1
|
Nguyen H, Staeva TP. Lupus ABC spearheading a new era of collaboration to advance lupus drug development. Nat Rev Rheumatol 2025; 21:253-254. [PMID: 40169881 DOI: 10.1038/s41584-025-01247-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
|
2
|
Bloostein A, Caricchio R. Patient-Reported Factors Influencing Clinical Trial Participation: An In-depth Analysis of Patients With Lupus and Rheumatoid Arthritis. ACR Open Rheumatol 2025; 7:e70035. [PMID: 40293092 PMCID: PMC12035868 DOI: 10.1002/acr2.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Systemic lupus erythematosus (SLE), a chronic systemic autoimmune disease, has only three approved therapies, despite a rapidly expanding number of clinical trials. Trials in SLE struggle to recruit patients and fail to fully enroll. Rheumatoid arthritis (RA), another chronic autoimmune disease, has had more success in clinical trial enrollment. This article's goal is to analyze literature to compare patient perspectives on factors determining clinical trial participation in SLE and RA populations. A literature review regarding patient attitudes, perceptions, perspectives about, and barriers toward clinical trials was conducted. RA literature used more quantitative methods, whereas SLE literature used more qualitative methods. Literature revealed that patients with SLE and patients with RA reported similar factors motivating trial participation, such as altruism, personal benefit, and trust in physician, but patients with SLE were uniquely motivated by strong social networks. In terms of disadvantages to trial participation, patients with SLE and RA similarly reported fear of unknown interventions, complications, and side effects; only in the population of patients with SLE were fears of a disease flare and of not qualifying reported. SLE literature had a specific emphasis on factors influencing Black patient participation. This review informs steps for future research to better evaluate perceptions of clinical trials in patients with SLE and provides a framework for methods to increase SLE trial participation, including recognizing fear of changing treatment regimen as an explanation for the paucity of SLE trial participation, incorporating clinical trials as usual care for patients with SLE, and using less stringent inclusion and exclusion criteria for trials.
Collapse
|
3
|
Merrill JT, Tanaka Y, D'Cruz D, Vila-Rivera K, Siri D, Zeng X, Saxena A, Aringer M, D'Silva KM, Cheng L, Mohamed MEF, Siovitz L, Bhatnagar S, Gaudreau MC, Doan TT, Friedman A. Efficacy and Safety of Upadacitinib or Elsubrutinib Alone or in Combination for Patients With Systemic Lupus Erythematosus: A Phase 2 Randomized Controlled Trial. Arthritis Rheumatol 2024; 76:1518-1529. [PMID: 38923871 DOI: 10.1002/art.42926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/11/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE The 48-week, phase 2 SLEek study (NCT03978520) evaluated the efficacy and safety of upadacitinib (JAK inhibitor) and elsubrutinib (BTK inhibitor) alone or in combination (ABBV-599) in adults with moderately to severely active systemic lupus erythematosus (SLE). METHODS Patients were randomized 1:1:1:1:1 to elsubrutinib 60 mg and upadacitinib 30 mg once daily (ABBV-599 high dose), elsubrutinib 60 mg and upadacitinib 15 mg once daily (ABBV-599 low dose), elsubrutinib 60 mg once daily (QD), upadacitinib 30 mg QD, or placebo QD. The primary endpoint was the proportion of patients achieving both Systemic Lupus Erythematosus Responder Index 4 (SRI-4) and glucocorticoid dose ≤10 mg QD at week 24. Additional assessments through week 48 included British Isles Lupus Assessment Group-Based Composite Lupus Assessment (BICLA) and Lupus Low Disease Activity State (LLDAS) responses, number of flares, time to first flare, and adverse events. RESULTS The study enrolled 341 patients. The ABBV-599 low dose and elsubrutinib arms were discontinued after a planned interim analysis showed lack of efficacy (no safety concerns). More patients achieved the primary endpoint with upadacitinib (54.8%; P = 0.028) and ABBV-599 high dose (48.5%; P = 0.081) versus placebo (37.3%). SRI-4, BICLA, and LLDAS response rates were higher for both upadacitinib and ABBV-599 high dose versus placebo at weeks 24 and 48. Flares were reduced, and time to first flare through week 48 was substantially delayed with both upadacitinib and ABBV-599 high dose versus placebo. No new safety signals were observed beyond those previously reported for upadacitinib or elsubrutinib. CONCLUSION Upadacitinib 30 mg alone or in combination with elsubrutinib (ABBV-599 high dose) demonstrated significant improvements in SLE disease activity and reduced flares and were well tolerated through 48 weeks.
Collapse
Affiliation(s)
| | - Yoshiya Tanaka
- University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - David D'Cruz
- King's College London and Guy's Hospital, London, United Kingdom
| | | | | | - Xiaofeng Zeng
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Amit Saxena
- New York University Grossman School of Medicine, New York
| | - Martin Aringer
- University Medical Center and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Echavarria R, Cardona-Muñoz EG, Ortiz-Lazareno P, Andrade-Sierra J, Gómez-Hermosillo LF, Casillas-Moreno J, Campos-Bayardo TI, Román-Rojas D, García-Sánchez A, Miranda-Díaz AG. The Role of the Oxidative State and Innate Immunity Mediated by TLR7 and TLR9 in Lupus Nephritis. Int J Mol Sci 2023; 24:15234. [PMID: 37894915 PMCID: PMC10607473 DOI: 10.3390/ijms242015234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/25/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Lupus nephritis (LN) is a severe complication of systemic lupus erythematosus (SLE) and is considered one of the leading causes of mortality. Multiple immunological pathways are involved in the pathogenesis of SLE, which makes it imperative to deepen our knowledge about this disease's immune-pathological complexity and explore new therapeutic targets. Since an altered redox state contributes to immune system dysregulation, this document briefly addresses the roles of oxidative stress (OS), oxidative DNA damage, antioxidant enzymes, mitochondrial function, and mitophagy in SLE and LN. Although adaptive immunity's participation in the development of autoimmunity is undeniable, increasing data emphasize the importance of innate immunity elements, particularly the Toll-like receptors (TLRs) that recognize nucleic acid ligands, in inflammatory and autoimmune diseases. Here, we discuss the intriguing roles of TLR7 and TLR9 in developing SLE and LN. Also included are the essential characteristics of conventional treatments and some other novel and little-explored alternatives that offer options to improve renal function in LN.
Collapse
Affiliation(s)
- Raquel Echavarria
- Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (R.E.); (P.O.-L.)
- Investigadores por México, Consejo Nacional de Ciencia y Tecnología (CONACYT), Ciudad de México 03940, Mexico
| | - Ernesto Germán Cardona-Muñoz
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Mexico; (E.G.C.-M.); (J.A.-S.); (L.F.G.-H.); (J.C.-M.); (T.I.C.-B.); (D.R.-R.); (A.G.-S.)
| | - Pablo Ortiz-Lazareno
- Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico; (R.E.); (P.O.-L.)
| | - Jorge Andrade-Sierra
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Mexico; (E.G.C.-M.); (J.A.-S.); (L.F.G.-H.); (J.C.-M.); (T.I.C.-B.); (D.R.-R.); (A.G.-S.)
| | - Luis Francisco Gómez-Hermosillo
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Mexico; (E.G.C.-M.); (J.A.-S.); (L.F.G.-H.); (J.C.-M.); (T.I.C.-B.); (D.R.-R.); (A.G.-S.)
| | - Jorge Casillas-Moreno
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Mexico; (E.G.C.-M.); (J.A.-S.); (L.F.G.-H.); (J.C.-M.); (T.I.C.-B.); (D.R.-R.); (A.G.-S.)
| | - Tannia Isabel Campos-Bayardo
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Mexico; (E.G.C.-M.); (J.A.-S.); (L.F.G.-H.); (J.C.-M.); (T.I.C.-B.); (D.R.-R.); (A.G.-S.)
| | - Daniel Román-Rojas
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Mexico; (E.G.C.-M.); (J.A.-S.); (L.F.G.-H.); (J.C.-M.); (T.I.C.-B.); (D.R.-R.); (A.G.-S.)
| | - Andrés García-Sánchez
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Mexico; (E.G.C.-M.); (J.A.-S.); (L.F.G.-H.); (J.C.-M.); (T.I.C.-B.); (D.R.-R.); (A.G.-S.)
