1
|
Marder G, Quach T, Chadha P, Nandkumar P, Tsang J, Levine T, Schiopu E, Furie R, Davidson A, Narain S. Belimumab treatment of adult idiopathic inflammatory myopathy. Rheumatology (Oxford) 2024; 63:742-750. [PMID: 37326854 PMCID: PMC10907809 DOI: 10.1093/rheumatology/kead281] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/16/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023] Open
Abstract
OBJECTIVE To evaluate belimumab addition to the standard of care in patents with refractory idiopathic inflammatory myopathy (IIM). METHODS We conducted a 40-week multicentre, randomized, double-blind, placebo-controlled trial with 1:1 IV belimumab 10 mg/kg or placebo randomization and a 24-week open-label extension. Clinical responses were measured by the definition of improvement (DOI) and total improvement score (TIS). Flow cytometry analyses were performed on available samples before randomization, at 24 and 60-64 weeks. Descriptive statistics, t-test, Fisher's exact test and analysis of variance tests were used. RESULTS A total of 17 patients were randomized, 15 received five or more doses of belimumab or placebo and were included in the intention-to-treat analysis. More belimumab patients vs placebo attained a TIS ≥40 [55.5% vs 33.3%; P = non-significant (NS)] and achieved the DOI (33.3% vs 16.7%; P = NS) at weeks 40 and 64; the mean TIS was similar among groups. Two patients achieved major responses (TIS = 72.5) after week 40 in the belimumab arm and none in the placebo arm. No improvement in the placebo arm after switching to the open-label phase was observed. There was no steroid-sparing effect. No new safety signals were detected. Although total B cells were not reduced, belimumab induced naïve B cell depletion while enhancing the number and frequency memory B cells. CONCLUSION The study did not meet the primary endpoint and no statistically significant differences were observed in clinical responses between arms. More patients achieved sustained TIS ≥40 and reached the DOI. Most patients who received belimumab for >40 weeks had clinical improvement. Phenotypic changes in B cell populations were not associated with clinical responses. CLINICAL TRIAL REGISTRATION NUMBER Clinicaltrials.gov (https://clinicaltrials.gov/), NCT02347891.
Collapse
Affiliation(s)
- Galina Marder
- Division of Rheumatology, Northwell Health, Donald and Barbara Zucker School of Medicine, Great Neck, NY, USA
| | - Tam Quach
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Priyal Chadha
- Division of Rheumatology, Northwell Health, Donald and Barbara Zucker School of Medicine, Great Neck, NY, USA
| | - Preeya Nandkumar
- Division of Rheumatology, Northwell Health, Donald and Barbara Zucker School of Medicine, Great Neck, NY, USA
| | - Jimmy Tsang
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Todd Levine
- Phoenix Neurological Associates, Phoenix, AZ, USA
| | - Elena Schiopu
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Richard Furie
- Division of Rheumatology, Northwell Health, Donald and Barbara Zucker School of Medicine, Great Neck, NY, USA
| | - Anne Davidson
- Division of Rheumatology, Northwell Health, Donald and Barbara Zucker School of Medicine, Great Neck, NY, USA
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Sonali Narain
- Division of Rheumatology, Northwell Health, Donald and Barbara Zucker School of Medicine, Great Neck, NY, USA
| |
Collapse
|
2
|
Gomez A, Jägerback S, Sjöwall C, Parodis I. Belimumab and antimalarials combined against renal flares in patients treated for extra-renal systemic lupus erythematosus: results from 4 phase III clinical trials. Rheumatology (Oxford) 2024; 63:338-348. [PMID: 37228028 PMCID: PMC10836979 DOI: 10.1093/rheumatology/kead253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/24/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
OBJECTIVES To determine the effect of antimalarial agents (AMA) and different doses and pharmaceutical forms of belimumab on preventing renal flares in patients with SLE treated for extra-renal disease. METHODS We pooled data from the BLISS-52, BLISS-76, BLISS-SC and BLISS-Northeast Asia trials of belimumab (n = 3225), that included patients with active SLE yet no severe ongoing nephritis. Participants were allocated to receive intravenous belimumab 1 mg/kg, intravenous belimumab 10 mg/kg, subcutaneous belimumab 200 mg, or placebo in addition to standard therapy. We estimated hazards of renal flare development throughout the study follow-up (52-76 weeks) using Cox regression analysis. RESULTS In total, 192 patients developed a renal flare after a median of 197 days. Compared with placebo, the risk of renal flares was lower among patients receiving intravenous belimumab 10 mg/kg (HR: 0.62; 95% CI: 0.41, 0.92; P = 0.018) and intravenous belimumab 1 mg/kg (HR: 0.42; 95% CI: 0.22, 0.79; P = 0.007), while no significant association was found for subcutaneous belimumab 200 mg. AMA use yielded a lower hazard of renal flares (HR: 0.66; 95% CI: 0.55, 0.78; P < 0.001). The protection conferred was enhanced when belimumab and AMA were co-administered; the lowest flare rate was observed for the combination intravenous belimumab 1 mg/kg and AMA (18.5 cases per 1000 person-years). CONCLUSIONS The protection conferred from belimumab against renal flare development in patients treated for extra-renal SLE appears enhanced when belimumab was administered along with AMA. The prominent effect of low-dose belimumab warrants investigation of the efficacy of intermediate belimumab doses. CLINICAL TRIAL IDENTIFICATION BLISS-52: NCT00424476; BLISS-76: NCT00410384; BLISS-SC: NCT01484496; BLISS-NEA: NCT01345253.
