1
|
Hu X, Wei Z, Wu Y, Zhao M, Zhou L, Lin Q. Pathogenesis and Therapy of Hermansky-Pudlak Syndrome (HPS)-Associated Pulmonary Fibrosis. Int J Mol Sci 2024; 25:11270. [PMID: 39457053 PMCID: PMC11508683 DOI: 10.3390/ijms252011270] [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: 08/27/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Hermansky-Pudlak syndrome (HPS)-associated pulmonary fibrosis (HPS-PF) is a progressive lung disease that is a major cause of morbidity and mortality in HPS patients. Previous studies have demonstrated that the HPS proteins play an essential role in the biogenesis and function of lysosome-related organelles (LROs) in alveolar epithelial type II (AT2) cells and found that HPS-PF is associated with dysfunction of AT2 cells and abnormal immune reactions. Despite recent advances in research on HPS and the pathology of HPS-PF, the pathological mechanisms underlying HPS-PF remain poorly understood, and no effective treatment has been established. Therefore, it is necessary to refresh the progress in the pathogenesis of HPS-PF to increase our understanding of the pathogenic mechanism of HPS-PF and develop targeted therapeutic strategies. This review summarizes the recent progress in the pathogenesis of HPS-PF provides information about the current treatment strategies for HPS-PF, and hopefully increases our understanding of the pathogenesis of HPS-PF and offers thoughts for new therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | - Qiong Lin
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China; (X.H.); (Z.W.); (Y.W.); (M.Z.); (L.Z.)
| |
Collapse
|
2
|
Goldman N, Ong VH, Denton CP. Pathogenesis of interstitial lung disease in systemic sclerosis. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2024; 5:141-151. [PMID: 39439973 PMCID: PMC11492824 DOI: 10.2478/rir-2024-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 10/25/2024]
Abstract
Interstitial lung disease (ILD) is a frequent important complication of systemic sclerosis (SSc). Factors relevant to aetiopathogenesis of SSc are also central to SSc-ILD. Severity of SSc-ILD is variable but it has a major impact on morbidity and mortality. Factors determining SSc-ILD susceptibility reflect the genetic architecture of SSc and are increasingly being defined. There are aspects linked to immunogenomics and non-immunological genetic factors that may be less conserved and underlie some of the geographical and racial diversity of SSc. These associations may also underlie important links between autoantibody subgroups and patient level risk of SSc-ILD. Examination of blood and tissue samples and observational clinical research together with integrated analysis of in vitro and in vivo preclinical models have elucidated pathogenic mechanisms of SSc-ILD. These have confirmed the potential importance of immune mechanisms in the innate and adaptive immune systemic as well as a significant role for profibrotic pathways especially transforming growth factor beta (TGFbeta) and its regulators and downstream mediators. Recent analysis of clinical trial cohorts as well as integrated and multilevel high dimensional analysis of bio-samples has shed further light on SSc-ILD. This is likely to underpin future advances in stratified and precision medicine for treatment of SSc.
