safety and effectiveness in specific patient segments should be considered., Competing Interests: Competing interests: Karolinska Institutet, with JA as principal investigator, has or has had research agreements with AbbVie, Astra-Zeneca, BMS, Eli Lilly, Galapagos, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi, and UCB, mainly in the context of safety monitoring of biologics via ARTIS/Swedish Biologics Register. CT has received speaker fees from AbbVie, BMS, Nordic Drugs, Pfizer and Roche., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
28. Comparative cardiovascular safety with janus kinase inhibitors and biological disease-modifying antirheumatic drugs as used in clinical practice: an observational cohort study from Sweden in patients with rheumatoid arthritis.
- Author
-
Bower H, Frisell T, di Giuseppe D, Delcoigne B, and Askling J
- Subjects
- Humans, Cohort Studies, Sweden epidemiology, Antirheumatic Agents adverse effects, Arthritis, Rheumatoid drug therapy, Arthritis, Rheumatoid epidemiology, Janus Kinase Inhibitors adverse effects
- Abstract
Objectives: To compare the incidence of cardiovascular (CV) events in rheumatoid arthritis (RA) treated with janus kinase inhibitors (JAKi), tumour necrosis factor inhibitors (TNFi), or other biological disease-modifying antirheumatic drugs (bDMARDs), in clinical practice, and to contextualise these findings by comparing to the Swedish RA population and general population at large., Methods: Patients with RA initiating JAKi, TNFi and non-TNFi bDMARDs were identified in the Swedish Rheumatology Quality Register between 2016 and 2021. Through linkages to national registers, a cohort of patients with RA, general population comparators, as well as covariates and incident major acute CV event (MACE, including myocardial infarction, stroke and fatal CV events) were identified until 2022. Crude and age-sex standardised rates were calculated and HRs estimated from multivariable Cox regression models using TNFi as reference., Results: We identified 13 492 patients with RA initiating a JAKi, non-TNFi bDMARD or TNFi treatment. Among 3037 JAKi-initiators, 59 MACE events were observed. The age-sex standardised rates for MACE were similar in the JAKi (0.88 per 100 person years) and TNFi (0.91) cohorts. Fully adjusted models showed no increased rate of MACE with JAKi (HR=0.71, 95% CI 0.51 to 0.99), or non-TNFi bDMARD (HR=0.98; 95% CI 0.78 to 1.23) in comparison to TNFi. We found no evidence that this HR changed over time since treatment initiation. In a CV-enriched subset, we observed higher rates but similar HRs., Conclusions: As used in present clinical practice in Sweden, we found no evidence that CV risk is higher with JAKis than TNFis in RA., Competing Interests: Competing interests: Karolinska Institutet, with JA as principal investigator, has or has had research agreements with AbbVie, Astra-Zeneca, BMS, Eli Lilly, Galapagos, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi, and UCB, mainly in the context of safety monitoring of biologics via ARTIS/Swedish Biologics Register., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
29. There is plenty of asbestos at the bottom. The case of magnesite raw material contaminated with asbestos fibres.
- Author
-
Gualtieri AF, Malferrari D, Di Giuseppe D, Scognamiglio V, Sala O, Gualtieri ML, Bersani D, Fornasini L, and Mugnaioli E
- Abstract
Although all six asbestos minerals (the layer silicate chrysotile and five chain silicate species actinolite asbestos, amosite, anthophyllite asbestos, crocidolite and tremolite asbestos) are classified as carcinogenic, chrysotile is still mined and used in many countries worldwide. Other countries, like Italy, impose zero tolerance for all asbestos species, but conflicting views repress the development of globally uniform treaties controlling international trade of asbestos-containing materials. Hence, countries with more severe legislations against the use of these hazardous materials lack of an international safety net against importation of non-compliant products. This research reports the first discovery of commercial magnesite raw materials contaminated with white asbestos (chrysotile). X-ray powder diffraction and thermogravimetric/thermodifferential measurements showed the presence of serpentine group minerals in both the semi-processed (powder) and quarried material. The univocal identification of chrysotile in the powders was confirmed by its peculiar Raman bands of the OH stretching vibrations between 3500 and 3800 cm
-1 , with an intense peak at ∼3695 cm-1 and a weak contribution at ∼3647 cm-1 . Transmission electron microscope showed that chrysotile forms fibres up to a few microns long and up to 80 nm thick with a nanotube structure characterized by inner channels as large as 30-40 nm. Fibres size analysis obtained by scanning electron microscopy indicates mean length and diameter of 5.95 and 0.109 μm with medians of 2.62 and 0.096 μm, respectively; some among the fibres analysed exhibit the so-called "Stanton size" (i.e., asbestos fibres longer than 8 μm and thinner than 0.25 μm that are strongly carcinogenic). Quantitative analysis showed a chrysotile content around 0.01 wt% not allowed by current regulations in Italy and many other countries. More generally, our findings demonstrate that without shared policies aimed at regulating asbestos circulation on the global market, "asbestos-free" national policies will inevitably fail., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. The authors wish to state that the commercial samples analysed in this study were provided by Italian suppliers and end-users and considered representative of batches present in the manufacture sites in Italy during the autumn period of the year 2021. Evidence of asbestos contamination from this work is intended only for the analysed samples and, in the absence of further experimental data, should be regarded as accidental. Under no circumstances should the results of this study be generalized in space and time and considered valid for other commercial magnesite samples., (Copyright © 2023 Elsevier B.V. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
30. Commonalities and differences in set-up and data collection across European spondyloarthritis registries - results from the EuroSpA collaboration.
- Author
-
Linde L, Ørnbjerg LM, Rasmussen SH, Love TJ, Loft AG, Závada J, Vencovský J, Laas K, Nordstrom D, Sokka-Isler T, Gudbjornsson B, Gröndal G, Iannone F, Ramonda R, Hellamand P, Kristianslund EK, Kvien TK, Rodrigues AM, Santos MJ, Codreanu C, Rotar Z, Tomšič M, Castrejon I, Díaz-Gonzáles F, Di Giuseppe D, Ljung L, Nissen MJ, Ciurea A, Macfarlane GJ, Heddle M, Glintborg B, Østergaard M, and Hetland ML
- Subjects
- Humans, Registries, Pain, Arthritis, Psoriatic drug therapy, Arthritis, Psoriatic epidemiology, Spondylarthritis drug therapy, Spondylarthritis epidemiology, Spondylitis, Ankylosing drug therapy
- Abstract
Background: In European axial spondyloarthritis (axSpA) and psoriatic arthritis (PsA) clinical registries, we aimed to investigate commonalities and differences in (1) set-up, clinical data collection; (2) data availability and completeness; and (3) wording, recall period, and scale used for selected patient-reported outcome measures (PROMs)., Methods: Data was obtained as part of the EuroSpA Research Collaboration Network and consisted of (1) an online survey and follow-up interview, (2) upload of real-world data, and (3) selected PROMs included in the online survey., Results: Fifteen registries participated, contributing 33,948 patients (axSpA: 21,330 (63%), PsA: 12,618 (37%)). The reported coverage of eligible patients ranged from 0.5 to 100%. Information on age, sex, biological/targeted synthetic disease-modifying anti-rheumatic drug treatment, disease duration, and C-reactive protein was available in all registries with data completeness between 85% and 100%. All PROMs (Bath Ankylosing Spondylitis Disease Activity and Functional Indices, Health Assessment Questionnaire, and patient global, pain and fatigue assessments) were more complete after 2015 (68-86%) compared to prior (50-79%). Patient global, pain and fatigue assessments showed heterogeneity between registries in terms of wording, recall periods, and scale., Conclusion: Important heterogeneity in registry design and data collection across fifteen European axSpA and PsA registries was observed. Several core measures were widely available, and an increase in data completeness of PROMs in recent years was identified. This study might serve as a basis for examining how differences in data collection across registries may impact the results of collaborative research in the future., (© 2023. BioMed Central Ltd., part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
31. Differences and similarities between the EULAR/ASAS-EULAR and national recommendations for treatment of patients with psoriatic arthritis and axial spondyloarthritis across Europe.