Collapse
Affiliation(s)
- Alvaro Gomez
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Medical Unit of Gastroenterology, Dermatology, and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Sandra Jägerback
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Division of Rheumatology, Danderyd University Hospital, Stockholm, Sweden
| | - Christopher Sjöwall
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping University, Linköping, Sweden
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Medical Unit of Gastroenterology, Dermatology, and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
3
|
Emamikia S, Gentline C, Enman Y, Parodis I. How Can We Enhance Adherence to Medications in Patients with Systemic Lupus Erythematosus? Results from a Qualitative Study. J Clin Med 2022; 11:jcm11071857. [PMID: 35407466 PMCID: PMC8999748 DOI: 10.3390/jcm11071857] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Medication non-adherence is common among patients with systemic lupus erythematosus (SLE) and may lead to poor clinical outcomes. Our aim was to identify influenceable contributors to medication non-adherence and suggest interventions that could increase adherence. Patients with SLE from two Swedish tertiary referral centres (n = 205) participated in a survey assessing self-reported adherence to medications. Responses were used to select patients for qualitative interviews (n = 15). Verbatim interview transcripts were analysed by two researchers using content analysis methodology. The median age of the interviewees was 32 years, 87% were women, and their median SLE duration was nine years. Reasons for non-adherence were complex and multifaceted; we categorised them thematically into (i) patient-related (e.g., unintentional non-adherence due to forgetfulness or intentional non-adherence due to disbelief in medications); (ii) healthcare-related (e.g., untrustworthy relationship with the treating physician, authority fear, and poor information about the prescribed medications or the disease); (iii) medication-related (e.g., fear of side-effects); and (iv) disease-related reasons (e.g., lacking acceptance of a chronic illness or perceived disease quiescence). Interventions identified that healthcare could implement to improve patient adherence to medications included (i) increased communication between healthcare professionals and patients; (ii) patient education; (iii) accessible healthcare, preferably with the same personnel; (iv) well-coordinated transition from paediatric to adult care; (v) regularity in addressing adherence to medications; (vi) psychological support; and (vii) involvement of family members or people who are close to the patient.
Collapse
Affiliation(s)
- Sharzad Emamikia
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden; (C.G.); (Y.E.)
- Correspondence: (S.E.); (I.P.)
| | - Cidem Gentline
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden; (C.G.); (Y.E.)
| | - Yvonne Enman
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden; (C.G.); (Y.E.)
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, 17176 Stockholm, Sweden; (C.G.); (Y.E.)
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, 70182 Örebro, Sweden
- Correspondence: (S.E.); (I.P.)
| |
Collapse
|
4
|
Lu C, Liu L, Chen S, Niu J, Li S, Xie W, Cheng X. Azathioprine pretreatment ameliorates myocardial ischaemia reperfusion injury in diabetic rats by reducing oxidative stress, apoptosis, and inflammation. Clin Exp Pharmacol Physiol 2021; 48:1621-1632. [PMID: 34370882 PMCID: PMC9291025 DOI: 10.1111/1440-1681.13569] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022]
Abstract
This study was presented to observe the therapeutic effects of azathioprine (AZA) pretreatment on myocardial ischaemia reperfusion (I/R) damage in diabetic rats. All rats were randomly separated into control + sham operation; control +I/R; diabetes mellitus (DM) +I/R and DM +I/R + AZA groups. Diabetic rat models were established by intraperitoneally injecting 60 mg/kg streptozotocin (STZ). Diabetic rats were given 3 mg/kg AZA daily by gavage for 5 days. Then, myocardial I/R rat models were constructed. Myocardial infarction size and myocardial damage were respectively detected by TTC and H&E staining. Cardiac injury markers (CK-MB and MPO) and oxidative stress factors (SOD and MDA) were measured via ELISA. The protein expression of apoptotic markers (Caspase8, Caspase3, BAX and Bcl2), inflammatory factors (TLR4 and TNF-α) and AKT1/GSK3β in myocardial tissues was measured by western blot, immunohistochemistry or immunofluorescence. Data showed that AZA pretreatment could lessen myocardial infarction size and myocardial damage, and could down-regulate serum CK-MB, MPO, SOD and MDA levels in diabetic rats under I/R. Furthermore, AZA pretreatment decreased Caspase8, Caspase3, BAX, TLR4 and TNF-α expression, and increased Bcl2 expression in myocardial tissues of diabetic rats following I/R. Also, AZA pretreatment lowered AKT1, p-AKT1, GSK3β and p-GSK3β expression in diabetic heart after I/R. This study found that AZA may reduce myocardial injury in diabetic rats following I/R via reducing oxidative stress, cardiomyocyte apoptosis, and inflammatory response, which could be related to AKT1/GSK3β pathway inactivation.