Collapse
Affiliation(s)
- Nina Goldman
- Center for Rheumatology, University College London, London, UK
| | - Voon H Ong
- Center for Rheumatology, University College London, London, UK
| | | |
Collapse
|
3
|
Al-Adwi Y, Westra J, van Goor H, van Kempen LC, Osman M, Gan CT, Timens W, Mulder DJ. Transcriptomic analyses of lung tissues reveal key genes associated with progression of systemic sclerosis-interstitial lung disease (SSc-ILD). J Autoimmun 2024; 148:103297. [PMID: 39098251 DOI: 10.1016/j.jaut.2024.103297] [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: 02/12/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024]
Abstract
OBJECTIVE Systemic sclerosis-interstitial lung disease (SSc-ILD) is the leading cause of death in SSc, affecting around 50 % of the patients. Lung tissue of patients with early-stage SSc-ILD is characterized by a predominant inflammatory response with inconspicuous fibrosis, which may progress to honeycombing fibrosis. Hence, a better understanding of the molecular mechanisms underpinning SSc-ILD pathogenesis is needed to improve treatment options and progression prediction. This transcriptomic study aims to reveal the differential gene expression between control (ctrl) lung tissue and inflammatory, prefibrotic and fibrotic lung tissue to capture progression of early to late phase SSc-ILD. METHODS Twelve explanted lungs from patients with SSc-ILD were used to analyze gene expression from formalin-fixed paraffin-embedded lung tissues with varying stages of ILD (n = 18) and control lung tissue (n = 6). The SSc-ILD tissues were stratified into three ROIs: inflammatory, prefibrotic, and fibrotic using histological assessments to define a longitudinal simulation of early to late phases of SSc-ILD. The nanoString (nS) nCounter Human Fibrosis Panel was used to profile the transcriptome in the regions of interest. Validation of potential targetswas performed with immunohistochemistry in the same tissues that were used for transcriptome analysis. RESULTS To validate our simulation model, we performed subgroup analysis that showed an incremental increase in pathway scores related to the severity of fibrosis. Ctrl vs SSc-ILD comparison demonstrated 24 differentially expressed genes, two of which had the most pronounced p-values. Cyclin-dependent kinase inhibitor (cdkn2c) was overexpressed (P = 0.00052) in SSc-ILD compared to ctrl, while expression of Pellino E3 ubiquitin-protein ligase 1 (peli1) showed lower expression (P = 0.0012). Additionally, in all four groups, cdkn2c and peli1 gene expression showed an incremental increase and decrease, respectively. Immunohistochemistry of cdkn2c showed consistent results with the nS analysis. CONCLUSION More cdkn2c and less peli1 expression were associated with more advanced stages of SSc-ILD on histologic assessment. We report the potential of the cell cycle inhibitor and senescence marker, cdkn2c (p18) to be associated with fibrosis progression.
Collapse
Affiliation(s)
- Yehya Al-Adwi
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Vascular Medicine, Groningen, The Netherlands.
| | - Johanna Westra
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, The Netherlands
| | - Harry van Goor
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Leon C van Kempen
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Mohammed Osman
- Division of Rheumatology, University of Alberta, Edmonton, Canada
| | - C Tji Gan
- University of Groningen, University Medical Center Groningen, Department of Pulmonary Diseases and Tuberculosis, The Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Douwe J Mulder
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Vascular Medicine, Groningen, The Netherlands
| |
Collapse
|
4
|
Pugashetti JV, Khanna D, Kazerooni EA, Oldham J. Clinically Relevant Biomarkers in Connective Tissue Disease-Associated Interstitial Lung Disease. Rheum Dis Clin North Am 2024; 50:439-461. [PMID: 38942579 DOI: 10.1016/j.rdc.2024.03.007] [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] [Indexed: 06/30/2024]
Abstract
Interstitial lung disease (ILD) complicates connective tissue disease (CTD) with variable incidence and is a leading cause of death in these patients. To improve CTD-ILD outcomes, early recognition and management of ILD is critical. Blood-based and radiologic biomarkers that assist in the diagnosis CTD-ILD have long been studied. Recent studies, including -omic investigations, have also begun to identify biomarkers that may help prognosticate such patients. This review provides an overview of clinically relevant biomarkers in patients with CTD-ILD, highlighting recent advances to assist in the diagnosis and prognostication of CTD-ILD.