- Author
-
Michelsen B, Østergaard M, Nissen MJ, Ciurea A, Möller B, Ørnbjerg LM, Zavada J, Glintborg B, MacDonald A, Laas K, Nordström D, Gudbjornsson B, Iannone F, Hellmand P, Kvien TK, Rodrigues AM, Codreanu C, Rotar Z, Castrejón Fernández I, Wallman JK, Vencovsky J, Loft AG, Heddle M, Vorobjov S, Hokkanen AM, Gröndal G, Sebastiani M, van de Sande M, Kristianslund EK, Santos MJ, Mogosan C, Tomsic M, Díaz-González F, Di Giuseppe D, and Hetland ML
- Abstract
This is the first report comparing EULAR and national treatment recommendations for PsA patients across Europe, and the first this decade to compare ASAS-EULAR and national treatment recommendations in axSpA patients. An electronic survey was completed from October 2021-April 2022 by rheumatologists in 15 European countries. One and four countries followed all EULAR and ASAS-EULAR recommendations, respectively. Five countries had no national treatment recommendations for PsA and/or axSpA, but followed other regulations. In several countries, national treatment recommendations predated the most recent EULAR/ASAS-EULAR recommendations. Entry criteria for starting biologic/targeted synthetic disease-modifying anti-rheumatic drugs varied considerably. In several countries, for PsA patients with significant skin involvement, interleukin-17 inhibitors were not given preference. The positioning of Janus Kinase inhibitors differed and Phosphodiesterase-4 inhibitors were not in use/reimbursed in most countries. This study may motivate European countries to update their national treatment recommendations, to align them better with the latest international recommendations., Competing Interests: BM, research grant from Novartis (for the present manuscript, paid to employer), Centre for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY) is funded as a Centre for Clinical Treatment Research by The Research Council of Norway (project 328657); MØ, Funding for the present manuscript from Novartis (paid to institution), The EuroSpA Research Collaboration Network was financially supported by Novartis Pharma AG, research grants from Abbvie, BMS, Merck, Novartis, UCB, consulting fees from Abbvie, BMS, Celgene, Eli-Lilly, Galapagos, Gilead, Janssen, MEDAC, Merck, Novartis, Pfizer, Sandoz, UCB, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Abbvie, BMS, Celgene, Eli-Lilly, Galapagos, Gilead, Janssen, MEDAC, Merck, Novartis, Pfizer, Sandoz, UCB, support for attending meetings and/or travel from UCB; MJN, grant from Novartis (payment to institution), consulting fees from AbbVie, Eli-Lilly, Janssen, Novartis, Pfizer (payment to institution), payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from AbbVie, Eli-Lilly, Janssen, Novartis, Pfizer (payment to institution), support for attending meetings and/or travel from Janssen, UCB (payment to institution), participation on a Data Safety Monitoring Board or Advisory Board from Eli-Lilly, Janssen, Novartis, Pfizer (payment to institution), scientific member of the SCQM registry and the EuroSpA collaboration (unpaid), ASAS-EULAR taskforce member (unpaid); AC, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Abbvie, Merck-Sharp & Dohme, Novartis; BMö, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Abbvie, Janssen, Novartis, Pfizer, support for attending meetings and/or travel from Abbvie, Janssen, Novartis, Pfizer; LMØ, research grant from Novartis (for the present manuscript, paid to employer); JZ, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Abbvie, Eli-Lilly, Sandoz, Novartis, Egis, UCB, Sanofi, Astra Zeneca, support for attending meetings and/or travel from Abbvie, Pfizer, Astra Zeneca, participation on a data safety monitoring board or advisory board from Abbvie, UCB, Sobi; BGli, Research grants from Pfizer, AbbVie, BMS (payments made to institution); AM; payment for work done in relation to research questions for this project from the University of Aberdeen, speakers fees for educational events from Galapagos; KL, Honoraria for lectures from Pfizer, Abbvie, Novartis, Janssen, support for attending meetings from Abbvie, Pfizer; DN, Consulting fees from BMS, Lilly, MSD, Novartis, Pfizer, UCB, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Pfizer, support for attending meetings and/or travel from Pfizer; BGu, consulting fees from Novartis, speaker bureaus from Novartis, Nordic Pharma; FI, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Abbvie, BMS, Galapagos, Eli-Lilly, Pfizer, UCB, support for attending meetings and/or travel from Pfizer, UCB; PH, research grant (payments made to employer) from Novartis; TKK, consulting fees from AbbVie, Gilead, Janssen, Novartis, Pfizer, Sandoz, UCB, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Grünenthal, Janssen, Sandoz, participation on a data safety monitoring board or advisory board from Abbvie, grants or contracts from AbbVie, BMS, Galapagos, Novartis, Pfizer, UCB; AMR, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Abbvie, Amgen, Novartis, support for attending meetings and/or travel from Amgen, Nordic, Theramex, grants or contracts from Novartis, Pfizer, Amgen, Astra Zeneca, Abbvie, MSD, Lilly, Boehringer Ingelheim; CC, None; ZR, Consulting fees from AbbVie, Janssen, Novartis, MSD and Lek Sandoz, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Eli Lilly, Janssen, Abbvie, Pfizer, Boehringer Ingelheim, support for attending meetings and/or travel from Gedeon Richter, Pfizer, SOBI (payments made to employer), participation on data safety monitoring board or advisory board from Abbvie, Pfizer, Astra Zeneca, Janssen, Eli Lilly, Novartis and Boehringer Ingelheim; ICF, consulting fees from Pfizer, Galapagos, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from BMS, Eli-Lilly, Gilead, Janssen, Novartis, MSD, Pfizer, GSK, support for attending meetings and/or travel from Pfizer, Eli-Lilly; JKW, Research support from Abbvie, Amgen, Eli Lilly, Novartis, Pfizer (unrelated to the present work, payment made to Lund University), speakers bureaus from Abbvie and Amgen (payed to Skåne University Hospital), acting co-chair of the Swedish Society for Rheumatology's working group annually updating Swedish treatment recommendations for axial spondyloarthritis and psoriatic arthritis (unpaid, fiduciary assignment); JV, support for the present manuscript from the Czech Ministry of Health–Conceptual Development of Research Organization 00023728 (Institute of Rheumatology), payment to institution, consulting fees from Eli Lilly, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Abbvie, Biogen, Eli Lilly, Gilead, MSD, Novartis, Pfizer, Roche, Sanofi, UCB, support for attending meetings and/or travel from Abbvie (payment to institution), participation on a data safety monitoring board or advisory board from Abbvie, Gilead, Pfizer, UCB, grants or contracts from Abbvie (payment to institution); AGL, Consulting fees from Jansen-Cilag A/S, Lilly Nordics, UCB, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Novartis Healthcare A/S, Pfizer, support for attending meetings and/or travel from Pfizer, UCB, participation on a data safety monitoring board or advisory board from Jansen-Cilag A/S, Lilly Nordics, UCB; MH; None; SV, None; AMH, Research grant from MSD, grant for attending meeting from Janssen, Pfizer, support for travel cost from Janssen; GG, None; MS, support for the present manuscript from Lilly and Boehringer-Ingelheim, payment or honoraria for lectures, presentations, speaker's bureaus, manuscript writing or educational events from Pfizer, Boehringer-Ingelheim, GSK, participation on data safety or advisory board from Janssen-Cilag, support for attending meetings and/or travel from Janssen-Cilag; MvdS, Research support/grant from UCB, Eli Lilly, Novartis, Janssen (payments made to institution, AMR Medical Research BV), consulting fees from Novartis, UCB, Abbvie (payments made to institution), payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Novartis, UCB, Janssen (payments made to institution), support for attending meetings and/or travel from UCB; EKK, None; MJS, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Abbvie, Janssen, Lilly, Novartis, Pfizer, support for attending meetings and/or travel from Janssen, Viatris, Medac and receipt of equipment, materials, drugs, medical writing, gifts or other services from Vifor; CM, None; MT, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from AbbVie, Amgen, Boehringer, Pfizer, Sanofi-Aventis, Roche, Lek-Sandoz, Janssen, Medis, Novartis, participation on data safety or advisory board from Astra Zeneca, Eli Lilly, Janssen, Boehringer, MSD, Novartis, Roche; FDG, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Abbvie, payment for expert testimony from Abbvie, Galapagos, Janssen, and support for attending meetings and/or travel from Pfizer, Astra Zeneca; DDG, None; MLH, payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Pfizer, Medac, Sandoz, research grants from Abbvie, Biogen, BMS, Celltrion, Eli Lilly, Janssen Biologics B.V, Lundbeck Foundation, MSD, Pfizer, Roche, Samsung Biopies, Sandoz, Novartis, Nordstar (payment to institution). MLH has chaired the steering committee of the Danish Rheumatology Quality Registry (DANBIO, DRQ), which receives public funding from the hospital owners and funding from pharmaceutical companies. MLH co-chairs EuroSpA, which generates real-world evidence of treatment of psoriatic arthritis and axial spondyloarthritis based on secondary data and is partly funded by Novartis., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