Collapse
Affiliation(s)
- Cuijie Lu
- Department of Basic MedicineSichuan Vocational College of Health and RehabilitationZigongChina
| | - Ling Liu
- Department of Basic MedicineSichuan Vocational College of Health and RehabilitationZigongChina
| | - Shuai Chen
- Department of Basic MedicineSichuan Vocational College of Health and RehabilitationZigongChina
| | - Junfei Niu
- Department of Basic MedicineSichuan Vocational College of Health and RehabilitationZigongChina
| | - Sheng Li
- Department of Basic MedicineSichuan Vocational College of Health and RehabilitationZigongChina
| | - Wenxian Xie
- Department of Basic MedicineSichuan Vocational College of Health and RehabilitationZigongChina
| | - Xiang Cheng
- Department of Basic MedicineSichuan Vocational College of Health and RehabilitationZigongChina
| |
Collapse
|
5
|
Parodis I, Vital EM, Hassan SU, Jönsen A, Bengtsson AA, Eriksson P, Leonard D, Gunnarsson I, Rönnblom L, Sjöwall C. De novo lupus nephritis during treatment with belimumab. Rheumatology (Oxford) 2021; 60:4348-4354. [PMID: 33341888 PMCID: PMC8409994 DOI: 10.1093/rheumatology/keaa796] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/26/2020] [Indexed: 11/16/2022] Open
Abstract
Objective In light of reports of de novo LN during belimumab (BLM) treatment, we sought to determine its frequency and contributing or protective factors in a real-life setting. Methods Patients with SLE who received BLM between 2011 and 2017 at five European academic practices were enrolled (n = 95) and followed longitudinally for a median time of 13.1 months [interquartile range (IQR): 6.0–34.7]; 52.6% were anti-dsDNA positive, 60.0% had low complement levels, and 69.5% had no renal involvement prior to/at BLM initiation [mean disease duration at baseline: 11.4 (9.3) years]. Age- and sex-matched patients with non-renal SLE who had similar serological profiles, but were not exposed to BLM, served as controls (median follow-up: 132.0 months; IQR: 98.3–151.2). Results We observed 6/66 cases (9.1%) of biopsy-proven de novo LN (4/6 proliferative) among the non-renal BLM-treated SLE cases after a follow-up of 7.4 months (IQR: 2.7–22.2). Among controls, 2/66 cases (3.0%) of de novo LN (both proliferative) were observed after 21 and 50 months. BLM treatment was associated with an increased frequency and/or shorter time to de novo LN [hazard ratio (HR): 10.7; 95% CI: 1.7, 67.9; P = 0.012], while concomitant use of antimalarial agents along with BLM showed an opposing association (HR: 0.2; 95% CI: 0.03, 0.97; P = 0.046). Conclusion Addition of BLM to standard-of-care did not prevent LN in patients with active non-renal SLE, but a favourable effect of concomitant use of antimalarials was implicated. Studies of whether effects of B-cell activating factor inhibition on lymphocyte subsets contribute to LN susceptibility are warranted.