Collapse
Affiliation(s)
- Janelle Vu Pugashetti
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan.
| | - Dinesh Khanna
- Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan
| | - Ella A Kazerooni
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan; Division of Cardiothoracic Radiology, Department of Radiology, University of Michigan
| | - Justin Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan; Department of Epidemiology, University of Michigan
| |
Collapse
|
5
|
Joerns EK, Karp D, Zhang S, Sparks JA, Adams TN, Makris UE, Newton CA. High Interleukin-13 level is associated with disease stability in interstitial Lung disease. Heliyon 2024; 10:e32118. [PMID: 38882341 PMCID: PMC11176841 DOI: 10.1016/j.heliyon.2024.e32118] [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: 01/12/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Purpose Cytokines can help predict prognosis in interstitial lung disease (ILD) and to differentiate between ILD subtypes. The objectives of our study were to evaluate association of baseline cytokine levels with time to ILD progression and to compare baseline cytokine levels between ILD subtypes. Methods We quantified 27 cytokines using a multiplex assay in peripheral blood samples from 77 patients. Cox proportional hazards regression analysis was performed to evaluate cytokine impact on the time to progression in the total cohort and within each ILD type. We evaluated for significant differences in cytokine levels between ILD types using ANOVA, Wilcoxon signed-rank test and Tukey method. Results Higher IL-13 level was associated with longer time to progression (hazard ratio 0.52 [0.33-0.81], p-value 0.004). FGF-β, GM-CSF, and IL-17 levels differed significantly between fibrotic and inflammatory ILD subgroups. Conclusion IL-13 may be a useful biomarker predicting ILD stability.
Collapse
Affiliation(s)
- Elena K Joerns
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Rheumatic Diseases, Dallas, TX, USA
- Mayo Clinic, Department of Internal Medicine, Division of Rheumatology, Rochester, MN, USA
| | - David Karp
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Rheumatic Diseases, Dallas, TX, USA
| | - Song Zhang
- University of Texas Southwestern Medical Center, Department of Population and Data Sciences, Division of Biostatistics, Dallas, TX, USA
| | - Jeffrey A Sparks
- Brigham and Women's Hospital, Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Harvard Medical School, Boston, MA, USA
| | - Traci N Adams
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Dallas, TX, USA
| | - Una E Makris
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Rheumatic Diseases, Dallas, TX, USA
- Dallas Veterans Affairs Medical Center, Dallas, TX, USA
| | - Chad A Newton
- University of Texas Southwestern Medical Center, Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Dallas, TX, USA
| |
Collapse
|
6
|
Kuwana M, Takehara K, Tanaka Y, Yamashita K, Katsumata K, Takata M, Shima Y. Efficacy and safety of tocilizumab in Japanese patients with systemic sclerosis and associated interstitial lung disease: A subgroup analysis of a global, randomised, controlled Phase 3 trial. Mod Rheumatol 2024; 34:530-540. [PMID: 37436828 DOI: 10.1093/mr/road068] [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: 03/06/2023] [Revised: 06/15/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023]
Abstract
OBJECTIVES The aim of this article is to investigate the efficacy and safety of tocilizumab in Japanese patients with systemic sclerosis. METHODS Post hoc subgroup analysis of a global, randomised, controlled trial in patients treated with weekly tocilizumab 162 mg or placebo subcutaneously in a 48-week double-blind period (tocilizumab and placebo groups) followed by tocilizumab for 48 weeks in an open-label extension (continuous-tocilizumab and placebo-tocilizumab groups). RESULTS Among 20 patients, 12 were randomised to tocilizumab (all had interstitial lung disease) and eight were randomised to placebo (six had interstitial lung disease). The modified Rodnan skin score improved in both treatment groups. The mean change in percent-predicted forced vital capacity was 3.3% [95% confidence interval (CI), -2.5 to 9.0] for tocilizumab and -3.8% (95% CI, -9.9 to 2.2) for placebo in the double-blind period and 2.0% (95% CI, -0.7 to 4.6) for continuous-tocilizumab and -1.4% (95% CI, -6.7 to 4.0) for placebo-tocilizumab in the open-label extension. Rates of serious adverse events per 100 patient-years were 19.3 for tocilizumab and 26.8 for placebo in the double-blind period and 0.0 for continuous-tocilizumab and 13.6 for placebo-tocilizumab in the open-label period. CONCLUSIONS The efficacy and safety of tocilizumab in patients with systemic sclerosis were consistent between the Japanese subpopulation and the global trial population.