| | - Alejandra Guillermina Miranda-Díaz
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Mexico; (E.G.C.-M.); (J.A.-S.); (L.F.G.-H.); (J.C.-M.); (T.I.C.-B.); (D.R.-R.); (A.G.-S.)
| |
Collapse
|
5
|
Tian J, Kang S, Zhang D, Huang Y, Yao X, Zhao M, Lu Q. Selection of indicators reporting response rate in pharmaceutical trials for systemic lupus erythematosus: preference and relative sensitivity. Lupus Sci Med 2023; 10:e000942. [PMID: 37798046 PMCID: PMC10565300 DOI: 10.1136/lupus-2023-000942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVE SLE is a common multisystem autoimmune disease with chronic inflammation. Many efficacy evaluation indicators of randomised clinical trials (RCTs) for SLE have been proposed but the comparability remains unknown. We aim to explore the preference and comparability of indicators reporting response rate and provide basis for primary outcome selection when evaluating the efficacy of SLE pharmaceutical treatment. METHODS We systematically searched three databases and three registries to identify pharmacological intervention-controlled SLE RCTs. Relative discriminations between indicators were assessed by the Bayesian hierarchical linear mixed model. RESULTS 33 RCTs met our inclusion criteria and we compared eight of the most commonly used indicators reporting response rate. SLE Disease Activity Index 4 (SLEDAI-4) and SLE Responder Index 4 were considered the best recommended indicators reporting response rate to discriminate the pharmacological efficacy. Indicator preference was altered by disease severity, classification of drugs and outcome of trials, but SLEDAI-4 had robust efficacy in discriminating ability for most interventions. Of note, BILAG Index-based Combined Lupus Assessment showed efficacy in trials covering all-severity patients, as well as non-biologics RCTs. The British Isles Lupus Assessment Group response and Physician's Global Assessment response were more cautious in evaluating disease changes. Serious adverse event was often applied to evaluate the safety and tolerability of treatments rather than efficacy. CONCLUSIONS The impressionable efficacy discrimination ability of indicators highlights the importance of flexibility and comprehensiveness when choosing primary outcome(s). As for trials that are only evaluated by SLEDAI-4, attention should be paid to outcome interpretation to avoid the exaggeration of treatment efficacy. Further subgroup analyses are limited by the number of included RCTs. PROSPERO REGISTRATION NUMBER CRD42022334517.
Collapse
Affiliation(s)
- Jingru Tian
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Shuntong Kang
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Dingyao Zhang
- Graduate Program in Biological and Biomedical Sciences, Yale University, New Haven, Connecticut, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA
| | - Yaqing Huang
- Department of Pathology, Yale University, New Haven, Connecticut, USA
| | - Xu Yao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Ming Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Khalili L, Tang W, Askanase AD. Lupus clinical trials and the promise of future therapies. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2023; 4:109-114. [PMID: 37781678 PMCID: PMC10538598 DOI: 10.2478/rir-2023-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 08/18/2023] [Indexed: 10/03/2023]
Affiliation(s)
- Leila Khalili
- Columbia University Irving Medical Center, New York , NY, USA
| | - Wei Tang
- Columbia University Irving Medical Center, New York , NY, USA
| | | |
Collapse
|
7
|
David T, Su L, Cheng Y, Gordon C, Parker B, Isenberg D, Reynolds JA, Bruce IN. Predictors of British Isles Lupus Assessment Group-based outcomes in patients with systemic lupus erythematosus: Analysis from the Systemic Lupus International Collaborating Clinics Inception Cohort. Lupus 2023; 32:1043-1055. [PMID: 37463793 PMCID: PMC7614893 DOI: 10.1177/09612033231183273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
BACKGROUND We aimed to identify factors associated with a significant reduction in SLE disease activity over 12 months assessed by the BILAG Index. METHODS In an international SLE cohort, we studied patients from their 'inception enrolment' visit. We also defined an 'active disease' cohort of patients who had active disease similar to that needed for enrolment into clinical trials. Outcomes at 12 months were; Major Clinical Response (MCR: reduction to classic BILAG C in all domains, steroid dose of ≤7.5 mg and SLEDAI ≤ 4) and 'Improvement' (reduction to ≤1B score in previously active organs; no new BILAG A/B; stable or reduced steroid dose; no increase in SLEDAI). Univariate and multivariate logistic regression with Least Absolute Shrinkage and Selection Operator (LASSO) and cross-validation in randomly split samples were used to build prediction models. RESULTS 'Inception enrolment' (n = 1492) and 'active disease' (n = 924) patients were studied. Models for MCR performed well (ROC AUC = .777 and .732 in the inception enrolment and active disease cohorts, respectively). Models for Improvement performed poorly (ROC AUC = .574 in the active disease cohort). MCR in both cohorts was associated with anti-malarial use and inversely associated with active disease at baseline (BILAG or SLEDAI) scores, BILAG haematological A/B scores, higher steroid dose and immunosuppressive use. CONCLUSION Baseline predictors of response in SLE can help identify patients in clinic who are less likely to respond to standard therapy. They are also important as stratification factors when designing clinical trials in order to better standardize overall usual care response rates.
Collapse
Affiliation(s)
- Trixy David
- The Kellgren Centre for Rheumatology, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Li Su
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Yafeng Cheng
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Caroline Gordon
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Benjamin Parker
- The Kellgren Centre for Rheumatology, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - David Isenberg
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - John A Reynolds
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- Rheumatology Department, City Hospital, Sandwell and West Birmingham NHS Trust, Birmingham, UK
| | - Ian N Bruce
- The Kellgren Centre for Rheumatology, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Centre for Musculoskeletal Research, The University of Manchester, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, Manchester, UK
| | | |
Collapse
|
8
|
Schmidtmann I, Kraus D, Weinmann A, Pütz K, Claßen P, Schleicher EM, Boedecker-Lips SC, Weinmann-Menke J. Validation of the 2019 EULAR/ACR classification criteria for systemic lupus erythematosus in an academic tertiary care centre. RMD Open 2023; 9:e003037. [PMID: 37419524 DOI: 10.1136/rmdopen-2023-003037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/22/2023] [Indexed: 07/09/2023] Open
Abstract
OBJECTIVES To assess the sensitivity and specificity of the 2019 EULAR/American College of Rheumatology (ACR) classification criteria for systemic lupus erythematosus (SLE) in outpatients at an academic tertiary care centre and to compare them to the 1997 ACR and the 2012 Systemic Lupus International Collaborating Clinics criteria. METHODS Prospective and retrospective observational cohort study. RESULTS 3377 patients were included: 606 with SLE, 1015 with non-SLE autoimmune-mediated rheumatic diseases (ARD) and 1756 with non-ARD diseases (hepatocellular carcinoma, primary biliary cirrhosis, autoimmune hepatitis). The 2019 criteria were more sensitive than the 1997 criteria (87.0% vs 81.8%), but less specific (98.1% vs 99.5% in the entire cohort and 96.5% vs 98.8% in patients with non-SLE ARD), resulting in Youden Indexes for patients with SLE/non-SLE ARD of 0.835 and 0.806, respectively. The most sensitive items were history of antinuclear antibody (ANA) positivity and detection of anti-double-stranded deoxyribonucleic acid (dsDNA) antibodies. These were also the least specific items. The most specific items were class III/IV lupus nephritis and the combination of low C3 and low C4 complement levels, followed by class II/V lupus nephritis, either low C3 or low C4 complement levels, delirium and psychosis, when these were not attributable to non-SLE causes. CONCLUSIONS In this cohort from an independent academic medical centre, the sensitivity and specificity of the 2019 lupus classification criteria were confirmed. Overall agreement of the 1997 and the 2019 criteria was very good.