32. Validity of clinical psoriatic arthritis diagnoses made by rheumatologists in the Swedish National Patient Register.
- Author
-
Wallman JK, Alenius GM, Klingberg E, Sigurdardottir V, Wedrén S, Exarchou S, Lindström U, Di Giuseppe D, Askling J, and Jacobsson L
- Subjects
- Humans, Sweden, Rheumatologists, Predictive Value of Tests, Rheumatoid Factor, Arthritis, Psoriatic diagnosis
- Abstract
Objectives: : Knowledge of the correspondence between clinical ICD diagnoses and classification criteria fulfilment is crucial to interpret studies identifying cases via ICD codes. We assessed the degree to which patients registered with ICD-10 diagnoses of psoriatic arthritis (PsA) in the Swedish National Patient Register (NPR) fulfil established PsA classification criteria., Method: Four hundred patients with at least one outpatient visit to one of five rheumatology or internal medicine departments (three university/two county departments across Sweden) in 2013-2015, with a main ICD-10 diagnosis of PsA (L40.5, M07.0-M07.3), were randomly selected (80 cases/site). Through a structured medical record review, positive predictive values (PPVs) for fulfilment of the following classification criteria were assessed: CASPAR, Moll and Wright, Vasey and Espinoza, and modified ESSG criteria for PsA. A subset analysis regarding CASPAR fulfilment was also performed among cases with available rheumatoid factor and peripheral X-ray status (central CASPAR items; n = 227)., Results: Of the 400 patients with a main ICD-10 diagnosis of PsA, 343 (86%) fulfilled at least one of the four PsA classification criteria. PPVs for the different criteria were: CASPAR 69% (82% in the subset analysis), Moll and Wright 51%, Vasey and Espinoza 76%, and modified ESSG 64%. Overall, only 6.5% of the 400 PsA diagnoses were judged as clearly incorrect by the medical record reviewers., Conclusion: The validity of rheumatologist-made, clinical ICD-10 diagnoses for PsA in the Swedish NPR is good, with PPVs of 69-82% for CASPAR fulfilment and 86% for meeting any established PsA classification criteria.
- Published
- 2023
- Full Text
- View/download PDF
33. The crystal structure of the killer fibre erionite from Tuzköy (Cappadocia, Turkey).
- Author
-
Giacobbe C, Moliterni A, Di Giuseppe D, Malferrari D, Wright JP, Mattioli M, Raneri S, Giannini C, Fornasini L, Mugnaioli E, Ballirano P, and Gualtieri AF
- Abstract
Erionite is a non-asbestos fibrous zeolite classified by the International Agency for Research on Cancer (IARC) as a Group 1 carcinogen and is considered today similar to or even more carcinogenic than the six regulated asbestos minerals. Exposure to fibrous erionite has been unequivocally linked to cases of malignant mesothelioma (MM) and this killer fibre is assumed to be directly responsible for more than 50% of all deaths in the population of the villages of Karain and Tuzköy in central Anatolia (Turkey). Erionite usually occurs in bundles of thin fibres and very rarely as single acicular or needle-like fibres. For this reason, a crystal structure of this fibre has not been attempted to date although an accurate characterization of its crystal structure is of paramount importance for our understanding of the toxicity and carcinogenicity. In this work, we report on a combined approach of microscopic (SEM, TEM, electron diffraction), spectroscopic (micro-Raman) and chemical techniques with synchrotron nano-single-crystal diffraction that allowed us to obtain the first reliable ab initio crystal structure of this killer zeolite. The refined structure showed regular T-O distances (in the range 1.61-1.65 Å) and extra-framework content in line with the chemical formula (K
2.63 Ca1 .57 Mg0.76 Na0.13 Ba0.01 )[Si28.62 Al7.35 ]O72 ·28.3H2 O. The synchrotron nano-diffraction data combined with three-dimensional electron diffraction (3DED) allowed us to unequivocally rule out the presence of offretite. These results are of paramount importance for understanding the mechanisms by which erionite induces toxic damage and for confirming the physical similarities with asbestos fibres., (open access.)- Published
- 2023
- Full Text
- View/download PDF
34. The National Prevalence of Clinically Diagnosed Psoriatic Arthritis in Sweden in 2017.
- Author
-
Exarchou S, Wallman JK, Di Giuseppe D, Klingberg E, Sigurdardottir V, Wedrén S, Lindström U, Turesson C, Jacobsson LTH, and Askling J
- Subjects
- Adult, Male, Humans, Sweden epidemiology, Prevalence, Arthritis, Psoriatic diagnosis, Arthritis, Psoriatic drug therapy, Arthritis, Psoriatic epidemiology, Antirheumatic Agents therapeutic use, Rheumatology
- Abstract
Objective: Psoriatic arthritis (PsA) prevalence estimates vary across studies; studies based on national data are few. We aimed to estimate the prevalence of clinically diagnosed PsA in Sweden in 2017, overall and stratified by sex, age, education, and geography, and to quantify disease-modifying antirheumatic drug (DMARD) use among those in contact with specialized rheumatology care between 2015 and 2017., Methods: Individuals who were 18 to 79 years of age, alive and residing in Sweden on December 31, 2017, and had a prior PsA diagnosis were identified from the National Patient Register (NPR) and/or the Swedish Rheumatology Quality Register (SRQ). PsA prevalence was estimated according to a base case (BC) definition (ie, ≥ 1 main PsA International Classification of Diseases code from rheumatology or internal medicine departments in the NPR or a PsA diagnosis in the SRQ), according to 4 sensitivity analysis definitions, and for those seen in specialized rheumatology care between 2015 and 2017. In the latter group, DMARD use during 2017 was also assessed. Data for stratifications were retrieved from national registers., Results: The crude national prevalence of PsA for adults, aged 18 to 79 years, was estimated at 0.39%, according to the BC definition; 0.34% after accounting for diagnostic misclassification; and 0.32% to 0.50% across all sensitivity analyses. The prevalence was lower in males and in those with a higher level of education. The prevalence for those seen in specialized rheumatology care between 2015 and 2017 was estimated at 0.24%. During 2017, 32% of patients in this population received biologic or targeted synthetic DMARDs, and 41% received conventional synthetic DMARDs only., Conclusion: The prevalence of clinically diagnosed PsA in adults, aged 18 to 79 years, in Sweden in 2017 was around 0.35%. Among PsA cases in recent contact with specialized rheumatology care, almost three-fourths received DMARD therapy in 2017., (Copyright © 2023 by the Journal of Rheumatology.)
- Published
- 2023
- Full Text
- View/download PDF
35. Uptake and effectiveness of newer biologic and targeted synthetic disease-modifying antirheumatic drugs in psoriatic arthritis: results from five Nordic biologics registries.