Collapse
Affiliation(s)
- Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Kazuhiko Takehara
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | | | | | - Mai Takata
- Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Yoshihito Shima
- Laboratory of Thermo-therapeutics for Vascular Dysfunction/Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
7
|
Stock CJW, Bray WG, Kouranos V, Jacob J, Kokosi M, George PM, Chua F, Wells AU, Sestini P, Renzoni EA. Serum C-reactive protein is associated with earlier mortality across different interstitial lung diseases. Respirology 2024; 29:228-234. [PMID: 37779266 DOI: 10.1111/resp.14609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/20/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND AND OBJECTIVE The acute-phase protein C-reactive protein (CRP) is known to be associated with poor outcomes in cancer and cardiovascular disease, but there is limited evidence of its prognostic implications in interstitial lung diseases (ILDs). We therefore set out to test whether baseline serum CRP levels are associated with mortality in four different ILDs. METHODS In this retrospective study, clinically measured CRP levels, as well as baseline demographics and lung function measures, were collected for ILD patients first presenting to the Royal Brompton Hospital between January 2010 and December 2019. Cox regression analysis was used to determine the relationship with 5-year mortality. RESULTS Patients included in the study were: idiopathic pulmonary fibrosis (IPF) n = 422, fibrotic hypersensitivity pneumonitis (fHP) n = 233, rheumatoid arthritis associated ILD (RA-ILD) n = 111 and Systemic Sclerosis associated ILD (SSc-ILD) n = 86. Patients with a recent history of infection were excluded. Higher CRP levels were associated with shorter 5-year survival in all four disease groups on both univariable analyses, and after adjusting for age, gender, smoking history, immunosuppressive therapy and baseline disease severity (IPF: HR (95% CI): 1.3 (1.1-1.5), p = 0.003, fHP: 1.5 (1.2-1.9), p = 0.001, RA-ILD: 1.4 (1.1-1.84), p = 0.01 and SSc-ILD: 2.7 (1.6-4.5), p < 0.001). CONCLUSION Higher CRP levels are independently associated with reduced 5-year survival in IPF, fHP, RA-ILD and SSc-ILD.
Collapse
Affiliation(s)
- Carmel J W Stock
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - William G Bray
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Vasilis Kouranos
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Joseph Jacob
- Satsuma Lab, Centre for Medical Image Computing, Department of Computer Science, UCL, London, UK
- UCL Respiratory, UCL, London, UK
| | - Maria Kokosi
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter M George
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Felix Chua
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Athol U Wells
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Elisabetta A Renzoni
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
8
|
D’Agnano V, Mariniello DF, Ruotolo M, Quarcio G, Moriello A, Conte S, Sorrentino A, Sanduzzi Zamparelli S, Bianco A, Perrotta F. Targeting Progression in Pulmonary Fibrosis: An Overview of Underlying Mechanisms, Molecular Biomarkers, and Therapeutic Intervention. Life (Basel) 2024; 14:229. [PMID: 38398739 PMCID: PMC10890660 DOI: 10.3390/life14020229] [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: 01/07/2024] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Interstitial lung diseases comprise a heterogenous range of diffuse lung disorders, potentially resulting in pulmonary fibrosis. While idiopathic pulmonary fibrosis has been recognized as the paradigm of a progressive fibrosing interstitial lung disease, other conditions with a progressive fibrosing phenotype characterized by a significant deterioration of the lung function may lead to a burden of significant symptoms, a reduced quality of life, and increased mortality, despite treatment. There is now evidence indicating that some common underlying biological mechanisms can be shared among different chronic fibrosing disorders; therefore, different biomarkers for disease-activity monitoring and prognostic assessment are under evaluation. Thus, understanding the common pathways that induce the progression of pulmonary fibrosis, comprehending the diversity of these diseases, and identifying new molecular markers and potential therapeutic targets remain highly crucial assignments. The purpose of this review is to examine the main pathological mechanisms regulating the progression of fibrosis in interstitial lung diseases and to provide an overview of potential biomarker and therapeutic options for patients with progressive pulmonary fibrosis.