Collapse
Affiliation(s)
- Irene Schmidtmann
- Institute for Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Daniel Kraus
- 1st Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Arndt Weinmann
- 1st Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Katharina Pütz
- 1st Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Paul Claßen
- 1st Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Eva Maria Schleicher
- 1st Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Julia Weinmann-Menke
- 1st Department of Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
9
|
Garces S, Karis E, Merrill JT, Askanase AD, Kalunian K, Mo M, Milmont CE. Improving resource utilisation in SLE drug development through innovative trial design. Lupus Sci Med 2023; 10:e000890. [PMID: 37491104 PMCID: PMC10373732 DOI: 10.1136/lupus-2022-000890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/29/2023] [Indexed: 07/27/2023]
Abstract
SLE is a complex autoimmune disease with considerable unmet need. Numerous clinical trials designed to investigate novel therapies are actively enrolling patients straining limited resources and creating inefficiencies that increase enrolment challenges. This has motivated investigators developing novel drugs and treatment strategies to consider innovative trial designs that aim to improve the efficiency of generating evidence; these strategies propose conducting fewer trials, involving smaller numbers of patients, while maintaining scientific rigour in safety and efficacy data collection and analysis. In this review we present the design of two innovative phase IIb studies investigating efavaleukin alfa and rozibafusp alfa for the treatment of SLE which use an adaptive study design. This design was selected as a case study, investigating efavaleukin alfa, in the Food and Drug Administration's Complex Innovative Trial Design Pilot Program. The adaptive design approach includes prospectively planned modifications at predefined interim timepoints. Interim assessments of futility allow for a trial to end early when the investigational therapy is unlikely to provide meaningful treatment benefits to patients, which can release eligible patients to participate in other-potentially more promising-trials, or seek alternative treatments. Response-adaptive randomisation allows randomisation ratios to change based on accumulating data, in favour of the more efficacious dose arm(s), while the study is ongoing. Throughout the trial the placebo arm allocation ratio is maintained constant. These design elements can improve the statistical power in the estimation of treatment effect and increase the amount of safety and efficacy data collected for the optimal dose(s). Furthermore, these trials can provide the required evidence to potentially serve as one of two confirmatory trials needed for regulatory approval. This can reduce the need for multiple phase III trials, the total patient requirements, person-exposure risk, and ultimately the time and cost of investigational drug development programmes.
Collapse
Affiliation(s)
| | | | - Joan T Merrill
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Anca D Askanase
- Department of Rheumatology, Columbia University Irving Medical Center, New York (City), New York, USA
| | - Kenneth Kalunian
- Division of Rheumatology, Allergy and Immunology, University of California San Diego School of Medicine, San Diego, California, USA
| | - May Mo
- Amgen Inc, Thousand Oaks, California, USA
| | | |
Collapse
|
10
|
Askanase AD, Tang W, Zuraw Q, Gordon R, Brotherton B, Merrill JT. Evaluation of the LFA-REAL clinician-reported outcome (ClinRO) and patient-reported outcome (PRO): prespecified analysis of the phase III ustekinumab trial in patients with SLE. Lupus Sci Med 2023; 10:10/1/e000875. [PMID: 37012059 PMCID: PMC10083883 DOI: 10.1136/lupus-2022-000875] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/16/2023] [Indexed: 04/05/2023]
Abstract
OBJECTIVE The Lupus Foundation of America Rapid Evaluation of Activity in Lupus (LFA-REAL) system is a novel and simple SLE disease activity instrument, consisting of a tandem clinician-reported (ClinRO) and patient-reported (PRO) outcome measure. The aim of this study was to compare the LFA-REAL system with other SLE activity measures in the phase III trial of ustekinumab in patients with active SLE. METHODS This was a prespecified analysis of data from a randomised, double-blind, placebo-controlled, parallel-group trial conducted at 140 sites in 20 countries. Correlations were evaluated between the LFA-REAL ClinRO and PRO with a panel of clinician-reported and patient-reported disease activity measures commonly used in SLE clinical trials at baseline, week 24 and week 52. All p values are reported as nominal. RESULTS Trial participants included 516 patients with SLE with a mean (SD) age of 43.5 (8.9), of whom 482 (93.4%) were female. The LFA-REAL ClinRO correlated with Physician Global Assessment (r=0.39, 0.65 and 0.74, p<0.001), British Isles Lupus Assessment Group Index (r=0.43, 0.67 and 0.73, p<0.001) and SLE Disease Activity Index-2000 (r=0.35, 0.60 and 0.62, p<0.001). The LFA-REAL ClinRO arthralgia/arthritis score correlated well with active joint counts (r=0.54, 0.73 and 0.68, p<0.001) and the mucocutaneous global score correlated strongly with Cutaneous Lupus Erythematosus Disease Area and Severity Index total activity (r=0.57, 0.77 and 0.81, p<0.001). The LFA-REAL PRO demonstrated a moderate correlation with Functional Assessment of Chronic Illness Therapy-Fatigue (r=-0.60, -0.55 and -0.58, p<0.001), Lupus QoL physical health (r=-0.42, -0.47 and -0.46, p<0.001), SF-36v2 vitality (r=-0.40, -0.43 and -0.58, p<0.001) and SF-36v2 Physical Component Summary (r=-0.45, -0.53 and -0.53, p<0.001). The LFA-REAL ClinRO and PRO showed a moderate correlation with each other (r=0.32, 0.45 and 0.50, p<0.001). CONCLUSIONS The LFA-REAL ClinRO and PRO showed varied levels of correlations (weak to strong) with existing physician-based lupus disease activity measures and patient-reported outcome instruments, respectively and were able to more accurately capture organ-specific mucocutaneous and musculoskeletal manifestations. More analyses are needed to determine areas in which patient-reported outcomes are most similar or different to physician-reported end points and the basis for differences.
Collapse
Affiliation(s)
- Anca D Askanase
- Division of Rheumatology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Wei Tang
- Department of Medicine, Westchester Medical Center, Valhalla, New York, USA
| | - Qing Zuraw
- Clinical Development Immunology, Janssen Research and Development LLC, Spring House, Pennsylvania, USA
| | - Robert Gordon
- Data Science, IQVIA Inc, Durham, North Carolina, USA
| | | | - Joan T Merrill
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| |
Collapse
|
11
|
Sun Y, Nascimento Da Conceicao V, Chauhan A, Sukumaran P, Chauhan P, Ambrus JL, Vissink A, Kroese FGM, Muniswamy M, Mishra BB, Singh BB. Targeting alarmin release reverses Sjogren's syndrome phenotype by revitalizing Ca 2+ signalling. Clin Transl Med 2023; 13:e1228. [PMID: 37006181 PMCID: PMC10068318 DOI: 10.1002/ctm2.1228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Primary Sjogren's syndrome (pSS) is a systemic autoimmune disease that is embodied by the loss of salivary gland function and immune cell infiltration, but the mechanism(s) are still unknown. The aim of this study was to understand the mechanisms and identify key factors that leads to the development and progression of pSS. METHODS Immunohistochemistry staining, FACS analysis and cytokine levels were used to detect immune cells infiltration and activation in salivary glands. RNA sequencing was performed to identify the molecular mechanisms involved in the development of pSS. The function assays include in vivo saliva collection along with calcium imaging and electrophysiology on isolated salivary gland cells in mice models of pSS. Western blotting, real-time PCR, alarmin release, and immunohistochemistry was performed to identify the channels involved in salivary function in pSS. RESULTS We provide evidence that loss of Ca2+ signaling precedes a decrease in saliva secretion and/or immune cell infiltration in IL14α, a mouse model for pSS. We also showed that Ca2+ homeostasis was mediated by transient receptor potential canonical-1 (TRPC1) channels and inhibition of TRPC1, resulting in the loss of salivary acinar cells, which promoted alarmin release essential for immune cell infiltration/release of pro-inflammatory cytokines. In addition, both IL14α and samples from human pSS patients showed a decrease in TRPC1 expression and increased acinar cell death. Finally, paquinimod treatment in IL14α restored Ca2+ homeostasis that inhibited alarmin release thereby reverting the pSS phenotype. CONCLUSIONS These results indicate that loss of Ca2+ signaling is one of the initial factors, which induces loss of salivary gland function along with immune infiltration that exaggerates pSS. Importantly, restoration of Ca2+ signaling upon paquinimod treatment reversed the pSS phenotype thereby inhibiting the progressive development of pSS.
