1
|
Abacar K, Macleod T, Direskeneli H, McGonagle D. How underappreciated autoinflammatory (innate immunity) mechanisms dominate disparate autoimmune disorders. Front Immunol 2024; 15:1439371. [PMID: 39372419 PMCID: PMC11449752 DOI: 10.3389/fimmu.2024.1439371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Historically inflammation against self was considered autoimmune which stems back to the seminal observations by Ehrlich who described serum factors, now known to be autoantibodies produced by B lineage cells that mediate "horror autotoxicus". The 20th century elucidation of B- and T-cell adaptive immune responses cemented the understanding of the key role of adaptive immune responses in mediating pathology against self. However, Mechnikov shared the Nobel Prize for the discovery of phagocytosis, the most rudimentary aspect of innate immunity. Fast forward some 100 years and an immunogenetic understanding of innate immunity led to the categorising of innate immunopathology under the umbrella term 'auto inflammation' and terminology such as "horror autoinflammaticus" to highlight the schism from the classical adaptive immune understanding of autoimmunity. These concepts lead to calls for a two-tiered classification of inflammation against self, but just as innate and adaptive immunity are functionally integrated, so is immunopathology in many settings and the concept of an autoimmune to autoinflammation continuum emerged with overlaps between both. Herein we describe several historically designated disorders of adaptive immunity where innate immunity is key, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), systemic juvenile idiopathic arthritis (sJIA) and adult-onset Still's disease (AOSD) where the immunopathology phenotype is strongly linked to major histocompatibility complex (MHC) class II associations and responds to drugs that target T-cells. We also consider MHC-I-opathies including psoriasis and Behcet's disease(BD) that are increasingly viewed as archetype CD8 T-cell related disorders. We also briefly review the key role of barrier dysfunction in eczema and ulcerative colitis (UC) where innate tissue permeability barrier dysfunction and microbial dysbiosis contributes to prominent adaptive immune pathological mechanisms. We also highlight the emerging roles of intermediate populations of lymphocytes including gamma delta (γδ) and mucosal-associated invariant T (MAIT) cells that represent a blend of adaptive immune plasticity and innate immune rapid responders that may also determine site specific patterns of inflammation.
Collapse
Affiliation(s)
- Kerem Abacar
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Tom Macleod
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
| | - Haner Direskeneli
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Dennis McGonagle
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
- National Institute for Health Research, Leeds Biomedical Research Centre, Leeds Teaching Hospitals, Leeds, United Kingdom
| |
Collapse
|
2
|
Lambuk F, Nordin NA, Mussa A, Lambuk L, Ahmad S, Hassan R, Kadir R, Mohamud R, Yahya NK. Towards understanding the role of nanomedicine in targeting TNFR2 in rheumatoid arthritis. Immunology 2024. [PMID: 39191474 DOI: 10.1111/imm.13855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by inflammation of the synovium and progressive joint destruction which significantly affects both quality of life and socioeconomic status. Admittedly, various treatments are available, but they are usually accompanied by various side effects, from mild to severe, and potentially with adverse events. Tumour necrosis factor-alpha (TNF-α) plays a crucial role in the pathophysiology of RA. It promotes inflammatory, apoptosis and necroptosis via TNF receptor-1 (TNFR1) but elicit anti-inflammatory effects via TNFR2. Herein, targeting TNFR2 has gained attention in RA studies. Understanding the role of nanomedicine in modulating TNFR2 signalling may be the instrument in development of RA therapies. Nanotechnology has made a significant progress in treating various conditions of diseases since its inception. Due to this, nanomedicine has emerged as a promising therapeutics approach for RA. Recent studies have demonstrated the potential of nanomedicine in RA theranostics, combining therapy and diagnostics for improved treatment outcomes. Owing to the challenges and advancements in the field of nanotechnology, nanoparticles are seen as an applicable candidate in the treatment of RA. In this review, we provide an overview of the role of nanomedicine in targeting TNFR2 for the treatment of RA and highlight the limitations of current therapies as well as the potential of nanocarriers with controlled drug release and active targeting abilities.
Collapse
Affiliation(s)
- Fatmawati Lambuk
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Nor Asyikin Nordin
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Ali Mussa
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, Omdurman, Sudan
| | - Lidawani Lambuk
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Suhana Ahmad
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Rosline Hassan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Nurul Khaiza Yahya
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| |
Collapse
|
3
|
Valdivieso Shephard JL, Alvarez Robles EJ, Cámara Hijón C, Hernandez Breijo B, Novella-Navarro M, Bogas Schay P, Cuesta de la Cámara R, Balsa Criado A, López Granados E, Plasencia Rodríguez C. Predicting anti-TNF treatment response in rheumatoid arthritis: An artificial intelligence-driven model using cytokine profile and routine clinical practice parameters. Heliyon 2024; 10:e22925. [PMID: 38163219 PMCID: PMC10754867 DOI: 10.1016/j.heliyon.2023.e22925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a heterogeneous disease in which therapeutic strategies used have evolved dramatically. Despite significant progress in treatment strategies such as the development of anti-TNF drugs, it is still not possible to differentiate those patients who will respond from who will not. This can lead to effective-treatment delays and unnecessary costs. The aim of this study was to utilize a profile of the patient's characteristics, clinical parameters, immune status (cytokine profile) and artificial intelligence to assess the feasibility of developing a tool that could allow us to predict which patients will respond to treatment with anti-TNF drugs. Methods This study included 38 patients with RA from the RA-Paz cohort. Clinical activity was measured at baseline and after 6 months of treatment. The cytokines measured before the start of anti-TNF treatment were IL-1, IL-12, IL-10, IL-2, IL-4, IFNg, TNFa, and IL-6. Statistical analyses were performed using the Wilcoxon-Rank-Sum Test and the Benjamini-Hochberg method. The predictive model viability was explored using the 5-fold cross-validation scheme in order to train the logistic regression models. Results Statistically significant differences were found in parameters such as IL-6, IL-2, CRP and DAS-ESR. The predictive model performed to an acceptable level in correctly classifying patients (ROC-AUC 0.804167 to 0.891667), suggesting that it would be possible to develop a clinical classification tool. Conclusions Using a combination of parameters such as IL-6, IL-2, CRP and DAS-ESR, it was possible to develop a predictive model that can acceptably discriminate between remitters and non-remitters. However, this model needs to be replicated in a larger cohort to confirm these findings.
Collapse
Affiliation(s)
| | | | | | - Borja Hernandez Breijo
- Immunology-Rheumatology Research Group, Hospital Universitario La Paz-Idipaz, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
4
|
Wang Z, Zhang C, Meng J, Jiao Z, Bao W, Tian H, Wu C, Chai W, Li R, Liu Z, Ma G, Mei X, Wei W. A Targeted Exosome Therapeutic Confers Both CfDNA Scavenging and Macrophage Polarization for Ameliorating Rheumatoid Arthritis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302503. [PMID: 37681753 DOI: 10.1002/adma.202302503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/20/2023] [Indexed: 09/09/2023]
Abstract
Only a minority of rheumatoid arthritis (RA) patients achieve disease remission, so the exploration of additional pathogenic factors and the development of new therapeutics are needed. Here, strong correlations among the cell-free DNA (cfDNA) level and the inflammatory response in clinical synovial fluid samples and RA disease activity are discovered. The important role of cfDNA in disease development in a collagen-induced arthritis (CIA) murine model is also demonstrated. Building on these findings, a novel therapeutic based on anti-inflammatory (M2) macrophage-derived exosomes as chassis, that are modified with both oligolysine and matrix metalloproteinase (MMP)-cleavable polyethylene glycol (PEG) on the membrane, is developed. After intravenous injection, PEG-enabled prolonged circulation and C─C motif chemokine ligand-directed accumulation together result in enrichment at inflamed joints. Following subsequent MMP cleavage, the positively charged oligolysine is exposed for cfDNA scavenging, while exosomes induce M2 polarization. By using a classical CIA murine model and a newly established CIA canine model, it is demonstrated that the rationally designed exosome therapeutic substantially suppresses inflammation in joints and provides strong chondroprotection and osteoprotection, revealing its potential for effective CIA amelioration.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121002, P. R. China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Chuanjie Zhang
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121002, P. R. China
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Jiaqi Meng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Zhouguang Jiao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Weier Bao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - He Tian
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Chao Wu
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Wei Chai
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100037, P. R. China
| | - Rui Li
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100037, P. R. China
| | - Zheng Liu
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100037, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xifan Mei
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121002, P. R. China
- Key Laboratory of Medical Tissue Engineering of Liaoning, Jinzhou Medical University, Jinzhou, 121001, P. R. China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
5
|
de Klaver PAG, Keizer RJ, Ter Heine R, Smits L, Boekema PJ, Kuntzel I, Schaap T, de Vries A, Bloem K, Rispens T, Hoentjen F, Derijks LJJ. Early At-Home Measurement of Adalimumab Concentrations to Guide Anti-TNF Precision Dosing: A Pilot Study. Eur J Drug Metab Pharmacokinet 2023:10.1007/s13318-023-00835-7. [PMID: 37322238 DOI: 10.1007/s13318-023-00835-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND AND OBJECTIVE Underdosing of adalimumab can result in non-response and poor disease control in patients with rheumatic disease or inflammatory bowel disease. In this pilot study we aimed to predict adalimumab concentrations with population pharmacokinetic model-based Bayesian forecasting early in therapy. METHODS Adalimumab pharmacokinetic models were identified with a literature search. A fit-for-purpose evaluation of the model was performed for rheumatologic and inflammatory bowel disease (IBD) patients with adalimumab peak (first dose) and trough samples (first and seventh dose) obtained by a volumetric absorptive microsampling technique. Steady state adalimumab concentrations were predicted after the first adalimumab administration. Predictive performance was calculated with mean prediction error (MPE) and normalised root mean square error (RMSE). RESULTS Thirty-six patients (22 rheumatologic and 14 IBD) were analysed in our study. After stratification for absence of anti-adalimumab antibodies, the calculated MPE was -2.6% and normalised RMSE 24.0%. Concordance between predicted and measured adalimumab serum concentrations falling within or outside the therapeutic window was 75%. Three patients (8.3%) developed detectable concentrations of anti-adalimumab antibodies. CONCLUSION This prospective study demonstrates that adalimumab concentrations at steady state can be predicted from early samples during the induction phase. CLINICAL TRIAL REGISTRATION The trial was registered in the Netherlands Trial Register with trial registry number NTR 7692 ( www.trialregister.nl ).
Collapse
Affiliation(s)
- Paul A G de Klaver
- Department of Clinical Pharmacy and Pharmacology, Máxima Medical Center, 5504 DB, Veldhoven, The Netherlands.
| | | | - Rob Ter Heine
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lisa Smits
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Paul J Boekema
- Department of Gastroenterology, Máxima Medical Center, Veldhoven, The Netherlands
| | - Inge Kuntzel
- Department of Rheumatology, Máxima Medical Center, Eindhoven, The Netherlands
| | - Tiny Schaap
- Biologics Laboratory, Sanquin Diagnostic Services, Amsterdam, The Netherlands
| | - Annick de Vries
- Biologics Laboratory, Sanquin Diagnostic Services, Amsterdam, The Netherlands
| | - Karien Bloem
- Biologics Laboratory, Sanquin Diagnostic Services, Amsterdam, The Netherlands
| | - Theo Rispens
- Biologics Laboratory, Sanquin Diagnostic Services, Amsterdam, The Netherlands
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Frank Hoentjen
- Department of Gastroenterology, Radboud University Medical Center, Nijmegen, The Netherlands
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Luc J J Derijks
- Department of Clinical Pharmacy and Pharmacology, Máxima Medical Center, 5504 DB, Veldhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
6
|
İnanç GN, Terzioğlu ME, Karabulut Y, Yılmaz Z, Tarhan EF, Enecik ME, Şahin A, Küçük A, Ayan A, Özgen M, Karasu U, Yolbaş S. A national, multicenter, retrospective study evaluating retention rate and efficacy of tocilizumab treatment in patients with active rheumatoid arthritis who had an inadequate response to csDMARDs and/or TNF inhibitors. Turk J Med Sci 2023; 53:731-743. [PMID: 37476902 PMCID: PMC10387905 DOI: 10.55730/1300-0144.5636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/18/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND To describe the disease activity and retention rate in rheumatoid arthritis (RA) patients with inadequate response (IR) to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) and/or tumor necrosis factor inhibitors (TNFis) who were prescribed tocilizumab (TCZ) as first-line or second-line biologic treatment in real-world setting. METHODS Data gathered from patients' files was used in a multicenter and retrospective context. Retention rates and the Disease Activity Score in 28 joints with CRP (DAS28-CRP) were evaluated at time points. The relationship of drug efficacy with factors such as smoking, obesity, and previous use of TNFis was also examined. RESULTS One hundred and twenty-four patients with a median (IQR) RA duration of 3.7 (7.4) years were included. Mean (SD) age was52.9 (12.9) and 75% of the patients were female. TCZ retention rates in the 6th and 12th months were 94.1% and 86.6%, respectively. In all patients, DAS28-CRP level decreased significantly from baseline to Months 3 and 6. There was an increase in patients with remission and/or low disease activity and a decrease in patients with high disease activity at Month 3 and Month 6 (p < 0.001 for both). Disease activity was similar between subgroups based on body mass index, smoking status, and previous use of TNFis at any time point. Regression analysis showed that absence of concomitant corticosteroid treatment independently was associated with remission/LDA achievement at Month 6 [OR = 0.31, 95% CI (0.14- 0.72), p = 0.006], and Month 12 [OR = 0.35, 95% CI (0.13-0.94), p = 0.037]. Overall, 25 mild adverse events were reported. DISCUSSION TCZ was found to be effective and safe in RA patients with IR to csDMARDs and/or TNFis. The drug retention rate was considered satisfactory with more than half of the patients continuing TCZ treatment at Month 12.