Collapse
Affiliation(s)
- Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Sabih-Ul Hassan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Andreas Jönsen
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Lund
| | - Anders A Bengtsson
- Department of Clinical Sciences Lund, Rheumatology, Lund University, Lund
| | - Per Eriksson
- Rheumatology/Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping
| | - Dag Leonard
- Department of Medical Sciences, Division of Rheumatology, Uppsala University, Uppsala, Sweden
| | - Iva Gunnarsson
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Division of Rheumatology, Uppsala University, Uppsala, Sweden
| | - Christopher Sjöwall
- Rheumatology/Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping
| |
Collapse
|
6
|
Lindblom J, Gomez A, Borg A, Emamikia S, Ladakis D, Matilla J, Pehr M, Cobar F, Enman Y, Heintz E, Regardt M, Parodis I. EQ-5D-3L full health state discriminates between drug and placebo in clinical trials of systemic lupus erythematosus. Rheumatology (Oxford) 2021; 60:4703-4716. [PMID: 33502473 PMCID: PMC8487305 DOI: 10.1093/rheumatology/keab080] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/17/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES The objectives of this study were to investigate the discriminative ability of EQ-5D-3L full health state (FHS) in clinical trials of SLE, and to identify factors associated with FHS after treatment. METHODS Data from the BLISS-52 (NCT00424476) and BLISS-76 (NCT00410384) trials of belimumab (N = 1684) were utilized. FHS was defined as a response of no problems in all five EQ-5D-3L dimensions, yielding an index score of 1. The Pearson's χ2 or Fisher's exact test was employed for comparisons, and logistic regression for adjustments and assessment of independence. RESULTS We demonstrated higher EQ-5D-3L FHS frequencies among patients given standard therapy (ST) plus the licensed belimumab dose vs ST alone (26.1% vs 19.4%; P = 0.001; week 52), and within SRI-4 responders vs non-responders (27.0% vs 19.8%; P < 0.001; week 52) from weeks 36 to 52. In multivariable regression analysis, SLEDAI-2K (OR: 0.90; 95% CI: 0.87, 0.94; P < 0.001) and SLICC/ACR Damage Index (OR: 0.79; 95% CI: 0.69, 0.91; P = 0.001) scores were independently associated with lower FHS frequencies at week 52, while adding monthly infusions of belimumab 10 mg/kg to ST favoured FHS perception (OR: 1.60; 95% CI: 1.15, 2.24; P = 0.006). Add-on belimumab 10 mg/kg yielded higher FHS frequencies in antimalarial users vs non-users (29.9% vs 20.1%; P = 0.011), and in anti-dsDNA- and anti-Sm- positive vs negative patients (31.4% vs 13.4%; P < 0.001 and 33.0% vs 22.6%; P = 0.010, respectively), whereas no significant differences were observed in patients given ST alone. CONCLUSION EQ-5D-3L FHS distinguished belimumab from placebo and responders from non-responders, and exhibited known-group validity in subgroup analysis. FHS may prove a useful patient-reported outcome in SLE studies.
Collapse
Affiliation(s)
- Julius Lindblom
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Alvaro Gomez
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Alexander Borg
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Sharzad Emamikia
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Dimitris Ladakis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Joaquin Matilla
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Martin Pehr
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Flordelyn Cobar
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Yvonne Enman
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| | - Emelie Heintz
- Department of Learning, Informatics, Management and Ethics (LIME)
| | - Malin Regardt
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet
- Occupational Therapy and Physiotherapy, Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital
| |
Collapse
|
7
|
Hernández-Breijo B, Brenis CM, Plasencia-Rodríguez C, Martínez-Feito A, Novella-Navarro M, Pascual-Salcedo D, Balsa A. Methotrexate Reduces the Probability of Discontinuation of TNF Inhibitors in Seropositive Patients With Rheumatoid Arthritis. A Real-World Data Analysis. Front Med (Lausanne) 2021; 8:692557. [PMID: 34268325 PMCID: PMC8275858 DOI: 10.3389/fmed.2021.692557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
Tumor necrosis factor inhibitors (TNFi) are widely used for the treatment of patients with rheumatoid arthritis (RA), however a considerable percentage of patients discontinued the therapy. The aim of this study is to explore real-world TNFi survival, stratified for seropositivity, and to determine the factors that may influence it. This is a retrospective, observational and longitudinal study, using real-world data of patients, who started their first TNFi therapy between 1999 and 2018 from the RA-PAZ cohort. Patients were considered seropositive if they showed positive serum levels of either RF, ACPA, or both. Treatment survival was analyzed using Kaplan-Meier curves, and Cox proportional hazards models were used to compare the risks of TNFi discontinuation for seronegative and seropositive patients. Of the included 250 patients, 213 (85%) were seropositive. Results showed that TNFi survival did not depend on seropositivity status. However, median survival time was significant longer for seropositive patients who received concomitant MTX compared to patients who did not receive it (median [95% CI]: 3.3 yr. [2.3-4.2] vs. 2.6 yr. [1.7-3.6], respectively; p = 0.008). Furthermore, seropositive patients who received concomitant MTX were 49% less likely to discontinue TNFi therapy than patients who did not receive it (HR: 0.51; 95% CI: 0.35-0.74). In addition, we found that in seropositive patients, the use of prednisone throughout the TNFi treatment was associated with a higher likelihood of therapy discontinuation (OR: 2.30; 95% CI: 1.01-5.23). In conclusion, these data provide evidence to support the use of concomitant MTX in seropositive patients to prolong the effectiveness and the survival of the TNFi therapy. Moreover, the co-administration of prednisone in seropositive patients receiving TNFi was highly associated with TNFi discontinuation.