Collapse
Affiliation(s)
- Yuyang Sun
- Department of PeriodonticsSchool of DentistryUniversity of Texas Health Science Center San AntonioSan AntonioTexasUSA
| | | | - Arun Chauhan
- Department of Developmental DentistrySchool of DentistryUniversity of Texas Health Science Center San AntonioSan AntonioTexasUSA
| | - Pramod Sukumaran
- Department of PeriodonticsSchool of DentistryUniversity of Texas Health Science Center San AntonioSan AntonioTexasUSA
| | - Pooja Chauhan
- Department of Developmental DentistrySchool of DentistryUniversity of Texas Health Science Center San AntonioSan AntonioTexasUSA
| | - Julian L. Ambrus
- Division of Allergy, Immunology, and RheumatologyDepartment of MedicineSchool of Medicine and Biomedical SciencesState University of New YorkBuffaloNew YorkUSA
| | - Arjan Vissink
- Department of Oral and Maxillofacial SurgeryUniversity of Groningen and University Medical Center GroningenGroningenThe Netherlands
| | - Frans G. M. Kroese
- Department of Rheumatology and Clinical ImmunologyUniversity of Groningen and University Medical Center GroningenGroningenThe Netherlands
| | - Madesh Muniswamy
- Department of MedicineUniversity of Texas Health Science Center San AntonioSan AntonioTexasUSA
| | - Bibhuti B. Mishra
- Department of Developmental DentistrySchool of DentistryUniversity of Texas Health Science Center San AntonioSan AntonioTexasUSA
- Department of Biomedical SciencesSchool of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Brij B. Singh
- Department of PeriodonticsSchool of DentistryUniversity of Texas Health Science Center San AntonioSan AntonioTexasUSA
| |
Collapse
|
12
|
Gavan SP, Bruce IN, Payne K. Valuing Health Gain from Composite Response Endpoints for Multisystem Diseases. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:115-122. [PMID: 36008224 DOI: 10.1016/j.jval.2022.07.001] [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: 02/03/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES This study aimed to demonstrate how to estimate the value of health gain after patients with a multisystem disease achieve a condition-specific composite response endpoint. METHODS Data from patients treated in routine practice with an exemplar multisystem disease (systemic lupus erythematosus) were extracted from a national register (British Isles Lupus Assessment Group Biologics Register). Two bespoke composite response endpoints (Major Clinical Response and Improvement) were developed in advance of this study. Difference-in-differences regression compared health utility values (3-level version of EQ-5D; UK tariff) over 6 months for responders and nonresponders. Bootstrapped regression estimated the incremental quality-adjusted life-years (QALYs), probability of QALY gain after achieving the response criteria, and population monetary benefit of response. RESULTS Within the sample (n = 171), 18.2% achieved Major Clinical Response and 49.1% achieved Improvement at 6 months. Incremental health utility values were 0.0923 for Major Clinical Response and 0.0454 for Improvement. Expected incremental QALY gain at 6 months was 0.020 for Major Clinical Response and 0.012 for Improvement. Probability of QALY gain after achieving the response criteria was 77.6% for Major Clinical Response and 72.7% for Improvement. Population monetary benefit of response was £1 106 458 for Major Clinical Response and £649 134 for Improvement. CONCLUSIONS Bespoke composite response endpoints are becoming more common to measure treatment response for multisystem diseases in trials and observational studies. Health technology assessment agencies face a growing challenge to establish whether these endpoints correspond with improved health gain. Health utility values can generate this evidence to enhance the usefulness of composite response endpoints for health technology assessment, decision making, and economic evaluation.
Collapse
Affiliation(s)
- Sean P Gavan
- Manchester Centre for Health Economics, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, England, UK.
| | - Ian N Bruce
- Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, England, UK; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, England, UK
| | - Katherine Payne
- Manchester Centre for Health Economics, Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, England, UK
| |
Collapse
|
13
|
Zhang W, Zhao H, Du P, Cui H, Lu S, Xiang Z, Lu Q, Jia S, Zhao M. Integration of metabolomics and lipidomics reveals serum biomarkers for systemic lupus erythematosus with different organs involvement. Clin Immunol 2022; 241:109057. [PMID: 35667550 DOI: 10.1016/j.clim.2022.109057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/22/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that affects various organs or systems. We performed metabolomic and lipidomic profiles analyses of 133 SLE patients and 30 HCs. Differential metabolites and lipids were integrated, and then the biomarker panel was identified using binary logistic regression. We found that a combination of four metabolites or lipids could distinguish SLE from HC with an AUC of 0.998. Three lipids were combined to differentiate inactive SLE and active SLE. The AUC was 0.767. In addition, we also identified the biomarkers for different organ phenotypes of SLE. The AUCs for diagnosing SLE patients with only kidney involvement, skin involvement, blood system involvement, and multisystem involvement were 0.766, 0.718, 0.951, and 0.909, respectively. Our study succeeded in identifying biomarkers associated with different clinical phenotypes in SLE patients, which could facilitate a more precise diagnosis and assessment of disease progression in SLE.
Collapse
Affiliation(s)
- Wenqian Zhang
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Hongjun Zhao
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pei Du
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha 410011, China
| | - Haobo Cui
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Shuang Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha 410011, China
| | - Zhongyuan Xiang
- Department of Clinical Laboratory, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Sujie Jia
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha 410011, China.
| |
Collapse
|
14
|
Ozaki T, Kamiyama N, Saechue B, Soga Y, Gotoh R, Nakayama T, Fukuda C, Dewayani A, Chalalai T, Ariki S, Ozaka S, Sonoda A, Hirose H, Gendo Y, Noguchi K, Sachi N, Hidano S, Maeshima K, Gotoh K, Masaki T, Ishii K, Osada Y, Shibata H, Kobayashi T. Comprehensive lipidomics of lupus-prone mice using LC-MS/MS identifies the reduction of palmitoylethanolamide that suppresses TLR9-mediated inflammation. Genes Cells 2022; 27:493-504. [PMID: 35485445 DOI: 10.1111/gtc.12944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/21/2022] [Accepted: 04/23/2022] [Indexed: 11/28/2022]
Abstract
Lipid mediators are known to play crucial roles not only in the onset of the inflammatory response but also in the induction of resolution of inflammation. Here we report that palmitoylethanolamide (PEA), an endogenous N-acylethanolamine, can suppress the inflammation induced by Toll-like receptor (TLR) signaling both in vitro and in vivo. PEA was found to be significantly reduced in the serum and spleen of lupus-prone MRL/lpr mice analyzed by lipidomics. PEA suppressed pro-inflammatory cytokine production in a mouse macrophage cell line stimulated with TLR ligands such as lipopolysaccharide, peptidoglycan, poly (I:C), imiquimod and CpG-ODN. PEA also inhibited both mRNA and protein levels of IL-6 in bone marrow derived dendritic cells (BMDCs) and B cells stimulated with CpG-ODN. Augmentation of cell surface CD86 and CD40 on BMDCs and B cells, IgM production and cell proliferation of B cells in response to CpG-ODN were attenuated by PEA. Moreover, PEA treatment significantly reduced mortality and serum IL-6 levels in mice injected with CpG-ODN plus D-galactosamine. Taken together, PEA ameliorates inflammation induced by TLR signaling, which could be a novel therapeutic target for inflammatory disorders. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Takashi Ozaki
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan.,Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Naganori Kamiyama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Benjawan Saechue
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Yasuhiro Soga
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Ryo Gotoh
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Tatsuya Nakayama
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Chiaki Fukuda
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Astri Dewayani
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Thanyakorn Chalalai
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Shimpei Ariki
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Sotaro Ozaka
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Akira Sonoda
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Haruna Hirose
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Yoshiko Gendo
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Kaori Noguchi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Nozomi Sachi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Shinya Hidano
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| | - Keisuke Maeshima
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Koro Gotoh
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Takayuki Masaki
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Koji Ishii
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Yoshio Osada
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Takashi Kobayashi
- Department of Infectious Disease Control, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|
15
|
Tsang-A-Sjoe MWP, Bultink IEM. New developments in systemic lupus erythematosus. Rheumatology (Oxford) 2021; 60:vi21-vi28. [PMID: 34951924 PMCID: PMC8709564 DOI: 10.1093/rheumatology/keab498] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
In this review, the results of recent and ongoing clinical trials in patients with SLE are discussed. After many unsuccessful trials in the past decade, belimumab was the first biologic specifically designed for SLE that met its primary end point. At the same time, studies on the pathophysiology of SLE have further elucidated the pathways involved in the disease, which has led to the identification of new possible therapeutics and has encouraged the initiation of new trials. These new drugs include biologics that target B cells, T cells and type 1 interferons, and small molecules that inhibit kinases. Other therapeutics aim to restore immunological balance by restoring tolerance. Results from phase II and even phase III trials are promising and it is likely that some of the therapeutics discussed will receive approval in the following years. Hopefully, this will allow for more tailor-made medicine for SLE patients in the future.