Collapse
Affiliation(s)
- Vito D’Agnano
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (V.D.); (D.F.M.); (M.R.); (G.Q.); (A.M.); (S.C.); (A.S.); (A.B.)
| | - Domenica Francesca Mariniello
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (V.D.); (D.F.M.); (M.R.); (G.Q.); (A.M.); (S.C.); (A.S.); (A.B.)
| | - Michela Ruotolo
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (V.D.); (D.F.M.); (M.R.); (G.Q.); (A.M.); (S.C.); (A.S.); (A.B.)
| | - Gianluca Quarcio
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (V.D.); (D.F.M.); (M.R.); (G.Q.); (A.M.); (S.C.); (A.S.); (A.B.)
| | - Alessandro Moriello
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (V.D.); (D.F.M.); (M.R.); (G.Q.); (A.M.); (S.C.); (A.S.); (A.B.)
| | - Stefano Conte
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (V.D.); (D.F.M.); (M.R.); (G.Q.); (A.M.); (S.C.); (A.S.); (A.B.)
| | - Antonio Sorrentino
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (V.D.); (D.F.M.); (M.R.); (G.Q.); (A.M.); (S.C.); (A.S.); (A.B.)
| | | | - Andrea Bianco
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (V.D.); (D.F.M.); (M.R.); (G.Q.); (A.M.); (S.C.); (A.S.); (A.B.)
| | - Fabio Perrotta
- Department of Translational Medical Sciences, University of Campania L. Vanvitelli, 80131 Naples, Italy; (V.D.); (D.F.M.); (M.R.); (G.Q.); (A.M.); (S.C.); (A.S.); (A.B.)
| |
Collapse
|
9
|
Ghuman A, Khanna D, Lin CJF, Furst DE, Raghu G, Martinez FJ, Zucchetto M, Huang S, Jennings A, Nihtyanova SI, Denton CP. Prognostic and predictive markers of systemic sclerosis-associated interstitial lung disease in a clinical trial and long-term observational cohort. Rheumatology (Oxford) 2024; 63:472-481. [PMID: 37228011 PMCID: PMC10836965 DOI: 10.1093/rheumatology/kead234] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/20/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
OBJECTIVES To explore prognostic and predictive markers of SSc-associated interstitial lung disease (SSc-ILD) outcomes in a phase 3 trial (focuSSced) and prognostic markers in a real-world cohort (SMART). METHODS The focuSSced SSc-ILD subgroup included 68 of 106 placebo-treated and 68 of 104 tocilizumab-treated patients. The SMART cohort included 505 patients with SSc-ILD. Linear mixed-effect models were used to identify factors associated with change in forced vital capacity (FVC). Kaplan-Meier estimation and Cox regression were used for time-to-event analyses. RESULTS In placebo-treated focuSSced patients, sex was a significant prognostic factor for FVC decline; males had increased risk for absolute decline ≥10% in percent-predicted FVC (ppFVC) and 0.22% faster weekly FVC decline than females (P = 0.0001). FVC was 9.8% lower in patients with CRP >6 mg/ml vs those with CRP ≤6 mg/ml (P = 0.0059). Tocilizumab reduced the risk for ≥10% decline in ppFVC in patients who were male, had earlier disease (<2 years duration), had IL-6 levels <10 pg/ml, or had anti-topoisomerase antibodies (ATA). In the SMART cohort, prognostic factors for ppFVC <70% were male sex, ATA, and low baseline FVC. Males had 3.3% lower FVC 1 year after disease onset (P < 0.001) and 0.6% faster yearly decline (P = 0.03) than females. CONCLUSION Prognostic markers in SSc-ILD were similar between focuSSced and SMART. Male sex and inflammatory markers were associated with lower FVC but IL-6 ≥10 pg/ml was not predictive of response to tocilizumab. TRIAL REGISTRATION ClinicalTrials.gov: NCT02453256.