Collapse
Affiliation(s)
- Güzide Nevsun İnanç
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Marmara University, İstanbul, Turkey
| | - Mustafa Ender Terzioğlu
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Yusuf Karabulut
- Division of Rheumatology, Department of Internal Medicine, Doruk Yıldırım Hospital, Bursa, Turkey
| | - Zevcet Yılmaz
- Division of Rheumatology, Department of Internal Medicine, Tepecik Education and Research Hospital, Health Sciences University, İzmir, Turkey
| | - Emine Figen Tarhan
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Muğla Sıtkı Koçman University, Muğla, Turkey
| | - Mehmet Emin Enecik
- Division of Rheumatology, Department of Internal Medicine, Medical Park Hospitals, Mersin, Turkey
| | - Ali Şahin
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Adem Küçük
- Division of Rheumatology, Department of Internal Medicine, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ayşe Ayan
- Division of Rheumatology, Department of Internal Medicine, Antalya Training and Research Hospital, Antalya, Turkey
| | - Metin Özgen
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Uğur Karasu
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Servet Yolbaş
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, İnonü University, Malatya, Turkey
| |
Collapse
|
7
|
Miyashiro M, Ishii Y, Miyazaki C, Shimizu H, Masuda J. A Real-World Claims Database Study Assessing Long-Term Persistence with Golimumab Treatment in Patients with Rheumatoid Arthritis in Japan. Rheumatol Ther 2023; 10:615-634. [PMID: 36802051 PMCID: PMC10140228 DOI: 10.1007/s40744-023-00539-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/01/2023] [Indexed: 02/21/2023] Open
Abstract
INTRODUCTION The persistence of golimumab (GLM) treatment in Japanese patients with rheumatoid arthritis (RA) has been evaluated previously, but evidence of long-term real-world use is lacking. This study assessed the long-term persistence of GLM use, its influencing factors, and impact of prior medications in patients with RA in actual clinical practice in Japan. METHODS This is a retrospective cohort study of patients with RA using data from a hospital insurance claims database in Japan. The identified patients were stratified as only GLM treatment (naïve), had one biological disease-modifying anti-rheumatic drug (bDMARD)/Janus kinase (JAK) inhibitor treatment prior to GLM [switch (1)] and had at least two bDMARDs/JAK prior to GLM treatment [switch (≥ 2)]. Patient characteristics were evaluated using descriptive statistics. Kaplan-Meier survival and Cox regression methods were used to analyze GLM persistence at 1, 3, 5, and 7 years and the associated factors. Treatment differences were compared using a log-rank test. RESULTS GLM persistence rate in the naïve group was 58.8%, 32.1%, 21.4%, and 11.4% at 1, 3, 5, and 7 years, respectively. Overall persistence rates in the naïve group were higher than in switch groups. Higher GLM persistence was observed among patients aged 61-75 years and those concomitantly using methotrexate (MTX). Also, women were less likely to discontinue treatment compared to men. Higher Charlson Comorbidity Index score, initial GLM dose of 100 mg, and switch from bDMARDs/JAK inhibitor were related to a lower persistence rate. As a prior medication, infliximab showed the longest persistence for subsequent GLM, and using this as a reference, tocilizumab, sarilumab, and tofacitinib subgroups had significantly shorter persistence, respectively (p = 0.001, 0.025, 0.041). CONCLUSION This study presents the long-term real-world results for persistence of GLM and its potential determinants. These most recent and long-term observations demonstrated that GLM and other bDMARDs continue to benefit patients with RA in Japan.
Collapse
Affiliation(s)
- Masahiko Miyashiro
- Medical Affairs Division, Medical Science Liaison Department, Janssen Pharmaceutical K.K., Nishi Kanda 3-5-2, Chiyoda-Ku, Tokyo, 101-0065, Japan. .,Medical Affairs Division, Immunology & Infectious Diseases Department, Janssen Pharmaceutical K.K., Nishi Kanda 3-5-2, Chiyoda-Ku, Tokyo, 101-0065, Japan.
| | - Yutaka Ishii
- Medical Affairs Division, Immunology & Infectious Diseases Department, Janssen Pharmaceutical K.K., Nishi Kanda 3-5-2, Chiyoda-Ku, Tokyo, 101-0065, Japan
| | - Celine Miyazaki
- Value, Evidence & Access Department, Janssen Pharmaceutical K.K., Nishi Kanda 3-5-2, Chiyoda-Ku, Tokyo, 101-0065, Japan
| | - Hirohito Shimizu
- Medical Affairs Division, Medical Science Liaison Department, Janssen Pharmaceutical K.K., Nishi Kanda 3-5-2, Chiyoda-Ku, Tokyo, 101-0065, Japan
| | - Junya Masuda
- Medical Affairs Division, Immunology & Infectious Diseases Department, Janssen Pharmaceutical K.K., Nishi Kanda 3-5-2, Chiyoda-Ku, Tokyo, 101-0065, Japan
| |
Collapse
|
8
|
Xiong H, Meng F, Luo M, Chen W, Tian J, Chen L, Ju Y, Mei Z. Anti-inflammatory and osteoprotective effects of Shi-Wei-Ru-Xiang pills on collagen-induced arthritis in rats via inhibiting MAPK and STAT3 pathways. JOURNAL OF ETHNOPHARMACOLOGY 2023; 300:115693. [PMID: 36075272 DOI: 10.1016/j.jep.2022.115693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/15/2022] [Accepted: 08/31/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shi-Wei-Ru-Xiang pills (SW) as a tradition Tibetan medicine has been clinically proved effective in rheumatoid arthritis (RA) treatment. However, the underlying mechanism of SW remains unclear. AIM OF THE STUDY This study aimed to investigate the anti-arthritic effect of SW and its possible mechanisms of action. MATERIALS AND METHODS A CIA rat model in vivo, and IL-1β-stimulated synoviocytes or chondrocytes and a co-culture system (IL-1β-stimulated synoviocytes/chondrocytes) in vitro were used to evaluate the effects of SW on the treatment of RA. Arthritic score, paw swelling rate, hematoxylin-eosin (HE) staining, and Safranin-O-Fast green (S-O) staining were used to evaluate the anti-arthritic activity of SW in CIA rats. TUNEL assay or flow cytometry were performed to measure chondrocytes apoptosis in vivo and invitro. The effects of SW on the expression and production of pro-inflammatory cytokines were assessed by qRT-PCR and Elisa. The inhibitory effects of SW on the phosphorylation of p38, Erk1/2, and STAT3 were analyzed by Western blot. RESULTS SW treatment significantly alleviated paw swelling, severity of arthritic and cartilage destruction in CIA rats. Moreover, SW decreased the expression of mRNAs of proinflammatory cytokines including TNF-α, IL-1β and IL-6 in the synovium, suppressed the production of these pro-inflammatory cytokines in serum and hind paws, downregulated the protein expression of p-p38, p-Erk1/2 and p-STAT3, and protected the chondrocytes apoptosis in CIA rats. Consistent with the results in vivo, SW also inhibited the activation of MAPK and STAT3 pathways, suppressed the expression of pro-inflammatory cytokines in IL-1β-stimulated synoviocytes, and attenuated chondrocytes apoptosis in IL-1β-stimulated chondrocytes. In the co-culture system, SW pre-treatment in IL-1β-stimulated synoviocytes exhibited inhibition of chondrocytes apoptosis, which was associated with attenuation of inflammation in synoviocytes. CONCLUSION These results suggested that the underlying mechanisms by which SW exerts its anti-arthritis effect may be related to the reduction of proinflammatory cytokine levels, inhibition of p38, Erk1/2 and STAT3 phosphorylation, and attenuating of chondrocyte apoptosis.
Collapse
Affiliation(s)
- Hui Xiong
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Fengping Meng
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Miao Luo
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Weiwu Chen
- Tibet Qizheng Tibetan Medicine Co.Ltd., Lasa, 850030, China
| | - Juan Tian
- Tibet Qizheng Tibetan Medicine Co.Ltd., Lasa, 850030, China
| | - Lunju Chen
- Tibet Qizheng Tibetan Medicine Co.Ltd., Lasa, 850030, China
| | - Yankun Ju
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Zhinan Mei
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China; College of Plant Science & Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
9
|
A national, multicenter, secondary data use study evaluating efficacy and retention of first-line biologic treatment with tocilizumab in patients with rheumatoid arthritis in real-life setting: results from TURKBIO registry. Sci Rep 2022; 12:21972. [PMID: 36539458 PMCID: PMC9767915 DOI: 10.1038/s41598-022-26106-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Tocilizumab (TCZ) is a recombinant humanized monoclonal antibody that targets the IL-6 receptor. TCZ found to be efficacious and has a good tolerated safety profile in rheumatoid arthritis (RA) patients. The aim of this study was to describe the disease activity and retention rate in Turkish RA patients who were prescribed TCZ as first-line biologic treatment in a real-world setting. Secondary data obtained from adult RA patients' files was used in a multicenter and retrospective context. Clinical Disease Activity Index (CDAI), Disease Activity Score in 28 joints with ESR (DAS28-ESR), and retention rates of TCZ were evaluated at related time points. 130 patients (87.7% female) with a mean age of 53 years (SD; 15.0) were included in the study. Mean RA duration was 14 years and median duration of follow-up was 18.5 months. Number of patients with ongoing TCZ treatment at 6, 12, and 24 months were 121 (93%), 85 (65%), and 46 (35%), respectively. Remission rates at 6, 12, and 24 months per CDAI (< 2.8) and DAS28-ESR (< 2.6) scores were 61.5, 44.6, 30%, and 54.6, 40.8, 27.7%, respectively. Both CDAI and DAS28-ESR scores significantly improved at 6, 12 and 24 months (p < 0.001 for both). At 24 months, 23 patients (17.6%) discontinued TCZ, of whom majority (17/23) were due to unsatisfactory response. Retention rates of TCZ at 6, 12, and 24 months were 93, 84.3, and 72.2%, respectively. In this real-world study, TCZ as a first-line biologic therapy was found to be efficacious and showing high retention rates. These real-world study results are in line with previous randomized studies.
Collapse
|
10
|
Zhang L, Qin Z, Sun H, Chen X, Dong J, Shen S, Zheng L, Gu N, Jiang Q. Nanoenzyme engineered neutrophil-derived exosomes attenuate joint injury in advanced rheumatoid arthritis via regulating inflammatory environment. Bioact Mater 2022; 18:1-14. [PMID: 35387158 PMCID: PMC8961303 DOI: 10.1016/j.bioactmat.2022.02.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by synovitis and destruction of cartilage, promoted by sustained inflammation. However, current treatments remain unsatisfactory due to lacking of selective and effective strategies for alleviating inflammatory environments in RA joint. Inspired by neutrophil chemotaxis for inflammatory region, we therefore developed neutrophil-derived exosomes functionalized with sub-5 nm ultrasmall Prussian blue nanoparticles (uPB-Exo) via click chemistry, inheriting neutrophil-targeted biological molecules and owning excellent anti-inflammatory properties. uPB-Exo can selectively accumulate in activated fibroblast-like synoviocytes, subsequently neutralizing pro-inflammatory factors, scavenging reactive oxygen species, and alleviating inflammatory stress. In addition, uPB-Exo effectively targeted to inflammatory synovitis, penetrated deeply into the cartilage and real-time visualized inflamed joint through MRI system, leading to precise diagnosis of RA in vivo with high sensitivity and specificity. Particularly, uPB-Exo induced a cascade of anti-inflammatory events via Th17/Treg cell balance regulation, thereby significantly ameliorating joint damage. Therefore, nanoenzyme functionalized exosomes hold the great potential for enhanced treatment of RA in clinic. uPB-Exo were firstly developed by combining NE-Exo with sub-5 nm ultrasmall PB nanoparticles via click chemistry. uPB-Exo selectively targeted inflamed joints via neutrophil-targeted biological molecules inherited from neutrophils. uPB-Exo accumulated in active FLS, and eventually scavenged reactive oxygen species and alleviated inflammatory stress. uPB-Exo induced a cascade of anti-inflammatory events via Th17/Treg cell balance regulation, thereby significantly ameliorating joint damage. uPB-Exo, as a drug free therapeutical agent, holds the great potential for enhanced treatment of RA in clinic.
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Ziguo Qin
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Han Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Xiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Jian Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Siyu Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Liming Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
- Corresponding author.
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
- Corresponding author. State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| |
Collapse
|
11
|
Alcaraz MJ, Guillén MI. Cellular and Molecular Targets of Extracellular Vesicles from Mesenchymal Stem/Stromal Cells in Rheumatoid Arthritis. Stem Cells Transl Med 2022; 11:1177-1185. [PMID: 36318277 PMCID: PMC9801303 DOI: 10.1093/stcltm/szac075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes progressive joint destruction. Despite the advances in the treatment of this condition there remains a clinical need for safe therapies leading to clinical remission. Mesenchymal stem/stromal cells (MSCs) play immunomodulatory and regenerative roles which can be partly mediated by their secretome. In recent years, the important contribution of extracellular vesicles (EVs) to MSC actions has received an increasing interest as a new therapeutic approach. We provide an extensive overview of the immunomodulatory properties of MSC EVs and their effects on articular cells such as fibroblast-like synoviocytes that play a central role in joint destruction. This review discusses the anti-arthritic effects of MSC EVs in vitro and in animal models of RA as well as their potential mechanisms. Recent preclinical data suggest that transfer of non-coding RNAs by MSC EVs regulates key signaling pathways involved in the pathogenesis of RA. We also examine a number of EV modifications for improving their anti-arthritic efficacy and carrier ability for drug delivery.
Collapse
Affiliation(s)
- María José Alcaraz
- Corresponding author: María José Alcaraz, PhD, Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Burjassot, Valencia, Spain. E-mail:
| | - María Isabel Guillén
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Av. Vicent A. Estellés s/n, Burjassot, Valencia, Spain,Department of Pharmacy, Faculty of Health Sciences, Cardenal Herrera-CEU University, Alfara del Patriarca, Valencia, Spain
| |
Collapse
|
12
|
miRNA-Mediated Epigenetic Regulation of Treatment Response in RA Patients—A Systematic Review. Int J Mol Sci 2022; 23:ijms232112989. [PMID: 36361779 PMCID: PMC9657910 DOI: 10.3390/ijms232112989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022] Open
Abstract
This study aimed to evaluate the role of microRNAs (miRNA) as biomarkers of treatment response in rheumatoid arthritis (RA) patients through a systematic review of the literature. The MEDLINE and Embase databases were searched for studies including RA-diagnosed patients treated with disease-modifying antirheumatic drugs (DMARDs) that identify miRNAs as response predictors. Review inclusion criteria were met by 10 studies. The main outcome of the study was the response to treatment, defined according to EULAR criteria. A total of 839 RA patients and 67 healthy donors were included in the selected studies. RA patients presented seropositivity for the rheumatoid factor of 74.7% and anti-citrullinated C-peptide antibodies of 63.6%. After revision, 15 miRNAs were described as treatment response biomarkers for methotrexate, anti-tumour necrosis factor (TNF), and rituximab. Among treatments, methotrexate presented the highest number of predictor miRNAs: miR-16, miR-22, miR-132, miR-146a and miR-155. The most polyvalent miRNAs were miR-146a, predicting response to methotrexate and anti-TNF, and miR-125b, which predicts response to infliximab and rituximab. Our data support the role of miRNAs as biomarkers of treatment response in RA and point to DMARDs modifying the miRNAs expression. Nevertheless, further studies are needed since a meta-analysis that allows definitive conclusions is not possible due to the lack of studies in this field.
Collapse
|
13
|
Malange KF, Navia-Pelaez JM, Dias EV, Lemes JBP, Choi SH, Dos Santos GG, Yaksh TL, Corr M. Macrophages and glial cells: Innate immune drivers of inflammatory arthritic pain perception from peripheral joints to the central nervous system. FRONTIERS IN PAIN RESEARCH 2022; 3:1018800. [PMID: 36387416 PMCID: PMC9644179 DOI: 10.3389/fpain.2022.1018800] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/03/2022] [Indexed: 07/22/2023] Open
Abstract
Millions of people suffer from arthritis worldwide, consistently struggling with daily activities due to debilitating pain evoked by this disease. Perhaps the most intensively investigated type of inflammatory arthritis is rheumatoid arthritis (RA), where, despite considerable advances in research and clinical management, gaps regarding the neuroimmune interactions that guide inflammation and chronic pain in this disease remain to be clarified. The pain and inflammation associated with arthritis are not isolated to the joints, and inflammatory mechanisms induced by different immune and glial cells in other tissues may affect the development of chronic pain that results from the disease. This review aims to provide an overview of the state-of-the-art research on the roles that innate immune, and glial cells play in the onset and maintenance of arthritis-associated pain, reviewing nociceptive pathways from the joint through the dorsal root ganglion, spinal circuits, and different structures in the brain. We will focus on the cellular mechanisms related to neuroinflammation and pain, and treatments targeting these mechanisms from the periphery and the CNS. A comprehensive understanding of the role these cells play in peripheral inflammation and initiation of pain and the central pathways in the spinal cord and brain will facilitate identifying new targets and pathways to aide in developing therapeutic strategies to treat joint pain associated with RA.