Collapse
Affiliation(s)
- Michel W. P. Tsang-A-Sjoe
- Department of Rheumatology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Irene E. M. Bultink
- Department of Rheumatology, Amsterdam Rheumatology and immunology Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Wu D, Ai L, Sun Y, Yang B, Chen S, Wang Q, Kuang H. Role of NLRP3 Inflammasome in Lupus Nephritis and Therapeutic Targeting by Phytochemicals. Front Pharmacol 2021; 12:621300. [PMID: 34489689 PMCID: PMC8417800 DOI: 10.3389/fphar.2021.621300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multisystem autoimmune inflammatory condition that affects multiple organs and provokes extensive and severe clinical manifestations. Lupus nephritis (LN) is one of the main clinical manifestations of SLE. It refers to the deposition of immune complexes in the glomeruli, which cause kidney inflammation. Although LN seriously affects prognosis and represents a key factor of disability and death in SLE patients, its mechanism remains unclear. The NACHT, leucine-rich repeat (LRR), and pyrin (PYD) domains-containing protein 3 (NLRP3) inflammasome regulates IL-1β and IL-18 secretion and gasdermin D-mediated pyroptosis and plays a key role in innate immunity. There is increasing evidence that aberrant activation of the NLRP3 inflammasome and downstream inflammatory pathways play an important part in the pathogenesis of multiple autoimmune diseases, including LN. This review summarizes research progress on the elucidation of NLRP3 activation, regulation, and recent clinical trials and experimental studies implicating the NLRP3 inflammasome in the pathophysiology of LN. Current treatments fail to provide durable remission and provoke several sides effects, mainly due to their broad immunosuppressive effects. Therefore, the identification of a safe and effective therapeutic approach for LN is of great significance. Phytochemicals are found in many herbs, fruits, and vegetables and are secondary metabolites of plants. Evidence suggests that phytochemicals have broad biological activities and have good prospects in a variety of diseases, including LN. Therefore, this review reports on current research evaluating phytochemicals for targeting NLRP3 inflammasome pathways in LN therapy.
Collapse
Affiliation(s)
- Dantong Wu
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, Harbin, China.,Department of Laboratory Diagnostics, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lianjie Ai
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanping Sun
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bingyou Yang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, Harbin, China
| | - Sisi Chen
- Department of Rheumatology, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiuhong Wang
- Department of Natural Medicinal Chemistry, College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Haixue Kuang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
17
|
Connelly K, Golder V, Kandane-Rathnayake R, Morand EF. Clinician-reported outcome measures in lupus trials: a problem worth solving. THE LANCET. RHEUMATOLOGY 2021; 3:e595-e603. [PMID: 38287623 DOI: 10.1016/s2665-9913(21)00119-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 02/08/2023]
Abstract
Systemic lupus erythematosus (SLE) remains a disease of high unmet clinical need. Because of substantial patient heterogeneity, the execution of clinical trials that successfully determine the efficacy of novel therapeutics compared with placebo is a continuous challenge. Clinician-reported outcome measures of treatment response used in SLE trials have evolved from the use of individual disease activity indices, including the SLE Disease Activity Index (SLEDAI) and British Isles Lupus Assessment Group (BILAG), to composite responder definitions such as the SLE Responder Index (SRI) and BILAG-Based Composite Lupus Assessment (BICLA), which are based on these indices. However, these approaches have notable drawbacks and defining the optimal clinical trial outcome measure for SLE remains a research goal. In this Viewpoint, we explore the strengths and limitations of existing indices and composite assessments, illustrating features which should be investigated in future analysis of trial data. Further, we provide a platform from which to advance new approaches to endpoint design, which is crucial to improve the interpretability and success of subsequent clinical trials in SLE.
Collapse
Affiliation(s)
- Kathryn Connelly
- School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia; Department of Rheumatology, Monash Health, Monash University, Clayton, VIC 3168, Australia
| | - Vera Golder
- School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia; Department of Rheumatology, Monash Health, Monash University, Clayton, VIC 3168, Australia
| | | | - Eric F Morand
- School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia; Department of Rheumatology, Monash Health, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
18
|
Reynolds JA, Prattley J, Geifman N, Lunt M, Gordon C, Bruce IN. Distinct patterns of disease activity over time in patients with active SLE revealed using latent class trajectory models. Arthritis Res Ther 2021; 23:203. [PMID: 34321096 PMCID: PMC8320218 DOI: 10.1186/s13075-021-02584-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/10/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a heterogeneous systemic autoimmune condition for which there are limited licensed therapies. Clinical trial design is challenging in SLE due at least in part to imperfect outcome measures. Improved understanding of how disease activity changes over time could inform future trial design. The aim of this study was to determine whether distinct trajectories of disease activity over time occur in patients with active SLE within a clinical trial setting and to identify factors associated with these trajectories. METHODS Latent class trajectory models were fitted to a clinical trial dataset of a monoclonal antibody targeting CD22 (Epratuzumab) in patients with active SLE using the numerical BILAG-2004 score (nBILAG). The baseline characteristics of patients in each class and changes in prednisolone over time were identified. Exploratory PK-PD modelling was used to examine cumulative drug exposure in relation to latent class membership. RESULTS Five trajectories of disease activity were identified, with 3 principal classes: non-responders (NR), slow responders (SR) and rapid-responders (RR). In both the SR and RR groups, significant changes in disease activity were evident within the first 90 days of the trial. The SR and RR patients had significantly higher baseline disease activity, exposure to epratuzumab and activity in specific BILAG domains, whilst NR had lower steroid use at baseline and less change in steroid dose early in the trial. CONCLUSIONS Longitudinal nBILAG scores reveal different trajectories of disease activity and may offer advantages over fixed endpoints. Corticosteroid use however remains an important confounder in lupus trials and can influence early response. Changes in disease activity and steroid dose early in the trial were associated with the overall disease activity trajectory, supporting the feasibility of performing adaptive trial designs in SLE.
Collapse
Affiliation(s)
- John A Reynolds
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Rheumatology Department, Sandwell and West Birmingham NHS Trust, Birmingham, UK
| | - Jennifer Prattley
- Centre for Epidemiology Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Nophar Geifman
- Centre for Health Informatics, Division of Informatics, Imaging & Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Mark Lunt
- Centre for Epidemiology Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Caroline Gordon
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Rheumatology Department, Sandwell and West Birmingham NHS Trust, Birmingham, UK
| | - Ian N Bruce
- Centre for Epidemiology Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
- Manchester University NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester, Greater Manchester, UK.
| |
Collapse
|
19
|
Yavuz S, Lipsky PE. Current Status of the Evaluation and Management of Lupus Patients and Future Prospects. Front Med (Lausanne) 2021; 8:682544. [PMID: 34124113 PMCID: PMC8193052 DOI: 10.3389/fmed.2021.682544] [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: 03/18/2021] [Accepted: 04/27/2021] [Indexed: 12/24/2022] Open
Abstract
The vastly diverse nature of systemic lupus erythematosus (SLE) poses great challenges to clinicians and patients, as well as to research and drug development efforts. Precise management of lupus patients would be advanced by the ability to identify specific abnormalities operative in individual patients at the time of encounter with the clinician. Advances in new technologies and bioinformatics have greatly improved the understanding of the pathophysiology of SLE. Recent research has focused on the discovery and classification of sensitive and specific markers that could aid early accurate diagnosis, better monitoring of disease and identification of appropriate therapy choices based on specific dysregulated molecular pathways. Here, we summarize some of the advances and discuss the challenges in moving toward precise patient-centric management modalities in SLE.
Collapse
Affiliation(s)
- Sule Yavuz
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Peter E Lipsky
- Ampel BioSolutions and Re-Imagine Lupus Investigation, Treatment and Education Research Institute, Charlottesville, VA, United States
| |
Collapse
|
20
|
Furie RA, Bruce IN, Dörner T, Leon MG, Leszczyński P, Urowitz M, Haier B, Jimenez T, Brittain C, Liu J, Barbey C, Stach C. Phase 2, Randomized, Placebo-Controlled Trial of Dapirolizumab Pegol in Patients with Moderate-to-Severe Active Systemic Lupus Erythematosus. Rheumatology (Oxford) 2021; 60:5397-5407. [PMID: 33956056 PMCID: PMC9194804 DOI: 10.1093/rheumatology/keab381] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/14/2021] [Indexed: 11/18/2022] Open
Abstract
Objective To evaluate the dose–response, efficacy and safety of dapirolizumab pegol (DZP) in
patients with SLE. Methods Adults with moderately to severely active SLE (SLEDAI-2K score ≥6 and ≥1 BILAG A or ≥2
BILAG B domain scores), receiving stable CS (≤40 mg/day prednisone-equivalent),
antimalarial or immunosuppressant drugs were included. Patients with stable LN
(proteinuria ≤2 g/day) not receiving high-dose CS or CYC were permitted entry.