Collapse
Affiliation(s)
- Borja Hernández-Breijo
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain
| | - Claudia M Brenis
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain
| | - Chamaida Plasencia-Rodríguez
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain.,Rheumatology, La Paz University Hospital, Madrid, Spain
| | - Ana Martínez-Feito
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain.,Immunology, La Paz University Hospital, Madrid, Spain
| | - Marta Novella-Navarro
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain.,Rheumatology, La Paz University Hospital, Madrid, Spain
| | - Dora Pascual-Salcedo
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain
| | - Alejandro Balsa
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain.,Rheumatology, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
8
|
BAFF predicts immunogenicity in older patients with rheumatoid arthritis treated with TNF inhibitors. Sci Rep 2021; 11:11632. [PMID: 34079038 PMCID: PMC8172642 DOI: 10.1038/s41598-021-91177-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/24/2021] [Indexed: 11/08/2022] Open
Abstract
Immunogenicity related to treatment with TNF inhibitors (TNFi) is one of the causes for the decreased attainment of clinical response in patients with rheumatoid arthritis (RA). The B-cell activating factor (BAFF) may be playing a role in the development of immunogenicity. The objective of this study was to analyse the association of baseline concentration of serum B-cell activating factor (BAFF) with immunogenicity after 6 months of TNFi treatment. A total of 127 patients with RA starting a TNFi (infliximab, adalimumab, certolizumab pegol or golimumab) were followed-up for 6 months. Serum samples were obtained at baseline and at 6 months and anti-drug antibody (ADA) and BAFF concentrations were measured. Logistic regression models were employed in order to analyse the association between BAFF concentrations and immunogenicity. Receiver operating characteristic analysis was performed to determine the BAFF concentrations with a greater likelihood of showing immunogenicity association. At 6 months, 31 patients (24%) developed ADA. A significant interaction between the age and baseline BAFF concentration was found for the development of ADA (Wald chi-square value = 5.30; p = 0.02); therefore, subsequent results were stratified according to mean age (≤ / > 55 years). Baseline serum BAFF concentration was independently associated with ADA development only in patients over 55 years (OR = 1.51; 95% CI 1.03–2.21). Baseline serum BAFF ≥ 1034 pg/mL predicted the presence of ADA at 6 months (AUC = 0.81; 95% confidence interval (CI) 0.69–0.93; p = 0.001; positive likelihood ratio = 3.7). In conclusion, our results suggest that the association of BAFF concentration and immunogenicity depends on the patient’s age. Baseline serum BAFF concentration predicts the presence of ADA within 6 months of TNFi therapy in older patients with RA.
Collapse
|
9
|
B Cells and Antibodies as Targets of Therapeutic Intervention in Neuromyelitis Optica Spectrum Disorders. Pharmaceuticals (Basel) 2021; 14:ph14010037. [PMID: 33419217 PMCID: PMC7825598 DOI: 10.3390/ph14010037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/02/2021] [Accepted: 01/03/2021] [Indexed: 12/11/2022] Open
Abstract
The first description of neuromyelitis optica by Eugène Devic and Fernand Gault dates back to the 19th century, but only the discovery of aquaporin-4 autoantibodies in a major subset of affected patients in 2004 led to a fundamentally revised disease concept: Neuromyelits optica spectrum disorders (NMOSD) are now considered autoantibody-mediated autoimmune diseases, bringing the pivotal pathogenetic role of B cells and plasma cells into focus. Not long ago, there was no approved medication for this deleterious disease and off-label therapies were the only treatment options for affected patients. Within the last years, there has been a tremendous development of novel therapies with diverse treatment strategies: immunosuppression, B cell depletion, complement factor antagonism and interleukin-6 receptor blockage were shown to be effective and promising therapeutic interventions. This has led to the long-expected official approval of eculizumab in 2019 and inebilizumab in 2020. In this article, we review current pathogenetic concepts in NMOSD with a focus on the role of B cells and autoantibodies as major contributors to the propagation of these diseases. Lastly, by highlighting promising experimental and future treatment options, we aim to round up the current state of knowledge on the therapeutic arsenal in NMOSD.