Collapse
Affiliation(s)
- Kaue Franco Malange
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | | | - Elayne Vieira Dias
- Department of Neurology, University of California, San Francisco, CA, United States
| | | | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, CA, United States
| | | | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, CA, United States
| |
Collapse
|
14
|
Jo HG, Seo J, Lee D. Clinical evidence construction of East Asian herbal medicine for inflammatory pain in rheumatoid arthritis based on integrative data mining approach. Pharmacol Res 2022; 185:106460. [PMID: 36152738 DOI: 10.1016/j.phrs.2022.106460] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to a significant social burden. East Asian herbal medicine (EAHM) has long been used to treat RA. Therefore, a systematic study of how EAHM treatments can be developed into new drugs using specific materials is needed. METHODS Eleven databases containing literature in English, Korean, Chinese, and Japanese were searched for randomized controlled trials comparing EAHM with conventional medicine (CM). A meta-analysis was performed on the variable data to assess their effects on inflammatory pain. Subsequently, we searched for core materials and combinations of core material-based data mining methods. RESULTS A total of 186 trials involving 19,716 patients with RA met the inclusion criteria. According to the meta-analysis, EAHM had a significantly superior effect on continuous pain intensity, tender joint count, and response rate. Patients treated with EAHM had a significantly reduced incidence of adverse events compared with those treated with CM. Based on additional analysis of the EAHM formula data included in this meta-analysis, 21 core materials and five core herbal combinations were identified. CONCLUSION EAHM remedies for RA have the adequate potential for use as candidate materials for treating inflammatory pain in RA. The candidate core herbs evaluated in this study act on multiple pathways and are expected to provide pain relief, sustained inflammation suppression, immune regulation, and prevention of joint destruction. It seems worthwhile to conduct follow-up research on drug development using the core materials derived from this review.
Collapse
Affiliation(s)
- Hee-Geun Jo
- BS Healthcare Co., Ltd., 11 Teheran-ro 33-gil, Gangnam-gu, Seoul 06141, Republic of Korea; Allbarun Kyunghee Korean Medicine Clinic, 18, Pungmu-ro 146-gil, Gimpo, Gyeonggi-do, Republic of Korea; Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam 13120, Republic of Korea.
| | - Jihye Seo
- BS Healthcare Co., Ltd., 11 Teheran-ro 33-gil, Gangnam-gu, Seoul 06141, Republic of Korea; Allbarun Kyunghee Korean Medicine Clinic, 18, Pungmu-ro 146-gil, Gimpo, Gyeonggi-do, Republic of Korea; Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam 13120, Republic of Korea
| | - Donghun Lee
- BS Healthcare Co., Ltd., 11 Teheran-ro 33-gil, Gangnam-gu, Seoul 06141, Republic of Korea; Allbarun Kyunghee Korean Medicine Clinic, 18, Pungmu-ro 146-gil, Gimpo, Gyeonggi-do, Republic of Korea; Department of Herbal Pharmacology, College of Korean Medicine, Gachon University, 1342 Seongnamdae-ro, Sujeong-gu, Seongnam 13120, Republic of Korea.
| |
Collapse
|
15
|
Caminade AM, Turrin CO, Poupot R. Curing inflammatory diseases using phosphorous dendrimers. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1783. [PMID: 35194953 DOI: 10.1002/wnan.1783] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 06/14/2023]
Abstract
Different types of water-soluble phosphorous dendrimers have been synthesized and display many different biological properties. It has been shown in particular that phosphorous dendrimers of first generation functionalized with azabisphosphonate terminal functions are able to stimulate the human immune system ex vivo. These dendrimers are internalized by monocytes within a few seconds, and induce their anti-inflammatory activation. The presence of the dendrimers induces also the inhibition of the differentiation of monocytes into osteoclasts, the maturation of dendritic cells, and inhibits the proliferation of the proinflammatory CD4+ T lymphocytes. Finally, after 2-3 weeks of culture of peripheral blood mononuclear cells, amplifications by several tens of natural killer cells is observed. In view of all these properties, the influence of these azabisphosphonate-dendrimers has been tested in vivo with several animal models, against different chronic or acute inflammatory diseases, such as multiple sclerosis, rheumatoid arthritis, uveitis, and psoriasis, but also against myeloid leukemia, a hematological cancer. The hematological safety has been demonstrated in mice, as there is no platelet aggregation, no hemolysis, and no disturbance in the hematological formula. The safety of the azabisphosphonate-dendrimer has been assessed also with non-human primates (cynomolgus monkeys) which received repeated injections, as a de-risking pre-clinical test. Biochemical, hematological, and all immunological parameters in peripheral blood remained within a normal physiological range throughout the study, and all survived well. Other phosphorous dendrimers also display anti-inflammatory properties in vivo, in particular dendrimers functionalized with mannose derivatives, which prevent acute lung diseases when given orally (per os) to mice. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Anne-Marie Caminade
- Laboratoire de Chimie de Coordination (LCC), CNRS UPR8241, Toulouse Cedex 4, France
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Cédric-Olivier Turrin
- Laboratoire de Chimie de Coordination (LCC), CNRS UPR8241, Toulouse Cedex 4, France
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse, France
- IMD-Pharma, Toulouse Cedex 4, France
| | - Rémy Poupot
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, CHU Purpan, Toulouse Cedex 3, France
- Infinity, Université Toulouse, CNRS, INSERM, UPS, Toulouse, France
| |
Collapse
|
16
|
Tahmasian H, Smeele HTW, de Jong PHP, Dolhain RJEM, van Mulligen E. Biological and Methotrexate Survival after Pregnancy in Patients With a Rheumatic Disease. Front Pharmacol 2022; 13:826034. [PMID: 35355725 PMCID: PMC8959570 DOI: 10.3389/fphar.2022.826034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/31/2022] [Indexed: 11/23/2022] Open
Abstract
Objective: Patients with a rheumatic disease who discontinue their disease-modifying anti-rheumatic drug (DMARD) due to pregnancy often wonder if treatment will be as effective after pregnancy. This study investigates the effect of a temporary discontinuation of DMARDs due to pregnancy on the effectiveness of the same DMARD postpartum in patients with a rheumatic disease. Methods: Pregnant, rheumatic patients were derived from the Preconceptional Counseling in Active Rheumatoid Arthritis (PreCARA) cohort. DMARD-survival after pregnancy, for biological and methotrexate (MTX) therapy, was analyzed and compared to controls with stable DMARD-treatment from a retrospective cohort. Results: In total, 234 patients were included, of whom 114 patients had stable biological or MTX treatment before their pregnancy. After pregnancy, 40 out of 56 (71%) patients restarted their biological, for MTX this was 49%. One year after restart, and censoring for a following pregnancy, 88.9% of patients were still using their biological, and 85% still used their MTX (p = 0.92). Compared to the matched controls the survival after pregnancy was significantly lower 1 year after restart for both biologicals (98.3%) and MTX (99.6%); p = 0.002 and p < 0.001 respectively; 3 years after restart this significant difference was no longer observed (p = 0.50 and p = 0.33, respectively). Conclusion: Effective DMARD (biological or MTX) treatment before pregnancy that was discontinued due to pregnancy seems effective after pregnancy. Although DMARD-survival was higher in the control group 1 year after restart, the percentage of patients with effective treatment was still very good (>85%). In addition, this difference was no longer observed after 3 years.
Collapse
Affiliation(s)
- Helena Tahmasian
- Department of Rheumatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Hieronymus T W Smeele
- Department of Rheumatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Pascal H P de Jong
- Department of Rheumatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Radboud J E M Dolhain
- Department of Rheumatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Elise van Mulligen
- Department of Rheumatology, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
17
|
Zhuo J, Xia Q, Sharma N, Gao S, Lama S, Cui J, Feathers V, Shadick N, Weinblatt ME. The Role of Shared Epitope in Rheumatoid Arthritis Prognosis in Relation to Anti-Citrullinated Protein Antibody Positivity. Rheumatol Ther 2022; 9:637-647. [PMID: 35174462 PMCID: PMC8964857 DOI: 10.1007/s40744-022-00427-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/20/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction Shared epitope (SE) is present in high proportions of anti-citrullinated protein antibody (ACPA) + patients with rheumatoid arthritis (RA) and is associated with poor prognosis. We assessed the role of SE in RA prognosis, in relation to ACPA positivity. Methods Patients enrolled in the Brigham and Women’s RA Sequential Study were included. Changes from baseline in disease activity (Disease Activity Score in 28 joints using C-reactive protein [DAS28 (CRP)], Clinical Disease Activity Index [CDAI], Simplified Disease Activity Index [SDAI]) to 1 year were assessed. Baseline characteristics were compared by SE and ACPA status (±; chi-squared, Kruskal-Wallis). Association between number of SE alleles and ACPA status (logistic regression models), relationships between baseline characteristics and changes in disease activity (adjusted linear regression model), and effect of ACPA on the association between SE and changes in disease activity (mediation analysis) were studied. Results Nine hundred twenty-six patients were included. SE + versus SE − patients had significantly longer disease duration and higher disease activity scores and were more likely to have erosive disease, have higher comorbidity burden, and be RF + (all p < 0.05). Among patients with one or two SE alleles (vs. 0), odds of being ACPA + were 1.97 (p = 0.0003) and 3.82 (p < 0.0001), respectively. SE + versus SE − patients had worse disease activity scores as indicated by mean increases in DAS28 (CRP) of 0.22, CDAI of 2.07, and SDAI of 2.43 over 1 year (all p < 0.05). Direct effect of SE + accounted for 76.4–80.1% of total effect in disease activity increases. Conclusions SE is strongly associated with ACPA positivity and higher disease activity in patients with RA. SE was associated with greater increases in disease activity over 1 year, which was partially mediated by the presence of ACPA. Trial Registration ClinicalTrials.gov identifier: NCT01793103; registration date: February 15, 2013, retrospectively registered. Patients with rheumatoid arthritis (RA) experience inflamed and damaged joints. RA is an autoimmune disease where proteins called autoantibodies in the blood of patients with RA target the patient’s own joint tissue and organs by mistake. This causes inflammation. Patients with certain autoantibodies, such as anti-citrullinated protein antibodies (ACPAs), may experience worse symptoms. There are certain genetic risk factors that may mean a person is more likely to develop RA. One example of a genetic risk factor is having the shared epitope (SE). Our study looked at almost 1000 patients with RA in the general population. It explored the impact of having SE and ACPAs on changes in RA disease activity. Patients with SE had RA for a longer time, had more severe disease, and were more likely to have other diseases compared with patients without SE. Patients with SE were also more likely to have ACPAs. Over the course of one year, patients with SE had larger increases in RA disease activity than those patients without SE, even though they were taking the same treatments. These results suggest that patients with the genetic risk factor, SE often have RA that is harder to treat. Doctors should take this into account when selecting treatment for RA.
Collapse
Affiliation(s)
- Joe Zhuo
- Bristol Myers Squibb, Princeton, NJ, USA.
| | - Qian Xia
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | - Sheng Gao
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Sonie Lama
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Jing Cui
- Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Vivi Feathers
- Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy Shadick
- Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | | |
Collapse
|
18
|
Wang Z, Huang J, Xie D, He D, Lu A, Liang C. Toward Overcoming Treatment Failure in Rheumatoid Arthritis. Front Immunol 2021; 12:755844. [PMID: 35003068 PMCID: PMC8732378 DOI: 10.3389/fimmu.2021.755844] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder characterized by inflammation and bone erosion. The exact mechanism of RA is still unknown, but various immune cytokines, signaling pathways and effector cells are involved. Disease-modifying antirheumatic drugs (DMARDs) are commonly used in RA treatment and classified into different categories. Nevertheless, RA treatment is based on a "trial-and-error" approach, and a substantial proportion of patients show failed therapy for each DMARD. Over the past decades, great efforts have been made to overcome treatment failure, including identification of biomarkers, exploration of the reasons for loss of efficacy, development of sequential or combinational DMARDs strategies and approval of new DMARDs. Here, we summarize these efforts, which would provide valuable insights for accurate RA clinical medication. While gratifying, researchers realize that these efforts are still far from enough to recommend specific DMARDs for individual patients. Precision medicine is an emerging medical model that proposes a highly individualized and tailored approach for disease management. In this review, we also discuss the potential of precision medicine for overcoming RA treatment failure, with the introduction of various cutting-edge technologies and big data.
Collapse
Affiliation(s)
- Zhuqian Wang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Jie Huang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Duoli Xie
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Dongyi He
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Chao Liang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
19
|
Bhushan V, Lester S, Briggs L, Hijjawi R, Shanahan EM, Pontifex E, Ninan J, Hill C, Cai F, Walker J, Goldblatt F, Wechalekar MD. Real-Life Retention Rates and Reasons for Switching of Biological DMARDs in Rheumatoid Arthritis, Psoriatic Arthritis, and Ankylosing Spondylitis. Front Med (Lausanne) 2021; 8:708168. [PMID: 34646840 PMCID: PMC8502861 DOI: 10.3389/fmed.2021.708168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/29/2021] [Indexed: 11/13/2022] Open
Abstract
Aims: To determine real-life biologic/targeted synthetic disease-modifying anti-rheumatic drug (b/tsDMARD) retention rates in rheumatoid arthritis (RA), psoriatic arthritis (PsA), and ankylosing spondylitis (AS), explore reasons for switching and to compare results to previously published data. Methods: Time-to-event analysis for mean treatment duration (estimated as the Restricted Mean Survival Time), b/tsDMARD failure, and b/tsDMARDs switching was performed for 230 patients (n = 147 RA, 46 PsA, 37 AS) who commenced their first b/tsDMARD between 2008 and 2018. Patients were managed in a dedicated “biologics” clinic in a tertiary hospital; the choice of b/tsDMARD was clinician driven based on medical factors and patient preferences. The effect of covariates on switching risk was analysed by a conditional risk-set Cox proportional-hazards model. Treatment retention data was compared to a historical analysis (2002–2008). Results: The proportions remaining on treatment (retention) were similar, throughout follow-up, for the first, second and third b/tsDMARDs across all patients (p = 0.46). When compared to RA patients, the risk of b/tsDMARD failure was halved in PsA patients [Hazard Ratio (HR) = 0.50], but no different in AS patients (HR = 1.0). The respective restricted mean (95%CI) treatment durations, estimated at 5 years of follow-up, were 3.1 (2.9, 3.4), 4.1 (3.7, 4.6), and 3.3 (2.8, 3.9) years, for RA, PsA, and AS, respectively. Age, gender, disease duration, smoking status and the use of concomitant csDMARDS were not associated with the risk of bDMARD failure. The most common reasons for switching in the first and subsequent years were secondary (n = 62) and primary (n = 35) failure. Comparison with historical data indicated no substantive differences in switching of the first biologic for RA and PsA. Conclusion: Similar retention rates of the second and third compared to the first b/tsDMARD in RA, PsA, and AS support a strategy of differential b/tsDMARDs use informed by patient presentation. Despite greater availability of b/tsDMARDs with differing mechanisms of action, retention rates of the first b/tsDMARD remain similar to previous years.