Collapse
Affiliation(s)
| | - Dinesh Khanna
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Daniel E Furst
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Ganesh Raghu
- Division of Pulmonary and Critical Care Medicine, University of Washington Medical Center, Seattle, WA, USA
| | | | | | - Suiyuan Huang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Svetlana I Nihtyanova
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Christopher P Denton
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| |
Collapse
|
10
|
Hamaguchi Y, Yoshimura Y, Horii M, Fushida N, Kitano T, Sawada K, Oishi K, Maeda S, Watanabe S, Matsushita T. Absence of interstitial lung disease at initial visit may predict a favorable outcome for Japanese patients with systemic sclerosis with anti-topoisomerase I antibody. J Dermatol 2024; 51:62-69. [PMID: 37864453 DOI: 10.1111/1346-8138.17001] [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: 03/13/2023] [Revised: 09/01/2023] [Accepted: 09/28/2023] [Indexed: 10/22/2023]
Abstract
Interstitial lung disease (ILD) is recognized a prognostic factor and leading cause of death in patients with systemic sclerosis (SSc). The aim of the present study is to clarify factors at an initial visit that are associated with the deterioration of ILD in SSc patients with anti-topoisomerase I (anti-topo I) antibodies. This was a single-center, retrospective, observational study. Fifty-three consecutive SSc patients with anti-topo I antibodies were included in this study. Of the 53 patients, 43 had ILD at their initial visit, whereas 10 did not. We examined the clinical and immunological factors at an initial visit that were associated with the deterioration of ILD. The deterioration of ILD was defined as the administration of intravenous cyclophosphamide (IVCY) therapy. In this cohort, 45 (85%) patients had ILD at the time of the final observation, and only two who did not have ILD at their initial visit developed ILD during the follow-up period. Until the final observation, 26 (49%) patients received IVCY therapy for the progression of ILD. The age at onset, disease duration, SSc subtype, and skin score were similar between patients with and those without IVCY therapy. Approximately 60% (26 of 43) of patients with ILD at their initial visit received IVCY therapy. On the other hand, none of the 10 patients without ILD at their initial visit received IVCY therapy. Our multivariate analyses using Cox proportional hazards regression model revealed that the presence of ILD at an initial visit was an independent factor associated with the introduction of IVCY therapy (odds ratio, 2.8e+7 [95% confidence interval, 1.8e+17-uncalculated], p = 0.0048). Although anti-topo I antibodies are strongly associated with ILD, it was unlikely for SSc patients with anti-topo I antibodies to receive IVCY therapy when they did not have ILD at an initial visit.
Collapse
Affiliation(s)
- Yasuhito Hamaguchi
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yukari Yoshimura
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Motoki Horii
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Natsumi Fushida
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Tasuku Kitano
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kaori Sawada
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kyosuke Oishi
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Shintaro Maeda
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Takashi Matsushita
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
11
|
Huang H, Wang Q, Xu Z. Advances in the identification and management of progressive pulmonary fibrosis: perspective from Chinese experts. Ther Adv Respir Dis 2024; 18:17534666241288417. [PMID: 39415340 PMCID: PMC11489892 DOI: 10.1177/17534666241288417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 09/16/2024] [Indexed: 10/18/2024] Open
Abstract
Fibrosing interstitial lung diseases (FILDs) other than idiopathic pulmonary fibrosis (IPF) can develop into progressive pulmonary fibrosis (PPF) despite initial management. A substantial proportion of patients with non-IPF interstitial lung diseases (ILDs) progress to PPF, including connective tissue disease-associated ILD (such as rheumatoid arthritis-associated ILD, systemic sclerosis-associated ILD, and idiopathic inflammatory myositis-associated ILD), fibrosing hypersensitivity pneumonitis, and fibrosing occupational ILD. The concept of PPF emerged only recently and several studies have confirmed the impact of PPF on mortality. In addition to poor prognosis among patients with PPF, there remains a lack of consensus in the diagnosis and treatment of PPF across different types of ILDs. There is a need to raise awareness of PPF in FILDs and to explore measures to improve PPF diagnosis and treatment, which in turn could potentially reduce the progression from FILD to PPF. This review discusses the disease burden of PPF and recent advances in the management of PPF among patients with ILDs, including antifibrotic medications that have emerged as promising treatment options. Additionally, this review highlights the perspectives of expert Chinese physicians with regard to their experience in managing PPF in clinical practice.