Randomized patients received placebo or i.v. DZP (6/24/45 mg/kg) and standard-of-care
(SOC) treatment every 4 weeks to week 24, after which patients received only SOC to week
48. The primary objective was to establish a dose–response relationship based on week 24
BILAG-Based Composite Lupus Assessment (BICLA) responder rates. Results All DZP groups exhibited improvements in clinical and immunological outcomes
vs placebo at week 24; however, BICLA responder rates did not fit
pre-specified dose–response models [best-fitting model
(Emax): P = 0.07]. Incidences of serious
treatment-emergent adverse events across DZP groups were low and similar to placebo.
Following DZP withdrawal, SLEDAI-2K, physician’s global assessment (PGA), BILAG, and
Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) scores stabilized;
BICLA and SLE Responder Index (SRI-4) responder rates declined (likely due to
interventions with disallowed escape medications), BILAG flares increased, and
immunologic parameters returned towards baseline. Conclusions Although the primary objective was not met, DZP appeared to be well tolerated, and
patients exhibited improvements across multiple clinical and immunological measures of
disease activity after 24 weeks relative to placebo. The potential clinical benefit of
DZP warrants further investigation.
Collapse
Affiliation(s)
| | - Ian N Bruce
- NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Trust, Manchester, UK.,Centre for Musculoskeletal Research, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Fanouriakis A, Tziolos N, Bertsias G, Boumpas DT. Update οn the diagnosis and management of systemic lupus erythematosus. Ann Rheum Dis 2021; 80:14-25. [PMID: 33051219 DOI: 10.1136/annrheumdis-2020-218272] [Citation(s) in RCA: 391] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022]
Abstract
Clinical heterogeneity, unpredictable course and flares are characteristics of systemic lupus erythematosus (SLE). Although SLE is-by and large-a systemic disease, occasionally it can be organ-dominant, posing diagnostic challenges. To date, diagnosis of SLE remains clinical with a few cases being negative for serologic tests. Diagnostic criteria are not available and classification criteria are often used for diagnosis, yet with significant caveats. Newer sets of criteria (European League Against Rheumatism (EULAR)/American College of Rheumatology (ACR) 2019) enable earlier and more accurate classification of SLE. Several disease endotypes have been recognised over the years. There is increased recognition of milder cases at presentation, but almost half of them progress overtime to more severe disease. Approximately 70% of patients follow a relapsing-remitting course, the remaining divided equally between a prolonged remission and a persistently active disease. Treatment goals include long-term patient survival, prevention of flares and organ damage, and optimisation of health-related quality of life. For organ-threatening or life-threatening SLE, treatment usually includes an initial period of high-intensity immunosuppressive therapy to control disease activity, followed by a longer period of less intensive therapy to consolidate response and prevent relapses. Management of disease-related and treatment-related comorbidities, especially infections and atherosclerosis, is of paramount importance. New disease-modifying conventional and biologic agents-used alone, in combination or sequentially-have improved rates of achieving both short-term and long-term treatment goals, including minimisation of glucocorticoid use.
Collapse
MESH Headings
- Anemia, Hemolytic, Autoimmune/physiopathology
- Anemia, Hemolytic, Autoimmune/therapy
- Antibodies, Monoclonal, Humanized/therapeutic use
- Autoantibodies/immunology
- Azathioprine/therapeutic use
- Calcineurin Inhibitors/therapeutic use
- Cardiovascular Diseases/epidemiology
- Cyclophosphamide/therapeutic use
- Disease Management
- Female
- Glucocorticoids/therapeutic use
- Heart Valve Diseases/physiopathology
- Heart Valve Diseases/therapy
- Humans
- Hydroxychloroquine/therapeutic use
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/therapy
- Immunosuppressive Agents/therapeutic use
- Lupus Erythematosus, Systemic/diagnosis
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/physiopathology
- Lupus Erythematosus, Systemic/therapy
- Lupus Nephritis/physiopathology
- Lupus Nephritis/therapy
- Lupus Vasculitis, Central Nervous System/physiopathology
- Lupus Vasculitis, Central Nervous System/therapy
- Macrophage Activation Syndrome/physiopathology
- Macrophage Activation Syndrome/therapy
- Methotrexate/therapeutic use
- Mycophenolic Acid/therapeutic use
- Myocarditis/physiopathology
- Myocarditis/therapy
- Outcome Assessment, Health Care
- Pericarditis/physiopathology
- Pericarditis/therapy
- Phenotype
- Pregnancy
- Pregnancy Complications/epidemiology
- Pregnancy Complications/physiopathology
- Pregnancy Complications/therapy
- Prognosis
- Purpura, Thrombocytopenic, Idiopathic/physiopathology
- Purpura, Thrombocytopenic, Idiopathic/therapy
- Quality of Life
- Recurrence
- Rituximab/therapeutic use
- Severity of Illness Index
- Survival Rate
- Uterine Cervical Neoplasms/epidemiology
Collapse
Affiliation(s)
| | - Nikolaos Tziolos
- 4th Department of Internal Medicine, "Attikon" University Hospital, Athens, Greece
| | - George Bertsias
- Rheumatology, Clinical Immunology and Allergy, University of Crete School of Medicine, Iraklio, Crete, Greece
- Laboratory of Autoimmunity-Inflammation, Institute of Molecular Biology and Biotechnology, Heraklion, Crete, Greece
| | - Dimitrios T Boumpas
- 4th Department of Internal Medicine, "Attikon" University Hospital, Athens, Greece
- Joint Rheumatology Program, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Medical School, University of Cyprus, Nicosia, Cyprus
- Laboratory of Autoimmunity and Inflammation, Biomedical Research Foundation of the Academy of Athens, Athens, Cyprus
| |
Collapse
|
22
|
Arnaud L, Tektonidou MG. Long-term outcomes in systemic lupus erythematosus: trends over time and major contributors. Rheumatology (Oxford) 2020; 59:v29-v38. [PMID: 33280012 PMCID: PMC7719040 DOI: 10.1093/rheumatology/keaa382] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/03/2020] [Indexed: 12/20/2022] Open
Abstract
SLE is a chronic autoimmune rheumatic disorder of high heterogeneity in clinical presentation, treatment response and prognosis. Long-term outcomes in SLE have been dramatically improved over the past decades, however, increased morbidity and mortality, especially among young individuals, still exists. Unmet needs include residual disease activity and frequent flares, glucocorticoid treatment dependency and toxicity, comorbidity burden, reduced health-related quality of life, health disparities and damage. The main determinants of long-term outcomes in SLE are age, sex, race/ethnicity, genetic profile, environmental factors including smoking, disease activity, major organ involvement such as lupus nephritis and CNS involvement, comorbidities including cardiovascular disease and serious infections, coexistence with APS, treatment adherence, socio-economic factors and access to care. In this review we discuss trends in long-term outcomes in SLE over the years and major contributors such as genetic, disease-related, treatment, comorbidity, socio-economic and other factors.