Collapse
|
10
|
Möckel T, Basta F, Weinmann-Menke J, Schwarting A. B cell activating factor (BAFF): Structure, functions, autoimmunity and clinical implications in Systemic Lupus Erythematosus (SLE). Autoimmun Rev 2020; 20:102736. [PMID: 33333233 DOI: 10.1016/j.autrev.2020.102736] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 08/28/2020] [Indexed: 12/30/2022]
Abstract
The B cell activating factor (BAFF), or B lymphocyte stimulator (BLyS), is a B cell survival factor which supports autoreactive B cells and prevents their deletion. BAFF expression is closely linked with autoimmunity and is enhanced by genetic alterations and viral infections. Furthermore, BAFF seems to be involved in adipogenesis, atherosclerosis, neuro-inflammatory processes and ischemia reperfusion (I/R) injury. BAFF is commonly overexpressed in Systemic Lupus Erythematosus (SLE) and strongly involved in the pathogenesis of the disease. The relationship between BAFF levels, disease activity and damage accrual in SLE is controversial, but growing evidence is emerging on its role in renal involvement. Belimumab, a biologic BAFF inhibitor, has been the first biologic agent licensed for SLE therapy so far. As Rituximab (RTX) has been shown to increase BAFF levels following B cell depletion, the combination therapy of RTX plus belimumab (being evaluated in two RCT) seems to be a valuable option for several clinical scenarios. In this review we will highlight the growing body of evidence of immune and non-immune related BAFF expression in experimental and clinical settings.
Collapse
Affiliation(s)
- Tamara Möckel
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Fabio Basta
- Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany
| | - Julia Weinmann-Menke
- Department of Internal Medicine I, Division of Nephrology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Schwarting
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany; Acura Rheumatology Center Rhineland Palatinate, Bad Kreuznach, Germany
| |
Collapse
|
11
|
Lambers WM, Westra J, Bootsma H, de Leeuw K. Hydroxychloroquine Suppresses Interferon-inducible Genes and B Cell Activating Factor in Patients With Incomplete and New-onset Systemic Lupus Erythematosus. J Rheumatol 2020; 48:847-851. [DOI: 10.3899/jrheum.200726] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 11/22/2022]
Abstract
Objective.Hydroxychloroquine (HCQ) is commonly used as first-line treatment for systemic lupus erythematosus (SLE). Interferon (IFN)-inducible gene expression, IFN-γ–induced protein 10 (IP-10) and B cell activating factor (BAFF) are early mediators in SLE. The purpose of this study was to analyze the effects of HCQ on these factors.Methods.Patients with incomplete SLE (iSLE; antinuclear antibody titer ≥ 1:80, symptoms < 5 years, ≥ 1 objectified clinical American College of Rheumatology or SLE International Collaborating Clinics criteria), or new-onset, mild SLE were included when HCQ treatment was started for clinical reasons. Blood samples were taken at start and after 16 weeks. Three SLE-related IFN-inducible genes were measured in whole blood by real-time PCR, and an IFN score was calculated. Serum levels of IP-10 and BAFF were measured using ELISA.Results.In total, 9 patients were included: 7 with iSLE and 2 with new-onset SLE. The median SLE Disease Activity Index (SLEDAI) was 4. After 16 weeks of treatment with HCQ, the expression of IFN-inducible genes decreased in 8 of 9 patients, and the IFN-3 score decreased significantly (P = 0.012). There was a trend towards lower IP-10 levels (P = 0.055), and a significant decrease in BAFF levels (P = 0.023).Conclusion.HCQ suppresses IFN score and BAFF levels in patients with iSLE or new-onset SLE, and there is a trend towards lowering IP-10 levels. As these biomarkers are early mediators in SLE, this might support the hypothesis that HCQ could influence disease progression. However, prospective research with a larger sample size and longer follow-up is needed.
Collapse
|
12
|
Differential Effects of MS Therapeutics on B Cells-Implications for Their Use and Failure in AQP4-Positive NMOSD Patients. Int J Mol Sci 2020; 21:ijms21145021. [PMID: 32708663 PMCID: PMC7404039 DOI: 10.3390/ijms21145021] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022] Open
Abstract
B cells are considered major contributors to multiple sclerosis (MS) pathophysiology. While lately approved disease-modifying drugs like ocrelizumab deplete B cells directly, most MS medications were not primarily designed to target B cells. Here, we review the current understanding how approved MS medications affect peripheral B lymphocytes in humans. These highly contrasting effects are of substantial importance when considering these drugs as therapy for neuromyelitis optica spectrum disorders (NMOSD), a frequent differential diagnosis to MS, which is considered being a primarily B cell- and antibody-driven diseases. Data indicates that MS medications, which deplete B cells or induce an anti-inflammatory phenotype of the remaining ones, were effective and safe in aquaporin-4 antibody positive NMOSD. In contrast, drugs such as natalizumab and interferon-β, which lead to activation and accumulation of B cells in the peripheral blood, lack efficacy or even induce catastrophic disease activity in NMOSD. Hence, we conclude that the differential effect of MS drugs on B cells is one potential parameter determining the therapeutic efficacy or failure in antibody-dependent diseases like seropositive NMOSD.