- Author
-
Glintborg B, Di Giuseppe D, Wallman JK, Nordström DC, Gudbjornsson B, Hetland ML, Askling J, Grondal G, Sokka T, Provan SA, Michelsen B, Kristianslund EK, Dreyer L, Love TJ, and Lindström U
- Subjects
- Humans, Adalimumab therapeutic use, Abatacept therapeutic use, Ustekinumab therapeutic use, Registries, Antirheumatic Agents therapeutic use, Arthritis, Psoriatic drug therapy, Biological Products therapeutic use
- Abstract
Background: We aimed to describe the uptake of newer biologic or targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs) in psoriatic arthritis (PsA) in the Nordic countries and to compare their retention and effectiveness., Methods: Patients with PsA starting a b/tsDMARD in 2012-2020 in five Nordic rheumatology registers were included. Uptake and patient characteristics were described, with comorbidities identified from linkages to national patient registries. One-year retention and 6-month effectiveness (proportions achieving low disease activity (LDA) on the Disease Activity Index for PSoriatic Arthritis based on 28-joint evaluation) for the newer b/tsDMARDs (abatacept/apremilast/ixekizumab/secukinumab/tofacitinib/ustekinumab) were compared with adalimumab through adjusted regression models stratified by treatment course (first, second/third, and fourth or more)., Results: In total, 5659 treatment courses with adalimumab (56% biologic-naïve) and 4767 courses with a newer b/tsDMARD (21% biologic-naïve) were included. The uptake of newer b/tsDMARDs increased from 2014 and plateaued in 2018. Patient characteristics appeared similar across treatments at treatment start. Adalimumab was more often used as the first course and newer b/tsDMARDs more often in biologic-experienced patients. Used as a second/third b/tsDMARD, the retention rate and the proportion achieving LDA were significantly better for adalimumab (rate 65%, proportion 59%) compared with abatacept (45%, 37%), apremilast (43%, 35%), ixekizumab (LDA only, 40%) and ustekinumab (LDA only, 40%), but not significantly different from other b/tsDMARDs., Conclusion: Uptake of newer b/tsDMARDs occurred mainly in biologic-experienced patients. Regardless of mode of action, only a minority of patients starting a second or later b/tsDMARD course remained on drug and achieved LDA. Superior outcomes for adalimumab indicate that the positioning of newer b/tsDMARDs in the PsA treatment algorithm remains to be established., Competing Interests: Competing interests: BG: Pfizer, BMS, Sandoz and chairs the steering committee of the Danish Rheumatology Registry (DANBIO), which receives public funding from hospital owners and funding from pharmaceutical companies. JKW: AbbVie, Amgen, Eli Lilly, Novartis and Pfizer. DCN: AbbVie, BMS, Lilly, MSD, Novartis, Pfizer, Roche and UCB. MLH: AbbVie, Biogen, BMS, Celltrion, Eli Lilly, Janssen Biologics BV, Lundbeckfonden, MSD, Pfizer, Roche, Samsung Bioepis, Sandoz and Novartis. JA: AbbVie, BMS, Eli Lilly, Galapagos, MSD, Pfizer, Roche, Samsung Bioepis and Sanofi. Agreements between Karolinska Institutet (with JA as PI) and the listed entities, mainly for the national safety monitoring of rheumatology immunomodulators in Sweden (ARTIS). TS: AbbVie, BMS, Celgene, Medac, Merck, Novartis, Orion Pharma, Pfizer, Roche, Sandoz, UCB and Boehringer Ingelheim. SAP: Boehringer Ingelheim. BM: Novartis. EKK: Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY) funded as a Centre for Clinical Treatment Research by the Research Council of Norway (project 328657). LD: BMS, Janssen, UCB and Boehringer Ingelheim., (© Author(s) (or their employer(s)) 2023. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
36. Does Risk of Hyperhomocysteinemia Depend on Thiol-Disulfide Exchange Reactions of Albumin and Homocysteine?
- Author
-
Coppo L, Scheggi S, DeMontis G, Priora R, Frosali S, Margaritis A, Summa D, Di Giuseppe D, Ulivelli M, and Di Simplicio P
- Subjects
- Humans, Disulfides, Homocystine, Homocysteine, Sulfhydryl Compounds, Hyperhomocysteinemia
- Abstract
Significance: Increased plasma concentrations of total homocysteine (tHcy; mild-moderate hyperhomocysteinemia: 15-50 μ M tHcy) are considered an independent risk factor for the onset/progression of various diseases, but it is not known about how the increase in tHcy causes pathological conditions. Recent Advances: Reduced homocysteine (HSH ∼1% of tHcy) is presumed to be toxic, unlike homocystine (∼9%) and mixed disulfide between homocysteine and albumin (HSS-ALB; homocysteine [Hcy]-albumin mixed disulfide, ∼90%). This and other notions make it difficult to explain the pathogenicity of Hcy because: (i) lowering tHcy does not improve pathological outcomes; (ii) damage due to HSH usually emerges at supraphysiological doses; and (iii) it is not known why tiny increments in plasma concentrations of HSH can be pathological. Critical Issues: Albumin may have a role in Hcy toxicity, because HSS-ALB could release toxic HSH via thiol-disulfide (SH/SS) exchange reactions in cells. Similarly, thiol-disulfide exchange processes of reduced albumin (albumin with free SH group of Cys34 [HS-ALB]) or N -homocysteinylated albumin are plausible alternatives for initiating Hcy pathological events. Adverse effects of albumin and other data reviewed here suggest the hypothesis of a role of albumin in Hcy toxicity. Future Directions : HSS-ALB might be involved in disruption of the antioxidant/oxidant balance in critical tissues (brain, liver, kidney). Since homocysteine-albumin mixed disulfide is a possible intermediate of thiol-disulfide exchange reactions, we suggest that homocysteinylated albumin could be a new pathological factor, and that studies on the redox role of albumin and mixed disulfide production via thiol-disulfide exchange reactions could offer new therapeutic insights for reducing Hcy toxicity.
- Published
- 2023
- Full Text
- View/download PDF
37. Methotrexate treatment in early psoriatic arthritis in comparison to rheumatoid arthritis: an observational nationwide study.
- Author
-
Lindström U, di Giuseppe D, Exarchou S, Alenius GM, Olofsson T, Klingberg E, Jacobsson L, Askling J, and Wallman JK
- Subjects
- Humans, Methotrexate therapeutic use, Pain drug therapy, Arthritis, Psoriatic diagnosis, Arthritis, Psoriatic drug therapy, Arthritis, Psoriatic epidemiology, Arthritis, Rheumatoid diagnosis, Arthritis, Rheumatoid drug therapy, Arthritis, Rheumatoid epidemiology, Antirheumatic Agents adverse effects
- Abstract
Introduction: We aimed to compare the proportions of patients with newly diagnosed psoriatic arthritis (PsA) and rheumatoid arthritis (RA) remaining on methotrexate (regardless of other disease-modifying antirheumatic drug (DMARD)-changes), and proportions not having started another DMARD (regardless of methotrexate discontinuation), within 2 years of starting methotrexate, as well as methotrexate effectiveness., Methods: Patients with DMARD-naïve, newly diagnosed PsA, starting methotrexate 2011-2019, were identified from high-quality national Swedish registers and matched 1:1 to comparable patients with RA. Proportions remaining on methotrexate and not starting another DMARD were calculated. For patients with disease activity data at baseline and 6 months, response to methotrexate monotherapy was compared through logistic regression, applying non-responder imputation., Results: In total, 3642/3642 patients with PsA/RA were included. Baseline patient-reported pain and global health were similar, whereas patients with RA had higher 28-joint scores and evaluator-assessed disease activity. Two years after methotrexate start, 71% of PsA vs 76% of patients with RA remained on methotrexate, 66% vs 60% had not started any other DMARD, and 77% vs 74% had not started specifically a biological or targeted synthetic DMARD. At 6 months, the proportions of patients with PsA versus RA achieving pain-scores ≤15 mm were 26% vs 36%; global health ≤20 mm: 32% vs 42%; evaluator-assessed 'remission': 20% vs 27%, with corresponding adjusted ORs (PsA vs RA) of 0.63 (95% CI 0.47 to 0.85); 0.57 (95% CI 0.42 to 0.76) and 0.54 (95% CI 0.39 to 0.75)., Discussion: In Swedish clinical practice, methotrexate use is similar in PsA and RA, both regarding initiation of other DMARDs and methotrexate retention. On a group level, disease activity improved during methotrexate monotherapy in both diseases, although more so in RA., Competing Interests: Competing interests: SE: Consultant of AbbVie, Amgen, Janssen, Novartis, UCB Pharma. TO: consulting tasks for Eli Lilly, and Merck Sharp & Dohme unrelated to the present work. EK: lecture fees from Pfizer, Eli-Lily, UCB Pharma and Janssen. LJ: lecture and consulting fees from Pfizer, Novartis, Eli-Lily and Janssen. JA: Agreements between Karolinska Institutet (with JA as PI) and Abbvie, BMS, Eli Lilly, Galapagos, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi, mainly for the national safety monitoring of rheumatology immunomodulators in Sweden (ARTIS). JKW: Speakers bureau fees from AbbVie, Amgen. Research support from AbbVie, Amgen, Eli Lilly, Novartis, Pfizer. The remaining authors have no competing interests., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
38. Safety of biological and targeted synthetic disease-modifying antirheumatic drugs for rheumatoid arthritis as used in clinical practice: results from the ARTIS programme.