Collapse
Affiliation(s)
- Laurent Arnaud
- Department of Rheumatology, Hôpitaux Universitaires de Strasbourg, INSERM UMR-S 1109, Centre National de Référence des Maladies Auto-immunes Systémiques Rares (RESO), Strasbourg, France
| | - Maria G Tektonidou
- Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Joint Rheumatology Program, Laiko Hospital, Athens, Greece
| |
Collapse
|
23
|
Lee YH, Song GG. Anifrolumab for the treatment of active systemic lupus erythematosus: a meta-analysis of randomized controlled trials. Z Rheumatol 2020; 80:988-994. [PMID: 33216191 DOI: 10.1007/s00393-020-00928-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2020] [Indexed: 01/04/2023]
Abstract
We surveyed randomized controlled trials (RCTs) examining the efficacy and safety of anifrolumab 300 mg in patients with active systemic lupus erythematosus (SLE) despite receiving standard therapy, using MEDLINE, EMBASE, the Cochrane Controlled Trials Register, and manual searches. Meta-analysis performed to determine treatment efficacy and safety outcomes of three RCTs (459 patients and 468 controls) revealed that the BICLA responses were significantly higher in the anifrolumab group than in the placebo group (OR = 2.071, 95%CI 1.575-2.725, p < 0.001). Steroid reduction and CLASI reduction were also significantly higher in the anifrolumab group than in the placebo group (OR = 1.811, 95%CI = 1.308-2.506, p < 0.001; OR = 2.245, 95%CI = 1.437-3.506, p < 0.001). Compared with placebo, anifrolumab significantly increased the SRI7 and SRI8 responses in SLE patients (OR = 1.866, 95%CI = 1.372-2.536, p < 0.001; OR = 1.925, 95%CI = 1.387-2.672, p < 0.001). The SRI4, 5, and 6 responses also tended to be higher in the anifrolumab group than in the placebo group. Adverse event incidence was significantly higher in the anifrolumab group than in the placebo group (OR = 1.815, 95%CI = 1.262-2.611, p = 0.001); serious adverse events were significantly lower in the anifrolumab group than in the placebo group (OR = 0.679, 95%CI = 0.468-0.986, p = 0.042). Herpes zoster infection was significantly higher in the anifrolumab group than in the placebo group (OR = 4.089, 95%CI = 1.750-9.522, p = 0.001). Anifrolumab is effective for treating active SLE. However, anifrolumab increased the incidence of herpes zoster infection compared with placebo.
Collapse
Affiliation(s)
- Y H Lee
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, 02841, Seoul, Korea (Republic of).
| | - G G Song
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, 73, Goryeodae-ro, Seongbuk-gu, 02841, Seoul, Korea (Republic of)
| |
Collapse
|
24
|
Verstappen GM, Kroese FGM, Bootsma H. Stumbles in Sjögren's syndrome drug development: where to look for the next big leap? Expert Rev Clin Immunol 2020; 16:1043-1045. [PMID: 33196342 DOI: 10.1080/1744666x.2021.1831915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Gwenny M Verstappen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| | - Frans G M Kroese
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| | - Hendrika Bootsma
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen , Groningen, The Netherlands
| |
Collapse
|
25
|
Kim TH, Lee SS, Park W, Song YW, Suh CH, Kim S, Lee YN, Yoo DH. A 5-year Retrospective Analysis of Drug Survival, Safety, and Effectiveness of the Infliximab Biosimilar CT-P13 in Patients with Rheumatoid Arthritis and Ankylosing Spondylitis. Clin Drug Investig 2020; 40:541-553. [DOI: 10.1007/s40261-020-00907-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
26
|
Protecting the kidney in systemic lupus erythematosus: from diagnosis to therapy. Nat Rev Rheumatol 2020; 16:255-267. [PMID: 32203285 DOI: 10.1038/s41584-020-0401-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2020] [Indexed: 12/20/2022]
Abstract
Lupus nephritis (LN) is a common manifestation of systemic lupus erythematosus that can lead to irreversible renal impairment. Although the prognosis of LN has improved substantially over the past 50 years, outcomes have plateaued in the USA in the past 20 years as immunosuppressive therapies have failed to reverse disease in more than half of treated patients. This failure might reflect disease complexity and heterogeneity, as well as social and economic barriers to health-care access that can delay intervention until after damage has already occurred. LN progression is still poorly understood and involves multiple cell types and both immune and non-immune mechanisms. Single-cell analysis of intrinsic renal cells and infiltrating cells from patients with LN is a new approach that will help to define the pathways of renal injury at a cellular level. Although many new immune-modulating therapies are being tested in the clinic, the development of therapies to improve regeneration of the injured kidney and to prevent fibrosis requires a better understanding of the mechanisms of LN progression. This mechanistic understanding, together with the development of clinical measures to evaluate risk and detect early disease and better access to expert health-care providers, should improve outcomes for patients with LN.
Collapse
|
27
|
Giles JL, Polak OJ, Landon J. Disease modifying drugs for rheumatological diseases: a brief history of everything. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 120:313-348. [PMID: 32085884 DOI: 10.1016/bs.apcsb.2019.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The rheumatological diseases are a group of chronic, painful, degenerative and debilitating conditions with an increasing prevalence across the globe. The pathogenesis of these disorders is complex, overlapping and not fully understood. As such, it is difficult and time consuming to achieve correct diagnosis and complete remission for an individual patient. In this review we describe the most common forms of inflammatory arthritis and discuss how the management and treatment options for these rheumatic diseases have developed over time. We outline the successes and the limitations of current treatment regimens and discuss the economic burden of the current options. With advancements in understanding of disease mechanisms, we discuss the importance of the biologics revolution in the context of rheumatological disease and how the development of biosimilars and small molecule inhibitors will impact current treatment options in order to alleviate some of the cost burden of biological therapies. The ideal treatment strategy for the future would involve personalized and predictive medicine where by treatments can be tailored to an individual patient's needs in order to achieve fast and successful remission with no adverse events.
Collapse
Affiliation(s)
- Joanna L Giles
- MicroPharm Ltd, Newcastle Emlyn, Carmarthenshire, United Kingdom
| | - Oktawia J Polak
- MicroPharm Ltd, Newcastle Emlyn, Carmarthenshire, United Kingdom
| | - John Landon
- MicroPharm Ltd, Newcastle Emlyn, Carmarthenshire, United Kingdom
| |
Collapse
|
28
|
Toward better outcomes in Sjögren's syndrome: The promise of a stratified medicine approach. Best Pract Res Clin Rheumatol 2020; 34:101475. [PMID: 32005417 DOI: 10.1016/j.berh.2019.101475] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Sjögren's syndrome is a systemic autoimmune disease defined by its targeted inflammation of the salivary and lacrimal glands, resulting in dry mouth and eyes in the majority and persistent or recurrent salivary gland enlargement in a minority of those affected. Involvement of major organs, an increased risk of lymphoma, and autoantibodies against ubiquitous cellular ribonucleoproteins define some of its systemic features. Those affected have a high symptom burden and the development of disease-modifying therapies is thus an urgent need. A stratified medicine approach offers promise as a means of targeting specific therapies to patients for whom the mechanism of action is most relevant. Implementation of this approach will require an understanding of the pathophysiological processes underlying different patient subsets, and then identifying or developing a drug that targets this pathway. Such therapies would be most effective if implemented early in the disease course before the advent of adverse outcomes or glandular damage. This review will provide a disease overview followed by an analysis of the feasibility of a stratified medicine approach, focusing on the disease heterogeneity, predictors of disease progression and adverse outcomes, and recent advances in the development of relevant outcome measures and new therapies.
Collapse
|
29
|
Winthrop KL, Weinblatt ME, Bathon J, Burmester GR, Mease PJ, Crofford L, Bykerk V, Dougados M, Rosenbaum JT, Mariette X, Sieper J, Melchers F, Cronstein BN, Breedveld FC, Kalden J, Smolen JS, Furst D. Unmet need in rheumatology: reports from the Targeted Therapies meeting 2019. Ann Rheum Dis 2020; 79:88-93. [PMID: 31662322 PMCID: PMC6937409 DOI: 10.1136/annrheumdis-2019-216151] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/04/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVES To detail the greatest areas of unmet scientific and clinical needs in rheumatology. METHODS The 21st annual international Advances in Targeted Therapies meeting brought together more than 100 leading basic scientists and clinical researchers in rheumatology, immunology, epidemiology, molecular biology and other specialties. During the meeting, breakout sessions were convened, consisting of 5 disease-specific groups with 20-30 experts assigned to each group based on expertise. Specific groups included: rheumatoid arthritis, psoriatic arthritis, axial spondyloarthritis, systemic lupus erythematosus and other systemic autoimmune rheumatic diseases. In each group, experts were asked to identify unmet clinical and translational research needs in general and then to prioritise and detail the most important specific needs within each disease area. RESULTS Overarching themes across all disease states included the need to innovate clinical trial design with emphasis on studying patients with refractory disease, the development of trials that take into account disease endotypes and patients with overlapping inflammatory diseases, the need to better understand the prevalence and incidence of inflammatory diseases in developing regions of the world and ultimately to develop therapies that can cure inflammatory autoimmune diseases. CONCLUSIONS Unmet needs for new therapies and trial designs, particularly for those with treatment refractory disease, remain a top priority in rheumatology.