Collapse
|
13
|
Belizna C, Meroni PL, Shoenfeld Y, Devreese K, Alijotas-Reig J, Esteve-Valverde E, Chighizola C, Pregnolato F, Cohen H, Fassot C, Mattera PM, Peretti P, Levy A, Bernard L, Saiet M, Lagarce L, Briet M, Rivière M, Pellier I, Gascoin G, Rakotonjanahary J, Borghi MO, Stojanovich L, Djokovic A, Stanisavljevic N, Bromley R, Elefant-Amoura E, Bahi Buisson N, Pindi Sala T, Kelchtermans H, Makatsariya A, Bidsatze V, Khizroeva J, Latino JO, Udry S, Henrion D, Loufrani L, Guihot AL, Muchardt C, Hasan M, Ungeheuer MN, Voswinkel J, Damian L, Pabinger I, Gebhart J, Lopez Pedrera R, Cohen Tervaert JW, Tincani A, Andreoli L. In utero exposure to Azathioprine in autoimmune disease. Where do we stand? Autoimmun Rev 2020; 19:102525. [PMID: 32240856 DOI: 10.1016/j.autrev.2020.102525] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/19/2022]
Abstract
Azathioprine (AZA), an oral immunosuppressant, is safe during pregnancy. Some reports suggested different impairments in the offspring of mothers with autoimmune diseases (AI) exposed in utero to AZA. These observations are available from retrospective studies or case reports. However, data with respect to the long-term safety in the antenatally exposed child are still lacking. The aim of this study is to summarize the current knowledge in this field and to focus on the need for a prospective study on this population. We performed a PubMed search using several search terms. The actual data show that although the risk of congenital anomalies in offspring, as well as the infertility risk, are similar to those found in general population, there is a higher incidence of prematurity, of lower weight at birth and an intra-uterine delay of development. There is also an increased risk of materno- fetal infections, especially cytomegalovirus infection. Some authors raise the interrogations about neurocognitive impairment. Even though the adverse outcomes might well be a consequence of maternal illness and disease activity, interest has been raised about a contribution of this drug. However, the interferences between the external agent (in utero exposure to AZA), with the host (child genetic susceptibility, immune system anomalies, emotional status), environment (public health, social context, availability of health care), economic, social, and behavioral conditions, cultural patterns, are complex and represent confounding factors. In conclusion, it is necessary to perform studies on the medium and long-term outcome of children born by mothers with autoimmune diseases, treated with AZA, in order to show the safety of AZA exposure. Only large-scale population studies with long-term follow-up will allow to formally conclude in this field. TAKE HOME MESSAGES.
Collapse
Affiliation(s)
- Cristina Belizna
- Vascular and Coagulation Department, University Hospital Angers, Angers, France; MITOVASC institute and CARFI facility, University of Angers, UMR CNRS 6015, INSERM U1083, Angers, France; Internal Medicine Department, Clinique de l'Anjou, Angers, France; UMR CNRS 6015, Angers, France; INSERM U1083, Angers, France.
| | - Pier Luigi Meroni
- Clinical Immunology and Rheumatology Research Department Auxologico Institute, Milan, Italy
| | - Yehuda Shoenfeld
- The Zabludowicz Center for Autoimmune Diseases, Affiliated to Sackler Faculty of Medicine, Tel-Aviv University, Israel; I.M. Sechenow First Moscow State Medical University, Moscow, Russia
| | - Katrien Devreese
- Coagulation Laboratory, Department of Clinical Biology, Immunology and Microbiology, Ghent University Hospital, Ghent, Belgium
| | - Jaume Alijotas-Reig
- Systemic Autoimmune Disease Unit, Department of Internal Medicine, Vall d'Hebron University Hospital, Barcelona, Spain; Department of Medicine, Universitat Autonòma, Barcelona, Spain
| | | | - Cecilia Chighizola
- Clinical Immunology and Rheumatology Research Department Auxologico Institute, Milan, Italy
| | - Francesca Pregnolato
- Clinical Immunology and Rheumatology Research Department Auxologico Institute, Milan, Italy
| | - Hannah Cohen
- Haematology Department, University College Hospital, London, UK
| | - Celine Fassot
- Internal Medicine Department, Clinique de l'Anjou, Angers, France
| | - Patrick Martin Mattera
- Faculty of Human and Social Sciences, Laboratory of Research in Psychopathology, 3 place André Leroy, 49008 Angers, France
| | - Pascale Peretti
- Faculty of Human and Social Sciences, Laboratory of Research in Psychopathology, 3 place André Leroy, 49008 Angers, France
| | - Alexandre Levy
- Faculty of Human and Social Sciences, Laboratory of Research in Psychopathology, 3 place André Leroy, 49008 Angers, France