Collapse
Affiliation(s)
- Vandana Bhushan
- Rheumatology Unit, Flinders Medical Centre, Adelaide, SA, Australia.,Division of Medicine, Flinders Medical Centre, Adelaide, SA, Australia
| | - Susan Lester
- Rheumatology Unit, Queen Elizabeth Hospital, Adelaide, SA, Australia.,Discipline of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Liz Briggs
- Rheumatology Unit, Flinders Medical Centre, Adelaide, SA, Australia
| | - Raif Hijjawi
- Division of Medicine, Flinders Medical Centre, Adelaide, SA, Australia
| | - E Michael Shanahan
- Rheumatology Unit, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Eliza Pontifex
- Rheumatology Unit, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Jem Ninan
- Rheumatology Unit, Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Catherine Hill
- Rheumatology Unit, Queen Elizabeth Hospital, Adelaide, SA, Australia.,Discipline of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Fin Cai
- Rheumatology Unit, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Jennifer Walker
- Rheumatology Unit, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Fiona Goldblatt
- Rheumatology Unit, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Mihir D Wechalekar
- Rheumatology Unit, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
20
|
Ba X, Huang Y, Shen P, Huang Y, Wang H, Han L, Lin WJ, Yan HJ, Xu LJ, Qin K, Chen Z, Tu SH. WTD Attenuating Rheumatoid Arthritis via Suppressing Angiogenesis and Modulating the PI3K/AKT/mTOR/HIF-1α Pathway. Front Pharmacol 2021; 12:696802. [PMID: 34646130 PMCID: PMC8502817 DOI: 10.3389/fphar.2021.696802] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/24/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Wutou Decoction (WTD), as a classic prescription, has been generally used to treat rheumatoid arthritis (RA) for two thousand years in China. However, the potential protective effects of WTD on rheumatoid arthritis and its possible mechanism have rarely been reported. Purpose: The aim of this study was to explore the possible mechanism of WTD against RA and a promising alternative candidate for RA therapy. Methods: A model of collagen-induced arthritis (CIA) was constructed in rats to assess the therapeutic effects of WTD. Histopathological staining, immunofluorescence, and western blotting of synovial sections were conducted to detect the antiangiogenic effects of WTD. Then, cell viability assays, flow cytometry, scratch healing assays, and invasion assays were conducted to explore the effects of WTD on MH7A human fibroblast-like synoviocyte (FLS) cell proliferation, apoptosis, migration, and invasion in vitro. The ability of WTD to induce blood vessel formation after MH7A cell and human umbilical vein endothelial cell line (HUVEC) coculture with WTD intervention was detected by a tube formation assay. The mechanisms of WTD were screened by network pharmacology and confirmed by in vivo and in vitro experiments. Results: WTD ameliorated the symptoms and synovial pannus hyperplasia of CIA rats. Treatment with WTD inhibited MH7A cell proliferation, migration, and invasion and promoted MH7A apoptosis. WTD could inhibit MH7A cell expression of proangiogenic factors, including VEGF and ANGI, to induce HUVEC tube formation. Furthermore, the PI3K-AKT-mTOR-HIF-1α pathway was enriched as a potential target of WTD for the treatment of RA through network pharmacology enrichment analysis. Finally, it was confirmed in vitro and in vivo that WTD inhibits angiogenesis in RA by interrupting the PI3K-AKT-mTOR-HIF-1α pathway. Conclusion: WTD can inhibit synovial hyperplasia and angiogenesis, presumably by inhibiting the migration and invasion of MH7A cells and blocking the production of proangiogenic effectors in MH7A cells. The possible underlying mechanism by which WTD ameliorates angiogenesis in RA is the PI3K-AKT-mTOR-HIF-1α pathway.
Collapse
Affiliation(s)
- Xin Ba
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Shen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Wang
- Rehabilitation and Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Liang Han
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Ji Lin
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Jia Yan
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Jun Xu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Qin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Hao Tu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Sánchez-Piedra C, Sueiro-Delgado D, García-González J, Ros-Vilamajo I, Prior-Español A, Moreno-Ramos MJ, Garcia-Magallon B, Calvo-Gutiérrez J, Perez-Vera Y, Martín-Domenech R, Ruiz-Montesino D, Vela-Casasempere P, Expósito L, Sánchez-Alonso F, González-Davila E, Díaz-González F. Changes in the use patterns of bDMARDs in patients with rheumatic diseases over the past 13 years. Sci Rep 2021; 11:15051. [PMID: 34302036 PMCID: PMC8302725 DOI: 10.1038/s41598-021-94504-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
The better understanding of the safety of biologic DMARDs (bDMARDs), as well as the emergence of new bDMARDs against different therapeutic targets and biosimilars have likely influenced the use patterns of these compounds over time. The aim of this study is to assess changes in demographic characteristics, disease activity and treatment patterns in patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA), or ankylosing spondylitis (AS) who started a first- or second-line biologic between 2007 and mid-2020. Patients diagnosed with RA, PsA or AS included in the BIOBADASER registry from January 2007 to July 2020 were included. According to the start date of a first- or second-line biologic therapy, patients were stratified into four time periods: 2007-2009; 2010-2013; 2014-2017; 2018-2020 and analyzed cross-sectionally in each period. Demographic and clinical variables, as well as the type of biologic used, were assessed. Generalized linear models were applied to study the evolution of the variables of interest over time periods, the diagnosis, and the interactions between them. A total of 4543 patients initiated a first biologic during the entire time frame of the study. Over the four time periods, disease evolution at the time of biologic initiation (p < 0.001), disease activity (p < 0.001), retention rate (p < 0.001) and the use of tumor necrosis factor inhibitors as a first-line treatment (p < 0.001) showed a significant tendency to decrease. Conversely, comorbidities, as assessed by the Charlson index (p < 0.001), and the percentage of patients using bDMARDs in monotherapy (p < 0.001), and corticosteroids (p < 0.001) tended to increase over time. Over the entire period of the study's analysis, 3289 patients started a second biologic. The following trends were observed: decreased DAS28 at switching (p < 0.001), lower retention rates (p = 0.004), and incremental changes to the therapeutic target between the first and second biologic (p < 0.001). From 2007 until now rheumatic patients who started a biologic were older, exhibited less clinical activity, presented more comorbidities, and switched to a different biologic more frequently and earlier.
Collapse
Affiliation(s)
| | | | | | | | - Agueda Prior-Español
- Servicio de Reumatología, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | | | | | | | - Yanira Perez-Vera
- Servicio de Reumatología, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas, Spain
| | | | | | | | - Lorena Expósito
- Servicio de Reumatología, Hospital Universitario de Canarias, Calle Ofra s/n 38320, La Laguna, Santa Cruz de Tenerife, Spain
| | | | - Enrique González-Davila
- Departamento de Matemáticas, Estadística e Investigación Operativa, Universidad de La Laguna, Tenerife, Spain
| | - Federico Díaz-González
- Servicio de Reumatología, Hospital Universitario de Canarias, Calle Ofra s/n 38320, La Laguna, Santa Cruz de Tenerife, Spain.
- Departamento de Medicina Interna, Dermatología y Cirugía, Dermatología y Psiquiatría, Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
22
|
Prediction of response to Certolizumab-Pegol in rheumatoid arthritis (PreCePRA) by functional MRI of the brain - Study protocol for a randomized double-blind controlled study. Contemp Clin Trials Commun 2021; 22:100770. [PMID: 34041413 PMCID: PMC8144682 DOI: 10.1016/j.conctc.2021.100770] [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: 07/28/2020] [Revised: 03/09/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
Background Tumor necrosis factor inhibitors (TNFi) signify a major advance in the treatment of rheumatoid arthritis (RA). However, treatment success initially remains uncertain as approximately half of the patients do not respond adequately to TNFi. Thus, an unmet need exists to better predict therapeutic outcome of biologicals. Objectives We investigated whether brain activity associated with arthritis measured by functional magnetic resonance imaging (fMRI) of the brain can serve as a predictor of response to TNFi in RA patients. Methods PreCePRA is a multi-center, randomized, double-blind, placebo-controlled fMRI trial on patients with RA [1] [2]. Active RA patients failing csDMARDs therapy with a DAS28 > 3.2 and at least three tender and/or swollen joints underwent a brain BOLD (blood-oxygen-level dependent) fMRI scan upon joint compression at screening. Patients were then randomized into a 12-week double-blinded treatment phase with 200 mg Certolizumab Pegol (CZP) every two weeks (arm 1: fMRI BOLD signal activated volume > 2000 voxel, i.e. 2 cm3; arm 2: fMRI BOLD signal activated volume <2000 voxel) or placebo (arm 3). DAS28 low disease activity at 12 weeks was assigned as primary endpoint. A 12-week follow-up phase in which patients were switched from the placebo to the treatment arm followed the blinded phase. fMRI was carried out at screening as well as after 12 and 24 weeks of receiving CZP or placebo. Conclusion We hypothesize that high-level central nervous representation of pain in patients with rheumatoid arthritis predicts response to the TNFi CZP which we further investigate in the PreCePRA trial.
Collapse
|
23
|
Human umbilical cord mesenchymal stem cell-derived small extracellular vesicles ameliorate collagen-induced arthritis via immunomodulatory T lymphocytes. Mol Immunol 2021; 135:36-44. [PMID: 33857817 DOI: 10.1016/j.molimm.2021.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/03/2020] [Accepted: 04/04/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease for which there are currently no effective therapies. Although mesenchymal stem cells (MSCs) can prevent arthritis through immunomodulatory mechanisms, there are several associated risks. Alternatively, MSC-derived small extracellular vesicles (sEVs) can mimic the effects of MSCs, while reducing the risk of adverse events. However, few studies have examined sEVs in the context of RA. Here, we evaluate the immunomodulatory effects of human umbilical cord MSC (hUCMSC)-derived sEVs on T lymphocytes in a collagen-induced arthritis (CIA) rat model to elucidate the possible mechanism of sEVs in RA treatment. We then compare these mechanisms to those of MSCs and methotrexate (MTX). METHODS The arthritis index and synovial pathology were assessed. T lymphocyte proliferation and apoptosis, Th17 and Treg proportions, and interleukin (IL)-17, IL-10, and transforming growth factor (TGF)-β expression were detected using flow cytometry. Retinoic acid receptor-related orphan receptor gamma t (RORγt) and forkhead box P3 (FOXP3), which are master transcriptional regulators of Th17 and Treg differentiation, were also assessed using immunohistochemistry and reverse transcription-polymerase chain reaction (RT-PCR). RESULTS sEV treatment ameliorated arthritis and inhibited synovial hyperplasia in a dose-dependent manner. These effects were mediated by inhibiting T lymphocyte proliferation and promoting their apoptosis, while decreasing Th17 cell proportion and increasing that of Treg cells in the spleen, resulting in decreased serum IL-17, and enhanced IL-10 and TGF-β expression. Transcriptionally, sEVs decreased RORγt and increased FOXP3 expression in the spleen, and decreased RORγt and FOXP3 expression in the joints. In some aspects sEVs were more effective than MSCs and MTX in treating CIA. CONCLUSIONS hUCMSC-derived sEVs ameliorate CIA via immunomodulatory T lymphocytes, and might serve as a new therapy for RA.
Collapse
|
24
|
Migliore A, Pompilio G, Integlia D, Zhuo J, Alemao E. Cycling of tumor necrosis factor inhibitors versus switching to different mechanism of action therapy in rheumatoid arthritis patients with inadequate response to tumor necrosis factor inhibitors: a Bayesian network meta-analysis. Ther Adv Musculoskelet Dis 2021; 13:1759720X211002682. [PMID: 33854570 PMCID: PMC8010806 DOI: 10.1177/1759720x211002682] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 02/15/2021] [Indexed: 11/17/2022] Open
Abstract
Introduction: For patients with rheumatoid arthritis (RA) with an inadequate response to tumor necrosis factor inhibitors (TNFi), main options include cycling onto a different TNFi or switching to a biologic/targeted synthetic disease-modifying antirheumatic drug with a different mechanism of action (MOA). This network meta-analysis (NMA) assessed comparative clinical efficacy of cycling versus switching. Methods: We conducted a literature search in MEDLINE, Embase, and Cochrane Library. Outcomes included proportion of patients with 20%, 50%, or 70% response to American College of Rheumatology criteria (ACR20/ACR50/ACR70 response), Disease Activity Score in 28 joints (DAS28) score below 2.6 or between 2.6 and 3.2, mean change in DAS28 score, mean reduction in and proportion of patients achieving a clinically meaningful reduction (⩾0.22) in Health Assessment Questionnaire score, number of serious adverse events (AEs), and withdrawals for any reason/due to AEs/lack of treatment efficacy. To account for the wide range of study populations and designs, we developed three models to conduct the NMA: fixed-effect, random-effects, and hierarchical Bayesian. PROSPERO ID: CRD42019122993. Results: We identified nine randomized controlled trials and 16 observational studies. The fixed-effect model suggested a 0.99 probability that switch was the better strategy for increasing odds of a clinically meaningful improvement in ACR50 [odds ratio (OR): 1.35 (95% credible interval (CI): 0.96–1.81)]. The fixed-effect model also suggested that switch was associated with lower rates of withdrawal for any reasons [OR: 0.53 (95% CI: 0.40–0.68)]. The random-effects and hierarchical Bayesian models suggested additional uncertainty as they considered more variability than the fixed-effect model. Discussion: Results suggest that switching to a drug with a different MOA is more effective and associated with lower rates of withdrawal than cycling to a different TNFi after failure of first-line TNFi. Further trials that directly compare cycling with switching are warranted to better assess comparative efficacy. Plain language summary Assessment of the effectiveness of different drug treatment strategies in patients with rheumatoid arthritis: an analysis of the published literature
Collapse
Affiliation(s)
- Alberto Migliore
- Unit of Rheumatology, Ospedale S. Pietro Fatebenefratelli ISPOR Italy, Via Cassia 600, Rome, 00189, Italy
| | | | | | - Joe Zhuo
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, Princeton, NJ, USA
| | - Evo Alemao
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
25
|
Eddaoudi M, Rostom S, Hmamouchi I, Binoune IE, Amine B, Abouqal R, Achemlal L, Allali F, Bouchti IE, Maghraoui AE, Ghozlani I, Hassikou H, Harzy T, Ichchou L, Mkinsi O, Niamane R, Bahiri R. The first biological choice in patients with rheumatoid arthritis: data from the Moroccan register of biotherapies. Pan Afr Med J 2021; 38:183. [PMID: 33995789 PMCID: PMC8106777 DOI: 10.11604/pamj.2021.38.183.27081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/05/2021] [Indexed: 12/29/2022] Open
Abstract
Introduction the aim of our study is to determine, from data of the Moroccan register of biotherapies, the factors influencing the choice of the first prescribed biological treatment. Methods cross-sectional multicenter study including rheumatoid arthritis patients who were initiated the first biological treatment either: Rituximab, an anti-TNF, or Tocilizumab. The determinants related to the patient and disease have been gathered. A univariate and then multivariate analysis to determine the factors associated with the choice of the first bDMARDs was realized. Results a total of 225 rheumatoid arthritis patients were included in the Moroccan registry. The mean age was 52 ± 11 years, with female predominance 88% (n = 197). The first prescribed biological treatment was Rituximab 74% (n = 166), the second one was Tocilizumab, 13.6% (n = 31) then comes the anti-TNF in 3rd position with 12.4% (n = 28). The factors associated with the choice of Rituximab as the first line bDMARDs prescribed in univariate analysis were: the insurance type, the positivity of the rheumatoid factor. In multivariate analysis, only the insurance type that remains associated with the choice of Rituximab as the first biological drugs. The Tocilizumab was associated with shorter disease duration and was more prescribed as mono-therapy compared to non Tocilizumab group. TNFi was associated with the insurance type. Conclusion our study suggests that Rituximab and TNFi are associated with the type of insurance and Tocilizumab is the most prescribed biologic mono-therapy in RA patients. Further studies are needed to confirm these results.