- Author
-
Frisell T, Bower H, Morin M, Baecklund E, Di Giuseppe D, Delcoigne B, Feltelius N, Forsblad-d'Elia H, Lindqvist E, Lindström U, and Askling J
- Subjects
- Humans, Sweden epidemiology, Cohort Studies, Antirheumatic Agents adverse effects, Biological Products therapeutic use, Arthritis, Rheumatoid drug therapy, Arthritis, Rheumatoid chemically induced, Janus Kinase Inhibitors adverse effects
- Abstract
Objective: Longitudinal clinical registry-infrastructures such as Anti-Rheumatic Therapies in Sweden (ARTIS) allow simultaneous comparison of the safety of individual immunomodulatory drugs used in clinical practice, with consistent definitions of treatment cohorts, follow-up and outcomes. Our objective was to assess and compare incidence rates of key safety outcomes for individual targeted synthetic or biological disease-modifying antirheumatic drugs (b/ts DMARDs) in rheumatoid arthritis (RA), updating previous reports and including newer treatments including Janus Kinase inhibitors (JAKi)., Methods: Nationwide register-based cohort study including all patients with RA in Sweden registered as starting any b/tsDMARD 1 January 2010 through 31 December 2020, followed until 30 June 2021 (N=20 117). The incidence rates of selected outcomes, identified through national healthcare registers, were compared between individual b/tsDMARDs, adjusted for confounding by demographics, RA disease characteristics and comorbidity., Results: There were marked differences in treatment discontinuations due to adverse events (rates per 1000 person-years ranged from 18 on rituximab to 57 on tofacitinib), but few significant differences were observed for the serious adverse events under study. Neither cardiovascular events nor general serious infections were more frequent on baricitinib or tofacitinib versus bDMARDs, but JAKi were associated with higher rates of hospital-treated herpes zoster (HR vs etanercept, 3.82 (95% CI 2.05 to 7.09) and 4.00 (1.59 to 10.06)). Low number of events limited some comparisons, in particular for sarilumab and tofacitinib., Conclusion: Data from ARTIS supports that the b/tsDMARDs currently used to treat RA have acceptable and largely similar safety profiles, but differences exist in particular concerning tolerability and specific infection risks., Competing Interests: Competing interests: The ARTIS project is supported by agreements between Karolinska Institutet (with JA as PI) and the listed entities, mainly for the national safety monitoring of rheumatology immunomodulators in Sweden (ARTIS): Abbvie, BMS, Eli Lilly, Galapagos, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi. TF, HB, DDG and BD are partly employed by the ARTIS project. EL, MM, NF, UL and HFdE report no conflict of interest., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
39. Venous thromboembolism with JAK inhibitors and other immune-modulatory drugs: a Swedish comparative safety study among patients with rheumatoid arthritis.
- Author
-
Molander V, Bower H, Frisell T, Delcoigne B, Di Giuseppe D, and Askling J
- Subjects
- Humans, Sweden epidemiology, Cohort Studies, Tumor Necrosis Factor Inhibitors therapeutic use, Venous Thromboembolism epidemiology, Janus Kinase Inhibitors therapeutic use, Arthritis, Rheumatoid drug therapy, Antirheumatic Agents therapeutic use
- Abstract
Objective: To assess and compare the incidence of venous thromboembolism (VTE) in patients with rheumatoid arthritis (RA) treated with Janus kinase inhibitors (JAKi), tumour necrosis factor inhibitors (TNFi) or other biological disease modifying antirheumatic drugs (bDMARDs). For contextualisation, to assess VTE incidences in the Swedish general population and in the RA source population., Methods: We performed a nationwide register-based, active comparator, new user design cohort study in Sweden from 2010 to 2021. The Swedish Rheumatology Quality Register was linked to national health registers to identify treatment cohorts (exposure) of initiators of a JAKi, a TNFi, or a non-TNFi bDMARD (n=32 737 treatment initiations). We also identified a general population cohort (matched 1:5, n=92 108), and an 'overall RA' comparator cohort (n=85 722). Outcome was time to first VTE during the follow-up, overall and by deep vein thrombosis (DVT) and pulmonary embolism (PE). We calculated incidence rates (IR) and multivariable-adjusted HRs using Cox regression., Results: Based on 559 incident VTE events, the age- and sex-standardised (to TNFi) IR (95% CI) for VTE was 5.15 per 1000 person-years (4.58 to 5.78) for patients treated with TNFi, 11.33 (8.54 to 15.04) for patients treated with JAKi, 5.86 (5.69 to 6.04) in the overall RA cohort and 3.28 (3.14 to 3.43) in the general population. The fully adjusted HR (95% CI) for VTE with JAKi versus TNFi was 1.73 (1.24 to 2.42), the corresponding HR for PE was 3.21 (2.11 to 4.88) and 0.83 (0.47 to 1.45) for DVT., Conclusions: Patients with RA treated with JAKi in clinical practice are at increased risk of VTE compared with those treated with bDMARDs, an increase numerically confined to PE., Competing Interests: Competing interests: Karolinska Institutet, with JA as principal investigator, has or has had research agreements with Abbvie, Astra-Zeneca, BMS, Eli Lilly, Galapagos, MSD, Pfizer, Roche, Samsung Bioepis, Sanofi, and UCB, mainly in the context of safety monitoring of biologics via ARTIS/Swedish Biologics Register., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
40. Is the risk of infection higher during treatment with secukinumab than with TNF inhibitors? An observational study from the Nordic countries.
- Author
-
Glintborg B, Di Giuseppe D, Wallman JK, Provan SA, Nordström D, Hokkanen AM, Österlund J, Kristianslund E, Kvien TK, Gudbjornsson B, Hetland ML, Michelsen B, Jacobsson L, Askling J, and Lindström U
- Subjects
- Humans, Adalimumab adverse effects, Tumor Necrosis Factor Inhibitors adverse effects, Scandinavian and Nordic Countries epidemiology, Treatment Outcome, Antirheumatic Agents adverse effects, Arthritis, Psoriatic drug therapy
- Abstract
Objectives: The positioning of secukinumab in the treatment of axial SpA (axSpA) and PsA is debated, partly due to a limited understanding of the comparative safety of the available treatments. We aimed to assess the risk of the key safety outcome infections during treatment with secukinumab and TNF inhibitors (TNFi)., Methods: Patients with SpA and PsA starting secukinumab or TNFi year 2015 through 2018 were identified in four Nordic rheumatology registers. The first hospitalized infection during the first year of treatment was identified through linkage to national registers. Incidence rates (IRs) with 95% CIs per 100 patient-years were calculated. Adjusted hazard ratios were estimated through Cox regression, with secukinumab as the reference. Several sensitivity analyses were performed to investigate confounding by indication., Results: Among 7708 patients with SpA and 5760 patients with PsA, we identified 16 229 treatment courses of TNFi (53% bionaïve) and 1948 with secukinumab (11% bionaïve). For secukinumab, the first-year risk of hospitalized infection was 3.5% (IR 5.0; 3.9-6.3), compared with 1.7% (IR 2.3; 1.7-3.0) during 3201 courses with adalimumab, with the IRs for other TNFi lying in between these values. The adjusted HR for adalimumab, compared with secukinumab, was 0.58 (0.39-0.85). In sensitivity analyses, the difference from secukinumab was somewhat attenuated and in some analyses no longer statistically significant., Conclusion: When used according to clinical practice in the Nordic countries, the observed first-year absolute risk of hospitalized infection was doubled for secukinumab compared with adalimumab. This excess risk seemed largely explained by confounding by indication., (© The Author(s) 2022. Published by Oxford University Press on behalf of the British Society for Rheumatology.)