| | - Laurence Bernard
- Faculty of Human and Social Sciences, Laboratory of Research in Psychopathology, 3 place André Leroy, 49008 Angers, France
| | - Mathilde Saiet
- Faculty of Human and Social Sciences, Laboratory of Research in Psychopathology, 3 place André Leroy, 49008 Angers, France
| | - Laurence Lagarce
- Departement of Pharmacovigilance, University Hospital Angers, Angers, France
| | - Marie Briet
- Departement of Pharmacovigilance, University Hospital Angers, Angers, France
| | - Marianne Rivière
- French Lupus and Other Autoimmune Disease Patients Association, AFL+, Cuvry, France
| | - Isabelle Pellier
- Department of Pediatrics, University Hospital Angers, Angers, France
| | - Géraldine Gascoin
- Department of Neonatology, University Hospital Angers, Angers, France
| | | | - Maria Orietta Borghi
- Clinical Immunology and Rheumatology Research Department Auxologico Institute, Milan, Italy
| | - Ljudmila Stojanovich
- Scientific Research Department, Internal Medicine-Rheumatology Bezhanijska Kosa, University Medical Center, Belgrade University, Serbia
| | - Aleksandra Djokovic
- Scientific Research Department, Internal Medicine-Rheumatology Bezhanijska Kosa, University Medical Center, Belgrade University, Serbia
| | - Natasa Stanisavljevic
- Scientific Research Department, Internal Medicine-Rheumatology Bezhanijska Kosa, University Medical Center, Belgrade University, Serbia
| | - Rebecca Bromley
- Manchester University Hospitals NHS Trust, Manchester, UK; Division of Evolution and Genomic Science, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Elisabeth Elefant-Amoura
- Genetical and Medical Embriology, CRAT Reference Center on Teratogenic Agents, Paris Est - Hôpital d'Enfants Armand-Trousseau, 26 avenue du Docteur Arnold Netter, 75571 Paris, France
| | - Nadia Bahi Buisson
- Neurology & Neurodevelopmental disorders Department Necker Enfants Malades University Hospital, APHP, Paris 149 Rue de Sèvres, 75015 Paris; INSERM U1163, Hôpital Necker Enfants Malades, 149 rue de Sèvres, 75015 Paris, France; INSERM U1163, Hôpital Necker Enfants Malades, 149 rue de Sèvres, 75015 Paris, France
| | - Taylor Pindi Sala
- EA 7334, Patient Centered Outcomes Research, University Paris Diderot, Paris, France
| | - Hilde Kelchtermans
- Synapse Research Institute, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Alexander Makatsariya
- Department of Obstetrics and Gynecology, I.M. Sechenow First Moscow State Medical University, Moscow, Russia
| | - Viktoria Bidsatze
- Department of Obstetrics and Gynecology, I.M. Sechenow First Moscow State Medical University, Moscow, Russia
| | - Jamilya Khizroeva
- Department of Obstetrics and Gynecology, I.M. Sechenow First Moscow State Medical University, Moscow, Russia
| | - Jose Omar Latino
- Autoimmune and thrombophilic disorders Department, Hospital Carlos G. Durand, Buenos Aires, Argentina
| | - Sebastian Udry
- Autoimmune and thrombophilic disorders Department, Hospital Carlos G. Durand, Buenos Aires, Argentina
| | - Daniel Henrion
- Internal Medicine Department, Clinique de l'Anjou, Angers, France
| | - Laurent Loufrani
- Internal Medicine Department, Clinique de l'Anjou, Angers, France
| | | | - Christian Muchardt
- Unit of Epigenetic Regulation, Department of Developmental and Stem Cell Biology, UMR3738 CNRS, Institut Pasteur, Paris, France
| | - Milena Hasan
- Cytometry and Biomarkers Unit of Technology and Service, Center for Translational Science, Institut Pasteur, 28, Rue Doct Roux, 75015 Paris, France
| | - Marie Noelle Ungeheuer
- Clinical Investigation and Acces to Bioresources Department, Institut Pasteur, 28, Rue Doct Roux, 75015 Paris, France
| | - Jan Voswinkel
- Department of Internal Medicine I, Saarland Medical School, University of Saarland, Homburg, Saarland, Germany
| | - Laura Damian
- Department of Rheumatology, County Emergency Hospital Cluj-Napoca, Cluj-Napoca, Romania
| | - Ingrid Pabinger
- Department of Medicine, Division of Hematology and Haemostasis, University Hospital of Vienna, Austria
| | - Johanna Gebhart
- Department of Medicine, Division of Hematology and Haemostasis, University Hospital of Vienna, Austria
| | - Rosario Lopez Pedrera
- Institute Maimónides of Biomedical Investigations, University Hospital Reina Sofía, Cordoba, Spain
| | | | - Angela Tincani
- Rheumatology and Clinical Immunology Unit, University of Brescia, Brescia, Italy; I.M. Sechenow First Moscow State Medical University, Moscow, Russia
| | - Laura Andreoli
- Rheumatology and Clinical Immunology Unit, University of Brescia, Brescia, Italy
| |
Collapse
|