Collapse
Affiliation(s)
- Meryem Eddaoudi
- Rheumatology Department A, El Ayachi Hospital, Ibn Sina University Hospital, Rabat-Sale, Morocco
| | - Samira Rostom
- Rheumatology Department A, El Ayachi Hospital, Ibn Sina University Hospital, Rabat-Sale, Morocco
| | - Ihsane Hmamouchi
- Laboratory of Epidemiology and Clinical Research, Faculty of Medicine and Pharmacy, Rabat, Morocco.,Provincial Hospital, Skhirat-Temara, Morocco
| | - Imane El Binoune
- Rheumatology Department A, El Ayachi Hospital, Ibn Sina University Hospital, Rabat-Sale, Morocco
| | - Bouchra Amine
- Rheumatology Department A, El Ayachi Hospital, Ibn Sina University Hospital, Rabat-Sale, Morocco
| | - Redouane Abouqal
- Laboratory of Epidemiology and Clinical Research, Faculty of Medicine and Pharmacy, Rabat, Morocco
| | - Lahsen Achemlal
- Rheumatology Department, Military Hospital Mohamed V, Ibn Sina University Hospital, Rabat, Morocco
| | - Fadoua Allali
- Rheumatology Department B, El Ayachi Hospital, Ibn Sina University Hospital, Rabat-Sale, Morocco
| | - Imane El Bouchti
- Rheumatology Department, Arrazi Hospital, University Hospital Mohammed VI, Marrakech, Morocco
| | | | - Imad Ghozlani
- Rheumatology Department, University Hospital, Agadir, Morocco
| | - Hasna Hassikou
- Rheumatology Department, Military Hospital, Hassan II University Hospital, Meknes, Morocco
| | - Taoufik Harzy
- Rheumatology Department, University Hospital Hassan II, Fez, Morocco
| | - Linda Ichchou
- Rheumatology Department, University Hospital Mohammed VI, Oujda, Morocco
| | - Ouafae Mkinsi
- Rheumatology Department, University Hospital Ibn Rochd, Casablanca, Morocco
| | - Redouane Niamane
- Rheumatology Department, Military Hospital Avicenne, University Hospital Mohammed VI, Marrakech, Morocco
| | - Rachid Bahiri
- Rheumatology Department A, El Ayachi Hospital, Ibn Sina University Hospital, Rabat-Sale, Morocco
| |
Collapse
|
26
|
Solomon DH, Xu C, Collins J, Kim SC, Losina E, Yau V, Johansson FD. The sequence of disease-modifying anti-rheumatic drugs: pathways to and predictors of tocilizumab monotherapy. Arthritis Res Ther 2021; 23:26. [PMID: 33446261 PMCID: PMC7807904 DOI: 10.1186/s13075-020-02408-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/26/2020] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND There are numerous non-biologic and biologic disease-modifying anti-rheumatic drugs (bDMARDs) for rheumatoid arthritis (RA). Typical sequences of bDMARDs are not clear. Future treatment policies and trials should be informed by quantitative estimates of current treatment practice. METHODS We used data from Corrona, a large real-world RA registry, to develop a method for quantifying sequential patterns in treatment with bDMARDs. As a proof of concept, we study patients who eventually use tocilizumab monotherapy (TCZm), an IL-6 antagonist with similar benefits used as monotherapy or in combination. Patients starting a bDMARD were included and were followed using a discrete-state Markov model, observing changes in treatments every 6 months and determining whether they used TCZm. A supervised machine learning algorithm was then employed to determine longitudinal patient factors associated with TCZm use. RESULTS 7300 patients starting a bDMARD were followed for up to 5 years. Their median age was 58 years, 78% were female, median disease duration was 5 years, and 57% were seropositive. During follow-up, 287 (3.9%) reported use of TCZm with median time until use of 25.6 (11.5, 56.0) months. Eighty-two percent of TCZm use began within 3 years of starting any bDMARD. Ninety-three percent of TCZm users switched from TCZ combination, a TNF inhibitor, or another bDMARD. Very few patients are given TCZm as their first DMARD (0.6%). Variables associated with the use of TCZm included prior use of TCZ combination therapy, older age, longer disease duration, seronegative, higher disease activity, and no prior use of a TNF inhibitor. CONCLUSIONS Improved understanding of treatment sequences in RA may help personalize care. These methods may help optimize treatment decisions using large-scale real-world data.
Collapse
Affiliation(s)
- Daniel H Solomon
- Division of Rheumatology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA. .,Division of Pharmacoepidemiology, Brigham and Women's Hospital, Boston, USA.
| | - Chang Xu
- Division of Rheumatology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Jamie Collins
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Boston, USA
| | - Seoyoung C Kim
- Division of Rheumatology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.,Division of Pharmacoepidemiology, Brigham and Women's Hospital, Boston, USA
| | - Elena Losina
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Boston, USA
| | - Vincent Yau
- Brigham and Women's Hospital, Genentech, San Francisco, California, USA
| | | |
Collapse
|
27
|
van Mulligen E, Ahmed S, Weel AEAM, Hazes JMW, van der Helm-van Mil AHM, de Jong PHP. Factors that influence biological survival in rheumatoid arthritis: results of a real-world academic cohort from the Netherlands. Clin Rheumatol 2021; 40:2177-2183. [PMID: 33415451 PMCID: PMC8121743 DOI: 10.1007/s10067-020-05567-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 11/28/2022]
Abstract
We aim to explore real-world biological survival stratified for discontinuation reason and determine its influenceability in rheumatoid arthritis (RA) patients. Data from the local pharmacy database and patient records of a university hospital in the Netherlands were used. RA patients who started a biological between 2000 and 2020 were included. Data on age, anti-citrullinated protein antibody (ACPA) and rheumatoid factor (RF) status, presence of erosions, gender, body mass index, time to first biological, biological survival time, use of csDMARDs, and discontinuation reasons were collected. Of the included 318 patients, 12% started their first biological within 6 months after diagnosis. The median time to first biological was 3.6 years (95% CI, 1.0–7.2). The median survival of the first- and second-line biological was respectively 1.7 years (95% CI, 1.3–2.2) and 0.8 years (95% CI, 0.5–1.0) (p = 0.0001). Discontinuation reasons for the first-line biological were ineffectiveness (47%), adverse events (17%), remission (16%), pregnancy (30%), or patient preference (10%). Multivariable Cox regression analyses for discontinuation due to inefficacy or adverse events showed that concomitant use of csDMARDs (HR = 1.32, p < 0.001) positively while RF positivity negatively (HR = 0.82, p = 0.03) influenced biological survival. ACPA positivity was associated with the inability to discontinue biologicals after achieving remission (HR = 1.43, p = 0.023). Second-line TNF inhibitor survival was similar between patients with a primary and secondary non-response on the first-line TNF inhibitor (HR = 1.28, p = 0.34). Biological survival diminishes with the number of biologicals used. Biological survival is prolonged if patients use csDMARDs. RF was negatively associated with biological survival. ACPA was negatively associated with the inability to discontinue biologicals after achieving remission. Therefore, tailoring treatment based upon autoantibody status might be the first step towards personalized medicine in RA.Key Points • Prolonged biological survival is a surrogate for treatment effectiveness; however, an increasing amount of patients will taper treatment due to remission, and factors influencing biological survival based on separate reasons for discontinuation have not been explored. • We found that combining a biological DMARD with a conventional synthetic DMARD increases biological DMARD survival. Rheumatoid factor is negatively associated with biological survival. Anti-citrullinated protein antibody is negatively associated with the inability to discontinue the biological when remission was reached. • The first step towards personalized medicine might be tailoring of treatment based upon autoantibody status. |
Collapse
Affiliation(s)
- Elise van Mulligen
- Department of Rheumatology, Erasmus MC, Room Na-523, Postbus 2040, 3000 CA, Rotterdam, the Netherlands.
| | - Saad Ahmed
- Department of Rheumatology, Erasmus MC, Room Na-523, Postbus 2040, 3000 CA, Rotterdam, the Netherlands
| | - Angelique E A M Weel
- Department of Rheumatology, Erasmus MC, Room Na-523, Postbus 2040, 3000 CA, Rotterdam, the Netherlands.,Department of Rheumatology, Maasstad Hospital, Rotterdam, the Netherlands.,Erasmus School of Health Policy & Management, Rotterdam, the Netherlands
| | - Johanna M W Hazes
- Department of Rheumatology, Erasmus MC, Room Na-523, Postbus 2040, 3000 CA, Rotterdam, the Netherlands
| | - Annette H M van der Helm-van Mil
- Department of Rheumatology, Erasmus MC, Room Na-523, Postbus 2040, 3000 CA, Rotterdam, the Netherlands.,Department of Rheumatology, LUMC, Leiden, the Netherlands
| | - Pascal H P de Jong
- Department of Rheumatology, Erasmus MC, Room Na-523, Postbus 2040, 3000 CA, Rotterdam, the Netherlands
| |
Collapse
|
28
|
Lopatina E, Marshall DA, Coupal L, Le Lorier J, Choquette D. Cost-utility analysis of second-line therapy with rituximab compared to tumour necrosis factor inhibitors in rheumatoid arthritis. Curr Med Res Opin 2021; 37:157-166. [PMID: 33136462 DOI: 10.1080/03007995.2020.1846172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To compare direct costs and treatment utility associated with the second-line therapy with rituximab and tumour necrosis factor inhibitors (TNFis) (adalimumab, etanercept, and infliximab) in patients with Rheumatoid Arthritis (RA) using data from a prospective registry. METHODS Health Assessment Questionnaire Disability Index (HAQ-DI) scores and RA-related healthcare resource utilization data (biologic agents and visits to rheumatologists) were extracted from a registry (Quebec, Canada) for patients with RA (n = 129) who had to discontinue a first-line TNFi and were treated with rituximab, adalimumab, etanercept, or infliximab as the second-line therapy between January 2007 and May 2016. A decision analytic model followed patients for 1 and 6 years. Treatment utility was measured as quality-adjusted life-years (QALYs) gained, which were calculated from HAQ-DI scores observed over the follow-up time. Quebec 2020 unit costs (Canadian Dollars, $) were used to value healthcare resource consumption. A probabilistic sensitivity analysis was performed with 10,000 Monte Carlo simulations to assess uncertainty around point-estimates of cost-utility. RESULTS Over 1-year, rituximab and etanercept resulted in the effectiveness of 0.80 QALYs gained at the cost of $14,291and $18,880, respectively, and were dominant (i.e. associated with lower costs and more QALYs gained) compared to adalimumab (0.79 QALYs, $18,825) and infliximab (0.76 QALYs, $20,158). Over 6-years, rituximab (4.42 QALYs, $82,402) was dominant compared to adalimumab (4.30 QALYs, $101,420), etanercept (4.02 QALYs, $99,191), and infliximab (3.71 QALYs, $100,396). In the probabilistic analysis, rituximab was dominant over adalimumab, etanercept, and infliximab with the probability of 0.51, 0.62, and 0.65, respectively. CONCLUSION Real-world data revealed differences between alternative biologic agents used as the second-line therapy in terms of both treatment costs for the healthcare system and utility of treatment for patients. Therefore, new guidelines on the order of selecting and switching biologic agents should be explored.
Collapse
Affiliation(s)
- Elena Lopatina
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Deborah A Marshall
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Louis Coupal
- Institut de Recherche en Rhumatologie de Montréal, Montreal, QC, Canada
| | - Jacques Le Lorier
- Département of Médicine, Université de Montréal, Montreal, QC, Canada
- Département of Pharmacologie, Université de Montréal, Montreal, QC, Canada
| | - Denis Choquette
- Institut de Recherche en Rhumatologie de Montréal, Montreal, QC, Canada
- Département of Médicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
29
|
Nahra V, Hasbani GE, Chaaya M, Uthman I. The Use of Infliximab (Remicade®) for the Treatment of Rheumatic Diseases at a Tertiary Center in Lebanon: A 17-Year Retrospective Chart Review. Mediterr J Rheumatol 2020; 31:400-405. [PMID: 33521572 PMCID: PMC7841097 DOI: 10.31138/mjr.31.4.400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 08/06/2020] [Accepted: 09/01/2020] [Indexed: 11/07/2022] Open
Abstract
Objectives: Infliximab (Remicade®) was the first tumour necrosis factor-α (TNF) inhibitor to receive its initial marketing approval from the US Food and Drug Administration (FDA) for the treatment of Crohn’s disease. Following that, infliximab became approved for several immune-mediated inflammatory diseases. No evidence exists in the Middle East and North Africa region on the experience with infliximab use over an extended period in terms of efficacy and safety. Methods: The Rheumatology division at the American University of Beirut Medical Centre (AUBMC), one of the largest tertiary centres in the Middle East and North Africa region, has been using infliximab infusions for the treatment of certain rheumatic diseases for around two decades. By reviewing retrospectively medical charts at AUBMC, we investigate indications, safety and efficacy, rate of withdrawals, rate of switching to another biologic, and financial coverage of the drug to present data for practitioners and patients in the region considering infliximab for treatment of immune-mediated inflammatory diseases. Results: A total of 198 patients were identified in the past 17 years to have taken infliximab. The largest proportion of treated patients had RA. Fourteen percent of the total cohort experienced serious adverse events, with 96.4% of those events being mild hypersensitivity reactions. Five patients withdrew the medication because of infectious complications, 4 of which were cases of tuberculosis reactivation. Despite that, around half of the patients were switched to another biologic agent such anti-TNF-α, anti-CD20, and anti-IL-6 due to partial response, and less than half were receiving add-on disease-modifying anti-rheumatic drugs (DMARDs) such as methotrexate, 70% of patients who used infliximab only or were switched achieved complete remission at their last hospital information. Around 98% of infliximab users were financially covered. Conclusion: According to our experience, infliximab has made remission and prevention of long-term disability realistic goals of therapy in the Middle East region.