Collapse
Affiliation(s)
| | | | - Joan Bathon
- Columbia University, College of Physicians & Surgeons, New York City, New York, USA
| | | | - Philip J Mease
- Swedish Medical Center, University of Washington, Seattle, Washington, USA
| | | | - Vivian Bykerk
- Hospital for Special Surgery, New York City, New York, USA
| | | | - James Todd Rosenbaum
- Oregon Health Sciences University, Portland, Oregon, USA
- Legacy Devers Eye Institute, Portland, Oregon, USA
| | - Xavier Mariette
- Paris-Sud University, APHP Université Paris-Saclay, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Joachim Sieper
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité, Berlin, Germany
| | - Fritz Melchers
- Max Planck Institute for Infection Biology, Berlin, Germany
| | | | | | | | - Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of, Vienna, Vienna, Austria
| | - Daniel Furst
- Swedish Medical Center, University of Washington, Seattle, Washington, USA
- University of California, Los Angeles Medical Center, Los Angeles, CA, USA
- University of Florence, Florence, Italy
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW The management of lupus nephritis remains unsatisfactory due to insufficiently effective treatment regimens and the dearth of reliable predictors of disease onset or progression to guide individualized therapeutic decisions. This review summarizes new findings related to lupus nephritis over the last 18 months and discusses clinical needs that should be considered to advance trials of mechanism-based therapeutic strategies. RECENT FINDINGS Collaborative teams are addressing how to improve disease definitions and are developing predictive models for disease onset, disease response and risk of flare in individual patients. More attention is being paid to clinical trial design. Advanced technologic approaches are allowing the analysis of small amounts of human tissue and urine in unprecedented detail so as to discover new pathogenic mechanisms and identify disease biomarkers. Novel therapies continue to be tested in disease models and include new strategies to protect renal tissue from cell damage and fibrosis. SUMMARY The collaborative efforts of patients, clinical and translational researchers, the pharmaceutical industry and funding sources are needed to advance therapies for lupus nephritis. Specialized clinical centers can then deliver optimal and more personalized patient care that will improve patient outcomes.
Collapse
Affiliation(s)
- Anne Davidson
- Center for Autoimmunity, Musculoskeletal and Hematologic Diseases, Feinstein Institute for Medical Research, New York, New York, USA
| | | | | |
Collapse
|
31
|
Jackson SW, Davidson A. BAFF inhibition in SLE-Is tolerance restored? Immunol Rev 2019; 292:102-119. [PMID: 31562657 PMCID: PMC6935406 DOI: 10.1111/imr.12810] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023]
Abstract
The B cell activating factor (BAFF) inhibitor, belimumab, is the first biologic drug approved for the treatment of SLE, and exhibits modest, but durable, efficacy in decreasing disease flares and organ damage. BAFF and its homolog APRIL are TNF-like cytokines that support the survival and differentiation of B cells at distinct developmental stages. BAFF is a crucial survival factor for transitional and mature B cells that acts as rheostat for the maturation of low-affinity autoreactive cells. In addition, BAFF augments innate B cell responses via complex interactions with the B cell receptor (BCR) and Toll like receptor (TLR) pathways. In this manner, BAFF impacts autoreactive B cell activation via extrafollicular pathways and fine tunes affinity selection within germinal centers (GC). Finally, BAFF and APRIL support plasma cell survival, with differential impacts on IgM- and IgG-producing populations. Therapeutically, BAFF and combined BAFF/APRIL inhibition delays disease onset in diverse murine lupus strains, although responsiveness to BAFF inhibition is model dependent, in keeping with heterogeneity in clinical responses to belimumab treatment in humans. In this review, we discuss the mechanisms whereby BAFF/APRIL signals promote autoreactive B cell activation, discuss whether altered selection accounts for therapeutic benefits of BAFF inhibition, and address whether new insights into BAFF/APRIL family complexity can be exploited to improve human lupus treatments.
Collapse
Affiliation(s)
- Shaun W Jackson
- Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Anne Davidson
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| |
Collapse
|
32
|
Manzi S, Raymond S, Tse K, Peña Y, Anderson A, Arntsen K, Bae SC, Bruce I, Dörner T, Getz K, Hanrahan L, Kao A, Morand E, Rovin B, Schanberg LE, Von Feldt JM, Werth VP, Costenbader K. Global consensus building and prioritisation of fundamental lupus challenges: the ALPHA project. Lupus Sci Med 2019; 6:e000342. [PMID: 31413854 PMCID: PMC6667778 DOI: 10.1136/lupus-2019-000342] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/24/2019] [Accepted: 06/29/2019] [Indexed: 11/14/2022]
Abstract
Objective Lupus is a complex, heterogeneous autoimmune disease that has yet to see significant progress towards more timely diagnosis, improved treatment options for short-term and long-term outcomes, and appropriate access to care. The Addressing Lupus Pillars for Health Advancement (ALPHA) project is the first step in establishing global consensus and developing concrete strategies to address the challenges limiting progress. Methods A Global Advisory Committee of 13 individuals guided the project and began barrier identification. Seventeen expert interviews were conducted to further characterise key barriers. Transcripts were analysed using Nvivo and a codebook was created containing a list of thematic ‘nodes’ (topics) and their descriptions. Findings were used to develop a final survey instrument that was fielded to a diverse, international stakeholder audience to achieve broad consensus. Results Expert interviews identified lupus heterogeneity as the primary barrier hindering advancement. Subsequent barriers were categorised into three areas: (1) Drug development. (2) Clinical care. (3) Access and value. The global survey received 127 completed responses from experts across 20 countries. Respondents identified barriers as high priority including the lack of biomarkers for clinical and drug development use, flawed clinical trial design, lack of access to clinicians familiar with lupus, and obstacles to effective management of lupus due to social determinants of care. Respondents also identified 30 autoimmune conditions that may be lupus-related based on overlapping features, shared autoantibodies and pathophysiology. Conclusions ALPHA is a comprehensive initiative to identify and prioritise the continuum of challenges facing people with lupus by engaging a global audience of lupus experts. It also explored views on lupus as a spectrum of related diseases. Conclusions from this effort provide a framework to generate actionable approaches to the identified high-priority barriers.
Collapse
Affiliation(s)
- Susan Manzi
- Medicine, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Sandra Raymond
- Lupus Foundation of America Inc, Washington, District of Columbia, USA
| | - Karin Tse
- Lupus Foundation of America Inc, Washington, District of Columbia, USA
| | - Yaritza Peña
- Tufts Center for the Study of Drug Development, Boston, Massachusetts, USA
| | - Annick Anderson
- Tufts Center for the Study of Drug Development, Boston, Massachusetts, USA
| | - Kathleen Arntsen
- Lupus and Allied Diseases Association, Inc, Verona, New York, USA
| | - Sang-Cheol Bae
- Rheumatology, Hanyang University Seoul Hospital, Seongdong-gu, Seoul, Korea (the Republic of)
| | - Ian Bruce
- Musculoskeletal and Dermatological Sciences, The University of Manchster, Manchester, England, UK
| | - Thomas Dörner
- Rheumatology and Clinical Immunology, Charite University Hospitals Berlin, Berlin, Germany
| | - Kenneth Getz
- Tufts Center for the Study of Drug Development, Boston, Massachusetts, USA
| | - Leslie Hanrahan
- Lupus Foundation of America Inc, Washington, District of Columbia, USA
| | - Amy Kao
- EMD Serono Research and Development Institute, Billerica, Massachusetts, USA
| | - Eric Morand
- Rheumatology, Monash Medical Centre, Melbourne, Victoria, Australia
| | - Brad Rovin
- Internal Medicine/Nephrology, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | | | | | - Victoria P Werth
- Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
33
|
Smolen JS. Greetings from the editor. Ann Rheum Dis 2019; 78:715. [PMID: 31088798 DOI: 10.1136/annrheumdis-2019-215621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 11/03/2022]
Affiliation(s)
- Josef S Smolen
- Rheumatology, Medical University of Vienna, Vienna 1090, Austria
| |
Collapse
|
34
|
Daikh DI. Antinuclear Antibodies and Lupus: Label Versus Meaning. Arthritis Care Res (Hoboken) 2019; 71:1401-1403. [PMID: 31058455 DOI: 10.1002/acr.23913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 11/11/2022]
Affiliation(s)
- David I Daikh
- San Francisco VA Medical Center and University of California, San Francisco
| |
Collapse
|