Collapse
Affiliation(s)
- Hui Huang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zuojun Xu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing 100730, China
| |
Collapse
|
12
|
Characteristics and risk factors of mortality in patients with systemic sclerosis-associated interstitial lung disease. Ann Med 2023; 55:663-671. [PMID: 37074318 PMCID: PMC9970221 DOI: 10.1080/07853890.2023.2179659] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) is a heterogeneous autoimmune disease characterized by dysregulation of fibroblast function, which often involves the lungs. Interstitial lung disease (ILD) associated with SSc (SSc-ILD) is a major cause of death among patients with SSc. Our study aimed to identify risk factors for mortality and compare the clinical characteristics of patients with SSc-ILD. PATIENTS AND METHODS Patients were retrospectively enrolled between 2010 and 2018 in a tertiary hospital in Korea. Patients with SSc-ILD were classified depending on the first pulmonary function test or radiologic findings: extensive (n = 46, >20% disease extent on computed tomography (CT) or forced vital capacity [FVC] < 70% in indeterminate cases) and limited (n = 60, <20% disease extent on CT or FVC ≥70% in indeterminate cases). RESULTS Patients in the extensive group were younger (mean age ± SD 49.3 ± 11.5) than those in the limited group (53.9 ± 12.5, p = .067) at diagnosis. The extensive group showed frequent pulmonary hypertension (43.5% vs. 16.7%, p = .009) and higher erythrocyte sedimentation rate (61.3 ± 33.7 vs. 42.1 ± 26.0, p = .003) and mortality (32.6%, mean duration of follow-up, 100.0 ± 44.7 months vs. 10.0%, 86.0 ± 53.4 months, p = .011). ILD was detected within five years from the first visit (median years 3.5 (1.0, 6.0) vs. 4.5 (0.6, 9.0), survivors vs. non-survivors), and mortality occurred in 19.8% of all patients during a 15-year follow-up. Older age, lower FVC, and initial disease stage (limited or extensive) were associated with mortality, but FVC decline was similar in the limited and extensive groups, such as 15-20% in the first year and 8-10% in the next year, regardless of the initial extent of the disease. CONCLUSIONS Approximately 10% of patients with SSc-ILD in the limited and extensive group showed progression. ILD was detected at a median of less than five years from the first visit; therefore, it is necessary to carefully monitor patients' symptoms and signs from an early stage. Long-term surveillance is also required.Key messagesPatients with systemic sclerosis-interstitial lung disease manifested a heterogeneous disease course.Approximately 10% of the patients in the limited group showed progression, which was similar to the proportion of patients in the extensive group.Interstitial lung disease was detected at a median of less than five years from the first visit.