Collapse
Affiliation(s)
- Vicky Nahra
- Division of Rheumatology, Department of Internal Medicine, American University of Beirut, Medical Center, Beirut, Lebanon
| | - Georges El Hasbani
- Division of Rheumatology, Department of Internal Medicine, American University of Beirut, Medical Center, Beirut, Lebanon
| | - Monique Chaaya
- Department of Epidemiology and Population Health, Faculty of Health Sciences, American University of Beirut, Medical Center, Beirut, Lebanon
| | - Imad Uthman
- Division of Rheumatology, Department of Internal Medicine, American University of Beirut, Medical Center, Beirut, Lebanon
| |
Collapse
|
30
|
Sauna ZE, Richards SM, Maillere B, Jury EC, Rosenberg AS. Editorial: Immunogenicity of Proteins Used as Therapeutics. Front Immunol 2020; 11:614856. [PMID: 33193460 PMCID: PMC7658604 DOI: 10.3389/fimmu.2020.614856] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 12/04/2022] Open
Affiliation(s)
- Zuben E Sauna
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Susan M Richards
- Translational Medicine and Early Development, Sanofi Research and Development, Sanofi, Framingham, MA, United States
| | - Bernard Maillere
- Université de Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, SIMoS, Gif-sur-Yvette, France
| | - Elizabeth C Jury
- Centre for Rheumatology, Division of Medicine, University College London, London, United Kingdom
| | - Amy S Rosenberg
- Laboratory of Immunology, Division of Biotechnology Product Review and Research 3, Office of Biotechnology Products, Center for Drugs Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
31
|
Park SH, Han X, Lobo F, Nanji S, Patel D. A Cost per Responder Model for Abatacept versus Adalimumab Among Rheumatoid Arthritis Patients with Seropositivity. CLINICOECONOMICS AND OUTCOMES RESEARCH 2020; 12:589-594. [PMID: 33116698 PMCID: PMC7571575 DOI: 10.2147/ceor.s263903] [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: 05/28/2020] [Accepted: 09/09/2020] [Indexed: 01/11/2023] Open
Abstract
Purpose The primary objective of this study was to compare the cost per responder (CPR) between abatacept and adalimumab among seropositive rheumatoid arthritis (RA) patients. Patients and Methods CPR analysis was conducted from a US payer perspective over 24 weeks for early moderate-to-severe seropositive RA patients. Efficacy data (American College of Rheumatology [ACR] improvement criteria [ACR20/50/70] and DAS28-C reactive protein <2.6) for abatacept and adalimumab were sourced from the post hoc analysis of the Early AMPLE trial (NCT02557100). Medication costs were considered assuming complete adherence. A 30% rebate was applied for adalimumab in the base case. Results At week 24, the total per patient pharmacy cost was $26,273.34 and $21,731.18, whereas the CPR (using ACR70 as the responder definition) was $46,337.46 and $74,935.10 (difference of -$28,597.64) for abatacept and adalimumab, respectively. The CPR was consistently lower for abatacept compared to adalimumab across all clinical measures, with differences ranging from -$7099.32 to -$43,608.97. Conclusion While the pharmacy cost was higher for abatacept compared to adalimumab, due to its higher clinical efficacy, the CPR was consistently lower for seropositive RA patients treated with abatacept. The results may be useful for healthcare decision-makers in understanding how to optimize treatment for seropositive RA patients while minimizing costs in today’s budget-constrained health environment.
Collapse
Affiliation(s)
- Sang Hee Park
- Modeling and Meta-Analysis, Pharmerit International, Bethesda, MD, USA
| | - Xue Han
- WW HEOR Markets-US, Bristol-Myers Squibb Company, Lawrenceville, NJ, USA
| | - Francis Lobo
- WW HEOR Markets-US, Bristol-Myers Squibb Company, Lawrenceville, NJ, USA
| | - Sakina Nanji
- Modeling and Meta-Analysis, Pharmerit International, Bethesda, MD, USA
| | - Dipen Patel
- Modeling and Meta-Analysis, Pharmerit International, Bethesda, MD, USA
| |
Collapse
|
32
|
Genovese MC, Gaylis NB, Sikes D, Kivitz A, Lewis Horowitz D, Peterfy C, Glass EV, Levine YA, Chernoff D. Safety and efficacy of neurostimulation with a miniaturised vagus nerve stimulation device in patients with multidrug-refractory rheumatoid arthritis: a two-stage multicentre, randomised pilot study. THE LANCET. RHEUMATOLOGY 2020; 2:e527-e538. [PMID: 38273617 DOI: 10.1016/s2665-9913(20)30172-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/02/2023]
Abstract
Background The inflammatory reflex plays a role in regulating innate and adaptive immunity by modulating cellular and molecular inflammatory pathways. The vagus nerve is a major constituent of the inflammatory reflex and studies have shown that the reflex can be activated by electrical stimulation of the vagus nerve. In this first in-human pilot study, we assessed the safety and efficacy of a novel miniaturised vagus nerve stimulation (VNS) device for the treatment of multidrug-refractory rheumatoid arthritis. METHODS Participants with moderately to severely active rheumatoid arthritis and prior insufficient response to two or more biological disease-modifying anti-rheumatic drugs or Janus kinase inhibitors with at least two different modes of action were enrolled in a two-stage study done at five clinical research sites in the USA. Stage 1 was open label; participants were implanted with a miniaturised VNS device, which was activated for 1 min once a day. In stage 2, participants were randomly assigned (1:1:1) to receive active stimulation (1 min once a day or 1 min four times a day) or sham stimulation (device implanted but not activated), with the sites and participants masked to treatment assignment. The primary outcome was incidence of treatment-emergent adverse events. Clinical efficacy was assessed as a key secondary outcome. The study was registered with ClinicalTrials.gov, NCT03437473. FINDINGS 14 patients were enrolled between March 13 and Aug 8, 2018. Three patients received stimulation in stage 1 and, following safety review board approval, the remaining 11 patients were implanted during stage 2 and randomly assigned to receive 1 min of stimulation once daily (n=3), 1 min of stimulation four times daily (n=4), or no stimulation (n=4) for 12 weeks. There were no device-related or treatment-related serious adverse events. Surgery-related adverse events were Horner's syndrome and vocal cord paralysis (in one patient each), which resolved without clinically significant sequelae. No deaths were recorded. INTERPRETATION VNS with a miniaturised neurostimulator was safe and well tolerated and reduced signs and symptoms of rheumatoid arthritis in patients with multidrug-refractory disease. These results support further evaluation in a larger randomised sham-controlled study. FUNDING SetPoint Medical.
Collapse
Affiliation(s)
- Mark C Genovese
- Division of Immunology and Rheumatology, Stanford University, Palo Alto, CA, USA.
| | - Norman B Gaylis
- Arthritis and Rheumatic Disease Specialties, Aventura, FL, USA
| | - David Sikes
- Department of Rheumatology, Florida Medical Clinic, Zephyrhills, FL, USA
| | - Alan Kivitz
- Altoona Center for Clinical Research, Duncansville, PA, USA
| | | | | | | | | | | |
Collapse
|
33
|
Favalli EG. Understanding the Role of Interleukin-6 (IL-6) in the Joint and Beyond: A Comprehensive Review of IL-6 Inhibition for the Management of Rheumatoid Arthritis. Rheumatol Ther 2020; 7:473-516. [PMID: 32734482 PMCID: PMC7410942 DOI: 10.1007/s40744-020-00219-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Indexed: 12/17/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, debilitating autoimmune disorder involving inflammation and progressive destruction of the joints, affecting up to 1% of the population. The majority of patients with RA have one or more comorbid conditions, the most common being cardiovascular disease, osteoporosis, and depression, the presence of which are associated with poorer clinical outcomes and lower health-related quality of life. RA pathogenesis is driven by a complex network of proinflammatory cells and cytokines, and of these, interleukin-6 (IL-6) plays a key role in the chronic inflammation associated with RA. Through cell signaling that can be initiated by both membrane-bound and soluble forms of its receptor, IL-6 acts both locally to promote joint inflammation and destruction, and in the circulation to mediate extra-articular manifestations of RA, including pain, fatigue, morning stiffness, anemia, and weight loss. This narrative review describes the role of IL-6 in the pathogenesis of RA, its comorbidities, and extra-articular systemic manifestations, and examines the effects of the IL-6 receptor inhibitors sarilumab and tocilizumab on clinical endpoints of RA, patient-reported outcomes, and common comorbidities and extra-articular manifestations.
Collapse
Affiliation(s)
- Ennio G Favalli
- Department of Rheumatology, ASST Gaetano Pini-CTO Institute, University of Milan, Milan, Italy.
| |
Collapse
|
34
|
Rodríguez-Martín E, Nieto-Gañán I, Hernández-Breijo B, Sobrino C, García-Hoz C, Bachiller J, Martínez-Feito A, Navarro-Compán V, Lapuente-Suanzes P, Bonilla G, Pascual-Salcedo D, Roy G, Jurado T, Nozal P, Vázquez-Díaz M, Balsa A, Villar LM, Plasencia-Rodríguez C. Blood Lymphocyte Subsets for Early Identification of Non-Remission to TNF Inhibitors in Rheumatoid Arthritis. Front Immunol 2020; 11:1913. [PMID: 32973793 PMCID: PMC7481468 DOI: 10.3389/fimmu.2020.01913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022] Open
Abstract
Background: TNF inhibitors (TNFis) are widely used for the treatment of rheumatoid arthritis (RA), although the response rates to this therapy in patients with RA remains heterogeneous and < 50% achieve remission (REM). Objective: To analyze baseline peripheral blood leukocytes profiles in order to search for biomarkers identifying patients who will most likely not achieve REM under TNFi treatment. Methods: A prospective bi-center pilot study including 98 RA patients treated with TNFis and followed-up during 6 months. Patients were classified according to DAS28 as follows: those who achieved REM (DAS28 ≤ 2.6) and those who did not (DAS28 > 2.6) at 6 months after starting TNFis. These rates were also assessed by simplified disease activity index (SDAI ≤ 3.3 and SDAI > 3.3, respectively). Peripheral blood immune cells were studied by flow cytometry before treatment initiation. Results: At 6 months, 61 or 80% of patients did not achieve REM by DAS28 or SDAI, respectively. Basal leukocyte profiles differed between REM vs. non-REM patients. Non-REM patients showed lower percentages of total and naïve B cells at baseline than REM subjects. A B lymphocyte/CD4+ lymphocyte ratio (BL/CD4 ratio) <0.2 clearly associated with a higher probability of non-REM status based on DAS28 at 6 months (OR = 9.2, p = 0.006). These data were confirmed when patient response was evaluated by SDAI index. Conclusion: Our results strongly suggest that BL/CD4 ratio could be considered as a useful biomarker for the early identification of non-remitters to TNFi in clinical practice.
Collapse
Affiliation(s)
| | - Israel Nieto-Gañán
- Department of Immunology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | - Cristina Sobrino
- Department of Rheumatology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Carlota García-Hoz
- Department of Immunology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Bachiller
- Department of Rheumatology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Ana Martínez-Feito
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | | | | | - Gema Bonilla
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Dora Pascual-Salcedo
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Garbiñe Roy
- Department of Immunology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Teresa Jurado
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Pilar Nozal
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Mónica Vázquez-Díaz
- Department of Rheumatology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandro Balsa
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Luisa M Villar
- Department of Immunology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | |
Collapse
|
35
|
Shimizu H, Kobayashi H, Kanbori M, Ishii Y. Effectiveness of golimumab in rheumatoid arthritis patients with inadequate response to first-line biologic therapy: Results from a Japanese post-marketing surveillance study. Mod Rheumatol 2020; 31:556-565. [PMID: 32677849 DOI: 10.1080/14397595.2020.1797266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVES To assess the real-world effectiveness of golimumab in Japanese patients with rheumatoid arthritis who had previously received first-line biologic therapy. METHODS A post-hoc analysis of post-marketing surveillance was performed. The effectiveness of golimumab was assessed in 731 patients with an inadequate response to first-line biologic therapy stratified by their prior biologic agents. Outcome variables included DAS28-CRP, DAS28-ESR, SDAI and CDAI, and medication persistence. Logistic regression analyses were conducted to identify factors associated with the likelihood of achieving a DAS28-CRP response (good/moderate) after 24 weeks of golimumab treatment. RESULTS Patients demonstrated significant improvement in the clinical signs and symptoms of rheumatoid arthritis at 24 weeks, as indicated by the reduction of DAS28-CRP (Δ0.87), DAS28-ESR (Δ0.85), SDAI (Δ7.32), and CDAI (Δ6.98) scores. This result was consistent across the subgroups stratified by previous biologic therapy. Multivariate analysis failed to identify any factors associated with response to golimumab. CONCLUSION In the real-world clinical setting, switching to golimumab was effective for Japanese patients with an inadequate response to first-line biologic therapy regardless of the biologic agent, including both TNF and non-TNF inhibitors.
Collapse
Affiliation(s)
- Hirohito Shimizu
- Immunology Department, Medical Affairs Division, Janssen Pharmaceutical K.K, Tokyo, Japan
| | - Hisanori Kobayashi
- External Collaboration and Portfolio Management Department, Clinical Science Division, R&D, Janssen Pharmaceutical K.K, Tokyo, Japan
| | - Masayoshi Kanbori
- Japan Safety & Surveillance Division, R&D, Janssen Pharmaceutical K.K, Tokyo, Japan
| | - Yutaka Ishii
- Immunology Department, Medical Affairs Division, Janssen Pharmaceutical K.K, Tokyo, Japan
| |
Collapse
|
36
|
Sullivan E, Kershaw J, Blackburn S, Choi J, Curtis JR, Boklage S. Biologic Disease-Modifying Antirheumatic Drug Prescription Patterns for Rheumatoid Arthritis Among United States Physicians. Rheumatol Ther 2020; 7:383-400. [PMID: 32318979 PMCID: PMC7211222 DOI: 10.1007/s40744-020-00203-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Some patients with rheumatoid arthritis (RA) using tumor necrosis factor inhibitors (TNFi) experience inefficacy or lack of tolerability and hence switch to another TNFi (cycling) or to a therapy with another mode of action (switching). This study examined patient characteristics, prescribing patterns and treatment practice for RA in the United States. METHODS Data were from the Adelphi Disease Specific Programme (Q2-Q3 2016). Rheumatologists completed a survey and patient record forms for adult patients with RA who had received ≥ 1 targeted therapy. Patients were grouped by class of first-used targeted therapy, and monotherapy vs. combination therapy. TNFi patients who received ≥ 1 targeted therapy were classified as cyclers or switchers. Univariate analyses compared patient characteristics and physician factors across the analysis groups. RESULTS Overall, 631 patients received ≥ 1 targeted therapy; 535 were prescribed a TNFi as first targeted therapy, 53 a nonTNFi biologic disease-modifying antirheumatic drug (bDMARD), and 43 tofacitinib. Of 577 patients with known conventional synthetic (cs) DMARD status, 18.7% were prescribed monotherapy and 81.3% combination therapy. Combination therapy patients received significantly more concomitant medications prior to initiation of first targeted therapy than monotherapy patients (P < 0.05). The top reason for physicians to prescribe first use targeted therapy was strong overall efficacy (79.9%). Of 163 patients who progressed to second targeted therapy, 60.7% were cyclers. A lower proportion of cyclers persisted on their first use targeted therapy versus switchers (P = 0.03). The main reason physicians gave for switching patients at this stage was worsening condition (46.6%). CONCLUSIONS Most patients were prescribed a TNFi as their first targeted therapy; over half then cycled to another TNFi. This suggests other factors may influence second use targeted treatment choice and highlights the need for greater understanding of outcomes associated with subsequent treatment choices and potential benefits of switching.