- Published
- 2023
- Full Text
- View/download PDF
41. The impact of a csDMARD in combination with a TNF inhibitor on drug retention and clinical remission in axial spondyloarthritis.
- Author
-
Nissen M, Delcoigne B, Di Giuseppe D, Jacobsson L, Hetland ML, Ciurea A, Nekvindova L, Iannone F, Akkoc N, Sokka-Isler T, Fagerli KM, Santos MJ, Codreanu C, Pombo-Suarez M, Rotar Z, Gudbjornsson B, van der Horst-Bruinsma I, Loft AG, Möller B, Mann H, Conti F, Yildirim Cetin G, Relas H, Michelsen B, Avila Ribeiro P, Ionescu R, Sanchez-Piedra C, Tomsic M, Geirsson ÁJ, Askling J, Glintborg B, and Lindström U
- Subjects
- Humans, Tumor Necrosis Factor Inhibitors therapeutic use, Tumor Necrosis Factor-alpha, Treatment Outcome, Antirheumatic Agents therapeutic use, Spondylarthritis drug therapy, Axial Spondyloarthritis
- Abstract
Objectives: Many axial spondylarthritis (axSpA) patients receive a conventional synthetic DMARD (csDMARD) in combination with a TNF inhibitor (TNFi). However, the value of this co-therapy remains unclear. The objectives were to describe the characteristics of axSpA patients initiating a first TNFi as monotherapy compared with co-therapy with csDMARD, to compare one-year TNFi retention and remission rates, and to explore the impact of peripheral arthritis., Methods: Data was collected from 13 European registries. One-year outcomes included TNFi retention and hazard ratios (HR) for discontinuation with 95% CIs. Logistic regression was performed with adjusted odds ratios (OR) of achieving remission (Ankylosing Spondylitis Disease Activity Score (ASDAS)-CRP < 1.3 and/or BASDAI < 2) and stratified by treatment. Inter-registry heterogeneity was assessed using random-effect meta-analyses, combined results were presented when heterogeneity was not significant. Peripheral arthritis was defined as ≥1 swollen joint at baseline (=TNFi start)., Results: Amongst 24 171 axSpA patients, 32% received csDMARD co-therapy (range across countries: 13.5% to 71.2%). The co-therapy group had more baseline peripheral arthritis and higher CRP than the monotherapy group. One-year TNFi-retention rates (95% CI): 79% (78, 79%) for TNFi monotherapy vs 82% (81, 83%) with co-therapy (P < 0.001). Remission was obtained in 20% on monotherapy and 22% on co-therapy (P < 0.001); adjusted OR of 1.16 (1.07, 1.25). Remission rates at 12 months were similar in patients with/without peripheral arthritis., Conclusion: This large European study of axial SpA patients showed similar one-year treatment outcomes for TNFi monotherapy and csDMARD co-therapy, although considerable heterogeneity across countries limited the identification of certain subgroups (e.g. peripheral arthritis) that may benefit from co-therapy., (© The Author(s) 2022. Published by Oxford University Press on behalf of the British Society for Rheumatology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
42. Correction to: Do patient-reported measures of disease activity in rheumatoid arthritis vary between countries? Results from a Nordic collaboration.
- Author
-
Delcoigne B, Provan SA, Hammer HB, Di Giuseppe D, Frisell T, Glintborg B, Grondal G, Gudbjornsson B, Hetland ML, Michelsen B, Nordström D, Relas H, and Askling J
- Published
- 2022
- Full Text
- View/download PDF
43. Do patient-reported measures of disease activity in rheumatoid arthritis vary between countries? Results from a Nordic collaboration.
- Author
-
Delcoigne B, Provan SA, Hammer HB, Di Giuseppe D, Frisell T, Glintborg B, Grondal G, Gudbjornsson B, Hetland ML, Michelsen B, Nordström D, Relas H, and Askling J
- Subjects
- Humans, Pain drug therapy, Patient Reported Outcome Measures, Severity of Illness Index, Antirheumatic Agents therapeutic use, Arthritis, Rheumatoid drug therapy
- Abstract
Objectives: To investigate whether patient-reported outcomes vary across countries and are influenced by cultural/contextual factors. Specifically, we aimed to assess inter-country differences in tender joint count (TJC), pain and patient's global health assessment (PGA), and their impact on disease activity (DAS28-CRP) in RA patients from five Nordic countries., Methods: We collected data (baseline, 3- and 12-months) from rheumatology registers in the five countries comprising RA patients starting a first ever MTX or a first ever TNF inhibitor (TNFi). In order to assess the role of context (=country), we separately modelled TJC, pain and PGA as functions of objective variables (CRP, swollen joint count, age, sex, calendar period and disease duration) with linear models. Analyses were performed at each time point and for both treatments. We further assessed the impact of inter-country differences on DAS28-CRP., Results: A total of 27 645 RA patients started MTX and 19 733 started a TNFi. Crude inter-country differences at MTX start amounted to up to 4 points (28 points scale) for TJC, 10 and 27 points (0-100 scale) for pain and PGA, respectively. Corresponding numbers at TNFi start were 3 (TJC), 27 (pain) and 24 (PGA) points. All differences were reduced at 3- and 12-months, and attenuated when adjusting for the objective variables. The variation in predicted DAS28-CRP across countries amounted to <0.5 units., Conclusions: Inter-country differences in TJC, pain and PGA are greater than expected based on differences in objective measures, but have a small clinical impact on DAS28-CRP across countries., (© The Author(s) 2022. Published by Oxford University Press on behalf of the British Society for Rheumatology.)
- Published
- 2022
- Full Text
- View/download PDF
44. Risk of lung cancer in rheumatoid arthritis and in relation to autoantibody positivity and smoking.
- Author
-
Chatzidionysiou K, di Giuseppe D, Soderling J, Catrina A, and Askling J
- Subjects
- Humans, Cohort Studies, Early Detection of Cancer adverse effects, Smoking adverse effects, Smoking epidemiology, Autoantibodies, Lung Neoplasms etiology, Lung Neoplasms complications, Arthritis, Rheumatoid complications, Arthritis, Rheumatoid epidemiology, Arthritis, Rheumatoid diagnosis
- Abstract
Objective: Lung cancer is a common malignancy in rheumatoid arthritis (RA). Since smoking is a risk factor for both (seropositive) RA and lung cancer, it remains unclear whether RA, in itself, increases lung cancer risk., Methods: We performed a population-based cohort study of patients with RA and individually matched general population reference individuals identified in Swedish registers and from the Epidemiological Investigation of RA early RA study, prospectively followed for lung cancer occurrence 1995-2018. We calculated incidence rates and performed Cox regression to estimate HRs including 95% CIs of lung cancer, taking smoking and RA serostatus into account., Results: Overall, we included 44 101 patients with RA (590 incident lung cancers, 56 per 100 000), and 216 495 matched general population individuals (1691 incident lung cancers, 33 per 100 000), corresponding to a crude HR (95% CI) of 1.76 (1.60 to 1.93). In subset analyses, this increased risk remained after adjustment for smoking (HR 1.77, 95% CI 1.06 to 2.97). Compared with general population subjects who were never smokers, patients with RA who were ever smokers had almost seven times higher risk of lung cancer. In RA, seropositivity was a significant lung cancer risk factor, even when adjusted for smoking, increasing the incidence 2-6 times. At 20 years, the risk in patients with RA was almost 3%, overall and over 4% for patients who were ever smokers and had at least one RA autoantibody., Conclusions: Seropositive RA is a risk factor for lung cancer over and above what can be explained by smoking, although residual confounding by smoking or other airway exposures cannot be formally excluded. There is a need for increased awareness and potentially for regular lung cancer screening, at least in a subset of patients with RA., Competing Interests: Competing interests: KC: consultancy fees from Eli Lilly, AbbVie and Pfizer. JS: none. JA: Karolinska Institutet has entered into agreements between Karolinska Institutet (JA as principal investigator) with AbbVie, BMS, MSD, Eli Lilly, Galapagos, Pfizer, Roche, Samsung Bioepis, Sanofi and UCB, mainly regarding safety monitoring of anti-rheumatic therapies. DdG: none., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
45. Comparison of treatment retention of originator vs biosimilar products in clinical rheumatology practice in Sweden.
- Author
-
Di Giuseppe D, Lindstrom U, Bower H, Delcoigne B, Frisell T, Chatzidionysiou K, Sjöwall C, Lindqvist E, and Askling J
- Subjects
- Adalimumab therapeutic use, Etanercept, Humans, Infliximab, Rituximab therapeutic use, Sweden, Treatment Outcome, Biosimilar Pharmaceuticals adverse effects, Rheumatology
- Abstract
Objectives: To compare treatment retention between biosimilars and their originator products among first starters (etanercept, infliximab, adalimumab and rituximab), as well as after non-medical switch., Methods: Patients with rheumatic diseases starting, for the first time, an originator or biosimilar etanercept, infliximab, adalimumab or rituximab were identified in the national Swedish Rheumatology Quality Register. Moreover, patients switching from an originator to its biosimilar were identified and individually matched to patients continuing on the originator. One-year treatment retention was calculated and hazard ratios (HR) for discontinuation with 95% CIs were estimated, adjusting for comorbidities and socio-economic factors., Results: In total, 21 443 first treatment courses were identified. The proportion of patients still on the drug at 1 year and the HR for discontinuation revealed no differences across adalimumab (Humira, Imraldi, Amgevita and Hyrimoz) nor across rituximab products (Mabthera, Ritemvia/Truxima and Rixathon). The proportions on the drug at 1 year were similar for Benepali (77%) and Enbrel (75%) and the adjusted HR for Benepali compared with Enbrel was 0.91 (95% CI 0.83, 0.99). For infliximab, the proportion still on the drug at 1 year was 67% for Remicade and 66% for Remsima/Inflectra and the HR compared with Remicade was 1.16 (95% CI 1.02, 1.33). Among 2925 patients switching from an originator drug to one of its biosimilars, we noted no statistically significant or clinically relevant differences in drug survival compared with those who remained on originator therapy., Conclusion: This large observational study supports the equivalence of biologic DMARD biosimilar products and originators when used in routine rheumatology care., (© The Author(s) 2021. Published by Oxford University Press on behalf of the British Society for Rheumatology.)
- Published
- 2022
- Full Text
- View/download PDF
46. European bio-naïve spondyloarthritis patients initiating TNF inhibitor: time trends in baseline characteristics, treatment retention and response.
- Author
-
Christiansen SN, Ørnbjerg LM, Rasmussen SH, Loft AG, Askling J, Iannone F, Zavada J, Michelsen B, Nissen M, Onen F, Santos MJ, Pombo-Suarez M, Relas H, Macfarlane GJ, Tomsic M, Codreanu C, Gudbjornsson B, Van der Horst-Bruinsma I, Di Giuseppe D, Glintborg B, Gremese E, Pavelka K, Kristianslund EK, Ciurea A, Akkoc N, Barcelos A, Sánchez-Piedra C, Peltomaa R, Jones GT, Rotar Z, Ionescu R, Grondal G, Van de Sande MGH, Laas K, Østergaard M, and Hetland ML
- Subjects
- Cohort Studies, Humans, Male, Treatment Outcome, Tumor Necrosis Factor Inhibitors therapeutic use, Arthritis, Psoriatic drug therapy, Spondylarthritis drug therapy
- Abstract
Objectives: To investigate time trends in baseline characteristics and retention, remission and response rates in bio-naïve axial spondyloarthritis (axSpA) and psoriatic arthritis (PsA) patients initiating TNF inhibitor (TNFi) treatment., Methods: Prospectively collected data on bio-naïve axSpA and PsA patients from routine care in 15 European countries were pooled. Three cohorts were defined according to year of TNFi initiation: A (1999-2008), B (2009-2014) and C (2015-2018). Retention, remission and response rates were assessed at 6, 12 and 24 months., Results: In total, 27 149 axSpA and 17 446 PsA patients were included. Cohort A patients had longer disease duration compared with B and C. In axSpA, cohort A had the largest proportion of male and HLA-B27 positive patients. In PsA, baseline disease activity was highest in cohort A. Retention rates in axSpA/PsA were highest in cohort A and differed only slightly between B and C. For all cohorts, disease activity decreased markedly from 0 to 6 months. In axSpA, disease activity at 24 months was highest in cohort A, where also remission and response rates were lowest. In PsA, remission rates at 6 and 12 months tended to be lowest in cohort A. Response rates were at all time points comparable across cohorts, and less between-cohort disease activity differences were seen at 24 months., Conclusion: Our findings indicate that over the past decades, clinicians have implemented more aggressive treatment strategies in spondyloarthritis. This was illustrated by shorter disease duration at treatment initiation, decreased retention rates and higher remission rates during recent years., (© The Author(s) 2021. Published by Oxford University Press on behalf of the British Society for Rheumatology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
47. The occurrence of multiple treatment switches in axial spondyloarthritis. Results from five Nordic rheumatology registries.
- Author
-
Di Giuseppe D, Lindström U, Aaltonen K, Relas H, Provan S, Gudbjornsson B, Hetland ML, Askling J, Kauppi M, Geirsson AJ, Chatzidionysiou K, Jørgensen TS, Dreyer L, Michelsen B, Jacobsson L, and Glintborg B
- Subjects
- Adult, Female, Humans, Male, Registries, Axial Spondyloarthritis, Biological Products therapeutic use, Rheumatology, Spondylarthritis drug therapy, Spondylarthritis epidemiology
- Abstract
Objectives: In axial spondyloarthritis (axSpA), switching between multiple biologic or targeted synthetic (b/ts-) DMARDs might indicate difficult-to-treat disease. We aimed to explore the occurrence of multiple switching in routine care axSpA patients using various definitions, and to identify associated clinical characteristics upon start of first b/tsDMARD (baseline)., Methods: Observational cohort study including patients with axSpA starting a first-ever b/tsDMARD 2009-2018 based on data from five biologic registries (Denmark/Sweden/Finland/Norway/Iceland). Comorbidities and extra-articular manifestations were identified through linkage to national registries. Multi-switching was defined in overlapping categories according to b/tsDMARD treatment history: treatment with ≥3, ≥4 or ≥5 b/tsDMARDs during follow-up. We explored the cumulative incidence of patients becoming multi-switchers with ≥3 b/tsDMARDs stratified by calendar-period (2009-2011, 2012-2013, 2014-2015, 2016-2018). In the subgroup of patients starting a first b/tsDMARD 2009-2015, baseline characteristics associated with multi-switching (within 3 years' follow-up) were explored using multiple logistic regression analyses., Results: Among 8398 patients included, 6056 patients (63% male, median age 42 years) started a first b/tsDMARD in 2009-2015, whereof proportions treated with ≥3, ≥4 or ≥5 b/tsDMARDs within 3 years' follow-up were 8%, 3% and 1%, respectively. Calendar-period did not affect the cumulative incidence of multi-switching. Baseline characteristics associated with multi-switching (≥3 b/tsDMARDs) were female gender, shorter disease duration, higher patient global score, comorbidities and having psoriasis but not uveitis., Conclusion: In this large Nordic observational cohort of axSpA patients, multiple switching was frequent with no apparent time-trend. Clinical associated factors included gender, but also previous comorbidities and extra-articular manifestations illustrating the ongoing challenge of treating this patient group., (© The Author(s) 2021. Published by Oxford University Press on behalf of the British Society for Rheumatology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
48. Neither low social support nor low decision latitude at work is associated with disease remission among patients with rheumatoid arthritis: results from the Swedish EIRA study.