Collapse
Affiliation(s)
| | | | | | | | | | - Susan Boklage
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA.
| |
Collapse
|
37
|
Gao J, Zhang G, Xu K, Ma D, Ren L, Fan J, Hou J, Han J, Zhang L. Bone marrow mesenchymal stem cells improve bone erosion in collagen-induced arthritis by inhibiting osteoclasia-related factors and differentiating into chondrocytes. Stem Cell Res Ther 2020; 11:171. [PMID: 32381074 PMCID: PMC7203805 DOI: 10.1186/s13287-020-01684-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is characterized by joint inflammation and damage to the cartilage and bone in collagen-induced arthritis (CIA). Mesenchymal stem cells (MSCs) can improve articular symptoms and reduce bone erosion in CIA rats; however, the underlying mechanism remains unknown. This study aimed to investigate the mechanism underlying MSC-induced improvement of bone destruction in CIA. Methods Wistar rats were divided into a normal group, CIA control group, MTX intervention group, and BMSC intervention group, each comprising 8 rats. Serum RANKL, OPG, and CXCL10 levels of all groups were determined via flow cytometry after 42 days of interventions. RANKL, OPG, TRAF6, CXCL10, and CXCR3 were detected on the synovial membrane via immunohistochemistry, and their relative mRNA levels were determined via RT-PCR analysis. BMSCs were labeled with GFP and administered to CIA rats via the tail vein. At different time points, the distribution of implanted GFP-MSCs in synovial tissues was observed using a fluorescence microscope, and the potential of GFP-MSCs to differentiate into chondrocytes was assessed via immunofluorescence analysis. Results BMSC transplantation improved joint inflammation and inhibited bone destruction in CIA rats. BMSCs inhibited the expression of serum CXCL10 and CXCL10 and CXCR3 expression at the synovial membrane. Moreover, protein and mRNA expression analyses revealed that BMSCs potentially regulated RANKL/OPG expression levels in the serum and synovial tissue. Upon implantation into CIA rats, GFP-MSCs were traced in the joints. GFP-positive cells were observed in the cartilage tissue from day 11 and until 42 days after transplantation. Anti-type II collagen/GFP double-positive cells were observed in the articular cartilage (especially damaged cartilage) upon immunofluorescence staining of anti-type II collagen. Conclusions BMSCs improve bone destruction in CIA by inhibiting the CXCL10/CXCR3 chemotactic axis, regulating the RANKL/OPG ratio, and directly differentiating into chondrocytes.
Collapse
Affiliation(s)
- Jinfang Gao
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.,Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Gailian Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Ke Xu
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Dan Ma
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.,Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Limin Ren
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Jingjing Fan
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Jianwen Hou
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Jian Han
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Liyun Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.
| |
Collapse
|
38
|
Favalli EG, Ingegnoli F, De Lucia O, Cincinelli G, Cimaz R, Caporali R. COVID-19 infection and rheumatoid arthritis: Faraway, so close! Autoimmun Rev 2020; 19:102523. [PMID: 32205186 PMCID: PMC7102591 DOI: 10.1016/j.autrev.2020.102523] [Citation(s) in RCA: 302] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023]
Abstract
The outbreak of the new coronavirus infections COVID-19 in December 2019 in China has quickly become a global health emergency. Given the lack of specific anti-viral therapies, the current management of severe acute respiratory syndrome coronaviruses (SARS-CoV-2) is mainly supportive, even though several compounds are now under investigation for the treatment of this life-threatening disease. COVID-19 pandemic is certainly conditioning the treatment strategy of a complex disorder as rheumatoid arthritis (RA), whose infectious risk is increased compared to the general population because of an overall impairment of immune system typical of autoimmune diseases combined with the iatrogenic effect generated by corticosteroids and immunosuppressive drugs. However, the increasing knowledge about the pathophysiology of SARS-CoV-2 infection is leading to consider some anti-rheumatic drugs as potential treatment options for the management of COVID-19. In this review we will critically analyse the evidences on either positive or negative effect of drugs commonly used to treat RA in this particular scenario, in order to optimize the current approach to RA patients.
Collapse
Affiliation(s)
- Ennio Giulio Favalli
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, Milan, Italy.
| | - Francesca Ingegnoli
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, Milan, Italy; Department of Clinical Sciences and Community Health, Research Center for Adult and Pediatric Rheumatic Diseases, Università degli Studi di Milano, Milano, Italy
| | - Orazio De Lucia
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, Milan, Italy
| | - Gilberto Cincinelli
- Department of Clinical Sciences and Community Health, Research Center for Adult and Pediatric Rheumatic Diseases, Università degli Studi di Milano, Milano, Italy
| | - Rolando Cimaz
- Department of Clinical Sciences and Community Health, Research Center for Adult and Pediatric Rheumatic Diseases, Università degli Studi di Milano, Milano, Italy; Division of Pediatric Rheumatology, ASST Gaetano Pini-CTO Institute, Milan, Italy
| | - Roberto Caporali
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, Milan, Italy; Department of Clinical Sciences and Community Health, Research Center for Adult and Pediatric Rheumatic Diseases, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
39
|
Targeting Granulocyte-Monocyte Colony-Stimulating Factor Signaling in Rheumatoid Arthritis: Future Prospects. Drugs 2020; 79:1741-1755. [PMID: 31486005 DOI: 10.1007/s40265-019-01192-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic, autoimmune disease that affects joints and extra-articular structures. In the last decade, the management of this chronic disease has dramatically changed with the introduction of several targeted mechanisms of action, such as tumor necrosis factor-α inhibition, T-cell costimulation inhibition, B-cell depletion, interleukin-6 blockade, and Janus kinase inhibition. Beyond its well-known hematopoietic role on the proliferation and differentiation of myeloid cells, granulocyte-monocyte colony-stimulating factor (GM-CSF) is a proinflammatory mediator acting as a cytokine, with a proven pathogenetic role in autoimmune disorders such as RA. In vitro studies clearly demonstrated the effect of GM-CSF in the communication between resident tissue cells and activated macrophages at chronic inflammation sites, and confirmed the elevation of GM-CSF levels in inflamed synovial tissue of RA subjects compared with healthy controls. Moreover, a pivotal role of GM-CSF in the perception of pain has been clearly confirmed. Therefore, blockade of the GM-CSF pathway by monoclonal antibodies directed against the cytokine itself or its receptor has been investigated in refractory RA patients. Overall, the safety profile of GM-CSF inhibitors seems to be very favorable, with a particularly low incidence of infectious complications. The efficacy of this new mechanism of action is comparable with main competitors, even though the response rates reported in phase II randomized controlled trials (RCTs) appear to be numerically lower than the response rates observed with other biological disease-modifying antirheumatic drugs already licensed for RA. Mainly because of this reason, nowadays the development program of most GM-CSF blockers for RA has been discontinued, with the exception of otilimab, which is under evaluation in two phase III RCTs with a head-to head non-inferiority design against tofacitinib. These studies will likely be useful for better defining the potential role of GM-CSF inhibition in the therapeutic algorithm of RA. On the other hand, the potential role of GM-CSF blockade in the treatment of other rheumatic diseases is now under investigation. Phase II trials are ongoing with the aim of evaluating mavrilimumab for the treatment of giant cell arteritis, and namilumab for the treatment of spondyloarthritis. Moreover, GM-CSF inhibitors have been tested in osteoarthritis and diffuse subtype of systemic sclerosis. This review aims to describe in detail the available evidence on the GM-CSF blocking pathway in RA management, paving the way to a possible alternative treatment for RA patients. Novel insights regarding the potential use of GM-CSF blockers for alternative indications will be also addressed.
Collapse
|
40
|
Cheng P, Wang J. The potential of circulating microRNA-125a and microRNA-125b as markers for inflammation and clinical response to infliximab in rheumatoid arthritis patients. J Clin Lab Anal 2020; 34:e23329. [PMID: 32281166 PMCID: PMC7439431 DOI: 10.1002/jcla.23329] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/15/2020] [Accepted: 03/18/2020] [Indexed: 12/21/2022] Open
Abstract
Objective This study was to investigate the changes in circulating microRNA (miR)‐125a and miR‐125b during infliximab (IFX) treatment, and their value in predicting clinical response to IFX in rheumatoid arthritis (RA) patients. Methods The plasma samples were obtained from 96 active RA patients who underwent 24‐week IFX treatment and from 96 healthy controls to detect miR‐125a and miR‐125b expressions by RT‐qPCR. Clinical response was assessed according to EULAR criteria based on disease activity alleviation at week 4, week 12, and week 24. Results MiR‐125a and miR‐125b expressions were both elevated in RA patients compared with healthy controls, and they could differentiate RA patients from healthy controls by receiver operating characteristic curve analysis. Baseline miR‐125a positively correlated with C‐reactive protein (CRP) level; meanwhile, baseline miR‐125b positively correlated with tender joint count (TJC), swollen joint count (SJC), erythrocyte sedimentation rate (ESR), CRP, and DAS28‐ESR score in RA patients. With the 24‐week IFX treatment, clinical response rate was gradually increased, while miR‐125a and miR‐125b expressions were gradually decreased in RA patients. At week 24, 69 (71.9%) patients responded to IFX treatment, while 27 (28.1%) patients did not respond to IFX treatment. Importantly, baseline miR‐125a and miR‐125b expressions were higher in responders than that in non‐responders, further multivariate logistic regression analysis disclosed that miR‐125b but not miR‐125a could independently predict better clinical response to IFX in RA patients. Conclusion Circulating miR‐125a and miR‐125b displays the potency for guiding personalized treatment strategy and improving clinical outcomes in RA patients.
Collapse
Affiliation(s)
- Peng Cheng
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jun Wang
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
41
|
Cohen S, Tuckwell K, Katsumoto TR, Zhao R, Galanter J, Lee C, Rae J, Toth B, Ramamoorthi N, Hackney JA, Berman A, Damjanov N, Fedkov D, Jeka S, Chinn LW, Townsend MJ, Morimoto AM, Genovese MC. Fenebrutinib versus Placebo or Adalimumab in Rheumatoid Arthritis: A Randomized, Double-Blind, Phase II Trial (ANDES Study). Arthritis Rheumatol 2020; 72:1435-1446. [PMID: 32270926 PMCID: PMC7496340 DOI: 10.1002/art.41275] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate fenebrutinib, an oral and highly selective non-covalent inhibitor of Bruton's tyrosine kinase (BTK), in patients with active rheumatoid arthritis (RA). METHODS Patients with RA and inadequate response to methotrexate (cohort 1, n=480) were randomized to fenebrutinib (50 mg once daily, 150 mg once daily, 200 mg twice daily), 40 mg adalimumab every other week, or placebo. Patients with RA and inadequate response to tumor necrosis factor inhibitors (cohort 2, n=98) received fenebrutinib (200 mg twice daily) or placebo. Both cohorts continued methotrexate therapy. RESULTS In cohort 1, American College of Rheumatology scores (ACR50) at week 12 were similar for fenebrutinib 50 mg once daily and placebo, and higher for fenebrutinib 150 mg once daily (28%) and 200 mg twice daily (35%) than placebo (15%) (p=0.017; p=0.0003). Fenebrutinib 200 mg twice daily and adalimumab (36%) were comparable (p=0.81). In cohort 2, more patients achieved ACR50 with fenebrutinib 200 mg twice daily (25%) than placebo (12%) (p=0.072). The most common adverse events for fenebrutinib included nausea, headache, anemia, and upper respiratory tract infections. Fenebrutinib had significant effects on myeloid and B cell biomarkers (CCL4 and rheumatoid factor). Fenebrutinib and adalimumab caused overlapping as well as distinct changes in B cell and myeloid biomarkers. CONCLUSION Fenebrutinib demonstrated efficacy comparable to adalimumab in patients with an inadequate response to methotrexate, and safety consistent with existing immunomodulatory therapies for RA. These data support targeting both B and myeloid cells via this novel mechanism for potential efficacy in the treatment of RA.
Collapse
Affiliation(s)
| | | | | | - Rui Zhao
- Genentech, Inc.South San FranciscoCalifornia
| | | | - Chin Lee
- Genentech, Inc.South San FranciscoCalifornia
| | - Julie Rae
- Genentech, Inc.South San FranciscoCalifornia
| | - Balazs Toth
- Genentech, Inc.South San FranciscoCalifornia
| | | | | | | | | | | | - Slawomir Jeka
- Collegium Medicum Jan Biziel University Hospital no 2BydgoszczPoland
| | | | | | | | | |
Collapse
|
42
|
Nguyen MVC, Courtier A, Adrait A, Defendi F, Couté Y, Baillet A, Guigue L, Gottenberg JE, Dumestre-Pérard C, Brun V, Gaudin P. Fetuin-A and thyroxin binding globulin predict rituximab response in rheumatoid arthritis patients with insufficient response to anti-TNFα. Clin Rheumatol 2020; 39:2553-2562. [PMID: 32212002 DOI: 10.1007/s10067-020-05030-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/07/2020] [Accepted: 03/05/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a debilitating disease, but patient management and treatment have been revolutionized since the advent of bDMARDs. However, about one third of RA patients do not respond to specific bDMARD treatment without clear identified reasons. Different bDMARDs must be tried until the right drug is found. Here, we sought to identify a predictive protein signature to stratify patient responsiveness to rituximab (RTX) among patients with an insufficient response to a first anti-TNFα treatment. METHODS Serum samples were collected at baseline before RTX initiation. A proteomics study comparing responders and nonresponders was conducted to identify and select potential predictive biomarkers whose concentration was measured by quantitative assays. Logistic regression was performed to determine the best biomarker combination to predict good or nonresponse to RTX (EULAR criteria after 6 months' treatment). RESULTS Eleven biomarkers potentially discriminating between responders and nonresponders were selected following discovery proteomics. Quantitative immunoassays and univariate statistical analysis showed that fetuin-A and thyroxine binding globulin (TBG) presented a good capacity to discriminate between patient groups. A logistic regression analysis revealed that the combination of fetuin-A plus TBG could accurately predict a patient's responsiveness to RTX with an AUC of 0.86, sensitivity of 80%, and a specificity of 79%. CONCLUSION In RA patients for whom a first anti-TNFα treatment has failed, the serum abundance of fetuin-A and TBG before initiating RTX treatment is an indicator for their response status at 6 months. ClinicalTrials.gov identifier: NCT01000441. Key Points • Proteomic analysis revealed 11 putative predictive biomarkers to discriminate rituximab responder vs. nonresponder RA patients. • Fetuin-A and TBG are significantly differentially expressed at baseline in rituximab responder vs. nonresponder RA patients. • Algorithm combining fetuin-A and TBG accurately predicts response to rituximab in RA patients with insufficient response to TNFi.