- Author
-
Hedenstierna L, Hedström AK, Klareskog L, Di Giuseppe D, Alfredsson L, Askling J, Ernestam S, Saevarsdottir S, and Ljung L
- Subjects
- C-Reactive Protein, Cohort Studies, Humans, Remission Induction, Severity of Illness Index, Social Support, Sweden epidemiology, Antirheumatic Agents therapeutic use, Arthritis, Rheumatoid drug therapy, Arthritis, Rheumatoid epidemiology
- Abstract
Objectives: To investigate the association between psychosocial vulnerability, defined as either low social support or low decision latitude at work, and disease remission at 3, 12, and 60 months in patients with rheumatoid arthritis (RA)., Methods: This cohort study included all patients enrolled in both the Swedish Epidemiological Investigation of Rheumatoid Arthritis (EIRA) 1996-2015 and the Swedish Rheumatology Quality Register (SRQ, n = 2820). Information on social support and decision latitude at work at RA diagnosis were identified from the EIRA questionnaire. Indexes for levels of social support and decision latitude at work, respectively, were calculated based on the questionnaire. Low social support and low decision latitude at work, respectively, were identified by a score in the lowest quartile and compared with the three other quartiles (not low). Disease-activity parameters were retrieved from SRQ at 3, 12, and 60 months. The associations between social support or decision latitude at work, respectively, and Disease Activity Score 28 joint count with C-reactive protein (DAS28-CRP) remission were analysed using logistic regression models adjusted for age, sex, smoking habits, alcohol habits, symptom duration, and educational level., Results: Having low social support (n = 591) was not associated with DAS28-CRP remission at 3 (OR 0.93, 95% CI 0.74-1.16), 12 (OR 0.96, 95%CI 0.75-1.23), or 60 (OR 0.89, 95%CI 0.72-1.10) months compared to not low social support (n = 2209). No association was observed for low (n = 212) versus not low (n = 635) decision latitude at work and DAS28-CRP remission at 3 (OR 0.84, 95%CI 0.54-1.31), 12 (OR 0.81, 95%CI 0.56-1.16), or 60 (OR 1.37, 95%CI 0.94-2.01) months., Conclusion: In a country with general access to healthcare, psychosocial vulnerability does not influence the likelihood of achieving remission in early RA., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
49. Investigation of the association between coffee and risk of RA-results from the Swedish EIRA study.
- Author
-
Westerlind H, Dukuzimana J, Lu X, Alfredsson L, Klareskog L, and Di Giuseppe D
- Subjects
- Case-Control Studies, Female, Humans, Male, Middle Aged, Risk Factors, Sweden epidemiology, Arthritis, Rheumatoid epidemiology, Arthritis, Rheumatoid etiology, Coffee adverse effects
- Abstract
Background: Studies on the association between coffee, a modifiable lifestyle factor, and rheumatoid arthritis (RA), a chronic autoimmune disease primarily affecting the joints, have been conflicting. The aim of the present study was to study the association between coffee consumption and risk of RA in the context of different lifestyle factors., Methods: We included 2184 cases (72% women, mean age 55 years) newly diagnosed with RA during 2005-2018 in Sweden and 4201 controls matched on age, sex, and residential area. Data on coffee consumption was collected through a food frequency questionnaire and categorized into < 2 (reference), 2-< 4, 4-< 6, and ≥ 6 cups/day. We calculated odds ratios (OR) with 95% confidence intervals (CI) for coffee consumption and risk of RA, in a crude model (taking matching factors into account), and then adjusted first for smoking and further for BMI, educational level, alcohol consumption, and physical activity. We also stratified analyses on sex, smoking, rheumatoid factor, and anti-CCP2 status., Results: In the crude model, high coffee consumption was associated with increased risk of RA (OR = 1.50, 95% CI 1.20-1.88 for ≥ 6 cups/day compared to < 2 cups). After adjusting for smoking, the OR decreased and was no longer statistically significant (OR = 1.16, 95% CI 0.92-1.46) and decreased further in the full model (OR = 1.14 95% CI 0.89-1.45). This pattern held true in all strata., Conclusion: The findings from this large, population-based case-control study did not support a significant association between coffee consumption and risk of RA as a whole nor within different subgroups., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
50. Real-World Six- and Twelve-Month Drug Retention, Remission, and Response Rates of Secukinumab in 2,017 Patients With Psoriatic Arthritis in Thirteen European Countries.
- Author
-
Michelsen B, Georgiadis S, Di Giuseppe D, Loft AG, Nissen MJ, Iannone F, Pombo-Suarez M, Mann H, Rotar Z, Eklund KK, Kvien TK, Santos MJ, Gudbjornsson B, Codreanu C, Yilmaz S, Wallman JK, Brahe CH, Möller B, Favalli EG, Sánchez-Piedra C, Nekvindova L, Tomsic M, Trokovic N, Kristianslund EK, Santos H, Löve TJ, Ionescu R, Pehlivan Y, Jones GT, van der Horst-Bruinsma I, Ørnbjerg LM, Østergaard M, and Hetland ML
- Subjects
- Europe, Humans, Interleukin-17 antagonists & inhibitors, Treatment Outcome, Antibodies, Monoclonal, Humanized therapeutic use, Antirheumatic Agents therapeutic use, Arthritis, Psoriatic diagnosis, Arthritis, Psoriatic drug therapy
- Abstract
Objective: There is a lack of real-life studies on interleukin-17 (IL-17) inhibition in psoriatic arthritis (PsA). We assessed real-life 6- and 12-month effectiveness (i.e., retention, remission, low disease activity [LDA], and response rates) of the IL-17 inhibitor secukinumab in PsA patients overall and across 1) number of prior biologic/targeted synthetic disease-modifying antirheumatic drugs (b/tsDMARDs), 2) years since diagnosis, and 3) European registries., Methods: Thirteen quality registries in rheumatology participating in the European Spondyloarthritis Research Collaboration Network provided longitudinal, observational data collected as part of routine care for secondary use. Data were pooled and analyzed with Kaplan-Meier plots, log rank tests, Cox regression, and multiple linear and logistic regression analyses., Results: A total of 2,017 PsA patients started treatment with secukinumab between 2015 and 2018. Overall secukinumab retention rates were 86% and 76% after 6 and 12 months, respectively. Crude (LUNDEX adjusted) 6-month remission/LDA (LDA including remission) rates for the 28-joint Disease Activity Index for Psoriatic Arthritis, the Disease Activity Score in 28 joints using the C-reactive protein level, and the Simplified Disease Activity Index (SDAI) were 13%/46% (11%/39%), 36%/55% (30%/46%), and 13%/56% (11%/47%), and 12-month rates were 11%/46% (7%/31%), 39%/56% (26%/38%), and 16%/62% (10%/41%), respectively. Clinical Disease Activity Index remission/LDA rates were similar to the SDAI rates. Six-month American College of Rheumatology 20%/50%/70% improvement criteria responses were 34%/19%/11% (29%/16%/9%); 12-month rates were 37%/21%/11% (24%/14%/7%). Secukinumab effectiveness was significantly better for b/tsDMARD-naive patients, similar across time since diagnosis (<2/2-4/>4 years), and varied significantly across the European registries., Conclusion: In this large real-world study on secukinumab treatment in PsA, 6- and 12-month effectiveness was comparable to that in previous observational studies of tumor necrosis factor inhibitors. Retention, remission, LDA, and response rates were significantly better for b/tsDMARD-naive patients, were independent of time since diagnosis, and varied significantly across the European countries., (© 2021 The Authors. Arthritis Care & Research published by Wiley Periodicals LLC on behalf of American College of Rheumatology.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.