Collapse
Affiliation(s)
- Minh Vu Chuong Nguyen
- GREPI EA 7408, Université Grenoble Alpes, 38000, Grenoble, France. .,Sinnovial, 38000, Grenoble, France.
| | | | - Annie Adrait
- Inserm, CEA, Biologie à Grande Echelle, Université Grenoble Alpes, F-38000, Grenoble, France
| | - Federica Defendi
- Laboratoire d'Immunologie, Pôle de Biologie, Centre Hospitalier Universitaire Grenoble Alpes, 38000, Grenoble Cedex 9, France
| | - Yohann Couté
- Inserm, CEA, Biologie à Grande Echelle, Université Grenoble Alpes, F-38000, Grenoble, France
| | - Athan Baillet
- GREPI EA 7408, Université Grenoble Alpes, 38000, Grenoble, France.,Rheumatology Department, Centre Hospitalier Universitaire Grenoble Alpes, Hôpital Sud Echirolles, 38130, Echirolles, France
| | | | - Jacques-Eric Gottenberg
- Department of Rheumatology, National Reference Center for Rare Systemic Autoimmune Diseases, Strasbourg. University Hospital, CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique/Laboratory of excellence MEDALIS, Université de Strasbourg, Hôpital Hautepierre, 1 Ave Molière, 67000, Strasbourg, France
| | - Chantal Dumestre-Pérard
- Laboratoire d'Immunologie, Pôle de Biologie, Centre Hospitalier Universitaire Grenoble Alpes, 38000, Grenoble Cedex 9, France
| | - Virginie Brun
- Inserm, CEA, Biologie à Grande Echelle, Université Grenoble Alpes, F-38000, Grenoble, France
| | - Philippe Gaudin
- GREPI EA 7408, Université Grenoble Alpes, 38000, Grenoble, France.,Rheumatology Department, Centre Hospitalier Universitaire Grenoble Alpes, Hôpital Sud Echirolles, 38130, Echirolles, France
| |
Collapse
|
43
|
Efficacy and retention rate of adalimumab in rheumatoid arthritis and psoriatic arthritis patients after first-line etanercept failure: the FEARLESS cohort. Rheumatol Int 2020; 40:263-272. [DOI: 10.1007/s00296-019-04416-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/08/2019] [Indexed: 10/26/2022]
|
44
|
Pan S, Tsakok T, Dand N, Lonsdale DO, Loeff FC, Bloem K, de Vries A, Baudry D, Duckworth M, Mahil S, Pushpa-Rajah A, Russell A, Alsharqi A, Becher G, Murphy R, Wahie S, Wright A, Griffiths CEM, Reynolds NJ, Barker J, Warren RB, David Burden A, Rispens T, Standing JF, Smith CH. Using Real-World Data to Guide Ustekinumab Dosing Strategies for Psoriasis: A Prospective Pharmacokinetic-Pharmacodynamic Study. Clin Transl Sci 2020; 13:400-409. [PMID: 31995663 PMCID: PMC7070790 DOI: 10.1111/cts.12725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022] Open
Abstract
Variation in response to biologic therapy for inflammatory diseases, such as psoriasis, is partly driven by variation in drug exposure. Real‐world psoriasis data were used to develop a pharmacokinetic/pharmacodynamic (PK/PD) model for the first‐line therapeutic antibody ustekinumab. The impact of differing dosing strategies on response was explored. Data were collected from a UK prospective multicenter observational cohort (491 patients on ustekinumab monotherapy, drug levels, and anti‐drug antibody measurements on 797 serum samples, 1,590 measurements of Psoriasis Area Severity Index (PASI)). Ustekinumab PKs were described with a linear one‐compartment model. A maximum effect (Emax) model inhibited progression of psoriatic skin lesions in the turnover PD mechanism describing PASI evolution while on treatment. A mixture model on half‐maximal effective concentration identified a potential nonresponder group, with simulations suggesting that, in future, the model could be incorporated into a Bayesian therapeutic drug monitoring “dashboard” to individualize dosing and improve treatment outcomes.
Collapse
Affiliation(s)
- Shan Pan
- St. John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Teresa Tsakok
- St. John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK.,St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Nick Dand
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Dagan O Lonsdale
- Institute of Infection and Immunity, St. George's, University of London, London, UK
| | - Floris C Loeff
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Karien Bloem
- Biologics Lab, Sanquin Diagnostic Services, Amsterdam, The Netherlands
| | - Annick de Vries
- Biologics Lab, Sanquin Diagnostic Services, Amsterdam, The Netherlands
| | - David Baudry
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Michael Duckworth
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Satveer Mahil
- St. John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK.,St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Angela Pushpa-Rajah
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Alice Russell
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Ali Alsharqi
- Dermatology Department, Royal Liverpool and Broadgreen University Hospital Trust, Liverpool, UK
| | | | - Ruth Murphy
- Department of Dermatology, Queens Medical Centre, Nottingham University Teaching Hospitals, Nottingham, UK
| | - Shyamal Wahie
- Dermatology Department, University Hospital of North Durham, Durham, UK
| | - Andrew Wright
- Centre for Skin Sciences, University of Bradford, Bradford, UK
| | - Christopher E M Griffiths
- Dermatology Centre, Salford Royal National Health Service Foundation Trust, Manchester, UK.,The University of Manchester, Manchester Academic Health Science Centre, National Institute for Health Research Manchester Biomedical Research Centre, Manchester, UK
| | - Nick J Reynolds
- Dermatological Sciences, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, UK.,Department of Dermatology, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Jonathan Barker
- St. John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK.,St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Richard B Warren
- Dermatology Centre, Salford Royal National Health Service Foundation Trust, Manchester, UK
| | - A David Burden
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Joseph F Standing
- Infection, Immunity, Inflammation Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Catherine H Smith
- St. John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK.,St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | | |
Collapse
|
45
|
Deviatkin AA, Vakulenko YA, Akhmadishina LV, Tarasov VV, Beloukhova MI, Zamyatnin Jr. AA, Lukashev AN. Emerging Concepts and Challenges in Rheumatoid Arthritis Gene Therapy. Biomedicines 2020; 8:biomedicines8010009. [PMID: 31936504 PMCID: PMC7168286 DOI: 10.3390/biomedicines8010009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/03/2020] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic inflammatory joint disease affecting about 1% of the population worldwide. Current treatment approaches do not ensure a cure for every patient. Moreover, classical regimens are based on nontargeted systemic immune suppression and have significant side effects. Biological treatment has advanced considerably but efficacy and specificity issues remain. Gene therapy is one of the potential future directions for RA therapy, which is rapidly developing. Several gene therapy trials done so far have been of moderate success, but experimental and genetics studies have yielded novel targets. As a result, the arsenal of gene therapy tools keeps growing. Currently, both viral and nonviral delivery systems are used for RA therapy. Herein, we review recent approaches for RA gene therapy.
Collapse
Affiliation(s)
- Andrei A. Deviatkin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
- Correspondence:
| | - Yulia A. Vakulenko
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (Y.A.V.); (L.V.A.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Ludmila V. Akhmadishina
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (Y.A.V.); (L.V.A.)
| | - Vadim V. Tarasov
- Department of Pharmacology and Pharmacy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Marina I. Beloukhova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
| | - Andrey A. Zamyatnin Jr.
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Alexander N. Lukashev
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.I.B.); (A.A.Z.J.); (A.N.L.)
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (Y.A.V.); (L.V.A.)
| |
Collapse
|
46
|
Post-rituximab immunoglobulin M (IgM) hypogammaglobulinemia. Autoimmun Rev 2020; 19:102466. [PMID: 31917267 DOI: 10.1016/j.autrev.2020.102466] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 12/19/2022]
Abstract
Rituximab is a B cell depleting monoclonal antibody that targets the B cell-specific cell surface antigen CD20 and is currently used to treat several autoimmune diseases. The elimination of mature CD20-positive B lymphocytes committed to differentiate into autoantibody-producing plasma cells is considered to be the major effect of rituximab, that makes it a beneficial biological agent in treating autoimmune diseases. Hypogammaglobulinemia has been reported after rituximab therapy in patients with lymphoma and rheumatoid arthritis. Similar data are scarce for other autoimmune diseases. Low immunoglobulin G (IgG) or hypogammaglobulinemia has attracted the most attention because of its significant role in protective immunity. However, the incidence and clinical implications of low immunoglobulin M (IgM) or hypogammaglobulinemia have not been studied in detail. This review will focus on the frequency and the clinical concerns of low IgM levels that result as a consequence of the administration of rituximab. The etiopathogenic mechanisms underlying post-rituximab IgM hypogammaglobulinemia and its implications are presented. The long-term consequences, if any, are not known or documented. Multiple factors may be involved in whether IgG or IgM decreases secondary to rituximab therapy. It is possible that the autoimmune disease itself may be one of the important factors. The dose, frequency and number of infusions appear to be important variables. Post-rituximab therapy immunoglobulin levels return to normal. During this process. IgM levels take a longer time to return to normal levels when compared to IgG or other immunoglobulins. IgM deficiency persists after B cell repopulation to normal levels has occurred. Laboratory animals and humans deficient in IgM can have multiple infections. Specific pharmacologic agents or biologic therapy that address and resolve IgM deficiency are currently unavailable. If the clinical situation so warrants, then prophylactic antibiotics may be indicated and perhaps helpful. Research in this iatrogenic phenomenon will provide a better understanding of not only the biology of IgM, but also the factor(s) that control its production and regulation, besides its influence if any, on rituximab therapy.
Collapse
|
47
|
Failure of anti-TNF treatment in patients with rheumatoid arthritis: The pros and cons of the early use of alternative biological agents. Autoimmun Rev 2019; 18:102398. [DOI: 10.1016/j.autrev.2019.102398] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 06/06/2019] [Indexed: 12/16/2022]
|
48
|
The Giants (biologicals) against the Pigmies (small molecules), pros and cons of two different approaches to the disease modifying treatment in rheumatoid arthritis. Autoimmun Rev 2019; 19:102421. [PMID: 31733368 DOI: 10.1016/j.autrev.2019.102421] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/13/2019] [Indexed: 02/08/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease that, if untreated, can lead to disability and reduce the life expectancy of affected patients. Over the last two decades the improvement of knowledge of the pathogenetic mechanisms leading to the development of the disease has profoundly changed the treatment strategies of RA through the development of biotechnological drugs (bDMARDs) directed towards specific pro-inflammatory targets involved in the RA network. To date, the therapeutic armamentarium for RA includes ten bDMARDs able to produce the depletion B-cells, the blockade of three different pro-inflammatory cytokines (tumour necrosis factor alpha, interleukin-6 and interleukin-1), or the inhibition of T-cell co-stimulation. The introduction of these new compounds has dramatically improved outcomes in the short and long term, although still a significant proportion of patients are unable to reach or maintain the treatment target over time. The identification of the fundamental role of Janus kinases in the process of transduction of the inflammatory signal within the immune cells has recently provided the opportunity to use the new pharmacological class of small molecules for the therapy of RA, further increasing the number of treatment options. In this review the PROS and CONS of these two drug classes will be discussed, trying to provide the evidence currently available to make the right choice based on the analysis of the efficacy and safety profile of the different drugs on the market and close to marketing.
Collapse
|
49
|
Biggioggero M, Becciolini A, Crotti C, Agape E, Favalli EG. Upadacitinib and filgotinib: the role of JAK1 selective inhibition in the treatment of rheumatoid arthritis. Drugs Context 2019; 8:212595. [PMID: 31692920 PMCID: PMC6821397 DOI: 10.7573/dic.212595] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease characterized by joint involvement, extra-articular manifestations, comorbidities, and increased mortality. In the last few decades, the management of RA has been dramatically improved by the introduction of a treat-to-target approach aiming to prevent joint damage progression. Moreover, the increasing knowledge about disease pathogenesis allowed the development of a new drug class of biologic agents targeted on immune cells and proinflammatory cytokines involved in RA network. Despite the introduction of several targeted drugs, a significant proportion of RA patients still fail to achieve the clinical target; so, more recently the focus of research has been shifted toward the inhibition of kinases involved in the transduction of the inflammatory signal into immune cells. In particular, two Janus kinase (JAK) inhibitors, baricitinib and tofacitinib, have been licensed for the treatment of RA as a consequence of a very favorable profile observed in randomized controlled trials (RCTs) conducted across different RA subpopulations. Both these new compounds are active on the majority of four JAK family members (JAK1, JAK2, JAK3, and TYK2), whereas the most recent emerging approach is directed toward the development of JAK1 selective inhibitors (upadacitinib and filgotinib) with the aim to improve the safety profile by minimizing the effects on JAK3 and, especially, JAK2. In this narrative review, we discuss the rationale for JAK inhibition in RA, with a special focus on the role of JAK1 selective blockade and a detailed description of available data from the results of clinical trials on upadacitinib and filgotinib.
Collapse
Affiliation(s)
| | | | - Chiara Crotti
- Department of Rheumatology, Gaetano Pini Institute, Milan, Italy
| | - Elena Agape
- Department of Clinical Sciences and Health Community, University of Milan, Division of Rheumatology, Gaetano Pini Institute, Milan, Italy
| | | |
Collapse
|
50
|
Irrera N, D'Ascola A, Pallio G, Bitto A, Mazzon E, Mannino F, Squadrito V, Arcoraci V, Minutoli L, Campo GM, Avenoso A, Bongiorno EB, Vaccaro M, Squadrito F, Altavilla D. β-Caryophyllene Mitigates Collagen Antibody Induced Arthritis (CAIA) in Mice Through a Cross-Talk between CB2 and PPAR-γ Receptors. Biomolecules 2019; 9:biom9080326. [PMID: 31370242 PMCID: PMC6723248 DOI: 10.3390/biom9080326] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/22/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023] Open
Abstract
β-caryophyllene (BCP) is a cannabinoid receptor 2 (CB2) agonist that tempers inflammation. An interaction between the CB2 receptor and peroxisome proliferator-activated receptor gamma (PPAR-γ) has been suggested and PPAR-γ activation exerts anti-arthritic effects. The aim of this study was to characterize the therapeutic activity of BCP and to investigate PPAR-γ involvement in a collagen antibody induced arthritis (CAIA) experimental model. CAIA was induced through intraperitoneal injection of a monoclonal antibody cocktail and lipopolysaccharide (LPS; 50 μg/100 μL/ip). CAIA animals were then randomized to orally receive either BCP (10 mg/kg/100 μL) or its vehicle (100 μL of corn oil). BCP significantly hampered the severity of the disease, reduced relevant pro-inflammatory cytokines, and increased the anti-inflammatory cytokine IL-13. BCP also decreased joint expression of matrix metalloproteinases 3 and 9. Arthritic joints showed increased COX2 and NF-ĸB mRNA expression and reduced expression of the PPARγ coactivator-1 alpha, PGC-1α, and PPAR-γ. These conditions were reverted following BCP treatment. Finally, BCP reduced NF-ĸB activation and increased PGC-1α and PPAR-γ expression in human articular chondrocytes stimulated with LPS. These effects were reverted by AM630, a CB2 receptor antagonist. These results suggest that BCP ameliorates arthritis through a cross-talk between CB2 and PPAR-γ.
Collapse
Affiliation(s)
- Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Angela D'Ascola
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Giovanni Pallio
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", 98124 Messina, Italy
| | - Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Violetta Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Vincenzo Arcoraci
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Giuseppe Maurizio Campo
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Angela Avenoso
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy
| | | | - Mario Vaccaro
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy.
| | - Domenica Altavilla
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy
| |
Collapse
|