Collapse
|
13
|
Qin L, Lin H, Zhu F, Wang J, Feng T, Xu T, Zhang G, Zhang X. CD4 +LAG3 +T cells are decreased in SSc-ILD and affect fibroblast mesenchymal transition by TGF-β3. iScience 2023; 26:108225. [PMID: 38025770 PMCID: PMC10661698 DOI: 10.1016/j.isci.2023.108225] [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: 05/08/2023] [Revised: 09/04/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Pulmonary fibrosis frequently occurs in rheumatic conditions, particularly systemic sclerosis-associated interstitial lung disease (SSc-ILD). The pathology involves cell transformation into interstitial structures and collagen accumulation. CD4+LAG3+T cells, known for immune inhibition, are relevant in autoimmunity. This study investigates CD4+LAG3+T cells in SSc-ILD. Clinical analysis revealed a correlation between CD4+LAG3+T cells and interleukin-6 (IL-6) and erythrocyte sedimentation rate (ESR). Using primary human lung fibroblasts (pHLFs) and murine bone marrow-derived macrophages (BMDMs), we showed that CD4+LAG3+T cells secreted TGF-β3 inhibits TGF-β1-induced mesenchymal transformation, modulates cellular function, and reduces collagen release. In mouse models, CD4+LAG3+T cells exhibited potential in alleviating bleomycin-induced pulmonary fibrosis. This study emphasizes CD4+LAG3+T cells' therapeutic promise against fibrosis and proposes their role as biomarkers.
Collapse
Affiliation(s)
- Linmang Qin
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Haobo Lin
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Fu Zhu
- Liuzhou Worker’s Hospital, Liuzhou 545007, China
| | - Jieying Wang
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Tianxiao Feng
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Ting Xu
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Guangfeng Zhang
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xiao Zhang
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| |
Collapse
|
14
|
Al-Adwi Y, Atzeni IM, Doornbos-van der Meer B, van der Leij MJ, Varkevisser RDM, Kroesen BJ, Stel A, Timens W, Gan CT, van Goor H, Westra J, Mulder DJ. High serum C-X-C motif chemokine ligand 10 (CXCL10) levels may be associated with new onset interstitial lung disease in patients with systemic sclerosis: evidence from observational, clinical, transcriptomic and in vitro studies. EBioMedicine 2023; 98:104883. [PMID: 37995465 PMCID: PMC10708993 DOI: 10.1016/j.ebiom.2023.104883] [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: 05/04/2023] [Revised: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Systemic sclerosis-interstitial lung disease (SSc-ILD) is the leading cause of death in patients with SSc. There is an unmet need for predictive biomarkers to identify patients with SSc at risk of ILD. Previous studies have shown that interferon (IFN) pathways may play a role in SSc. We assessed the use of C-X-C motif chemokine ligand 10 (CXCL10) as a predictive biomarker for new onset of ILD in patients with SSc. METHODS One-hundred-sixty-five (Female, N = 130) patients with SSc (SSc-ILD, N = 41) and 13 (Female, N = 8) healthy controls were investigated retrospectively. CXCL10 protein levels were measured by ELISA. We performed log rank analysis with baseline CXCL10 serum levels. CXCL10 nanoString data from lung tissues obtained from transplanted patients with SSc-ILD were extracted. Fifteen (Female, N = 10) patients with SSc (SSc-ILD, N = 7) were recruited for bronchoalveolar lavage (BAL) procedure. Lung fibroblasts were treated with BAL-fluid or serum from patients with SSc with or without ILD. Inflammatory/fibrotic genes were assessed. FINDINGS Serum CXCL10 levels were higher in patients with SSc-ILD compared to SSc patients without ILD [Median (IQR):126 pg/ml (66-282.5) vs. 78.5 pg/ml (50-122), P = 0.029, 95% CI: 1.5 × 10-6 to 0.4284]. Survival analysis showed that baseline CXCL10 levels >78.5 pg/ml have a 2.74-fold increased risk of developing new onset of ILD (Log-rank: P = 0.119) on follow-up. CXCL10 levels in BAL supernatant were not different in patients with SSc-ILD compared to SSc without ILD [76.1 pg/ml (7.2-120.8) vs. 22.3 pg/ml (12.1-43.7), P = 0.24, 95% CI: -19.5 to 100]. NanoString showed that CXCL10 mRNA expression was higher in inflammatory compared to fibrotic lung tissues [4.7 (4.2-5.6) vs. 4.3 (3.6-4.7), P = 0.029]. Fibroblasts treated with SSc-ILD serum or BAL fluids overexpressed CXCL10. INTERPRETATIONS Clinical, transcriptomic, and in vitro data showed that CXCL10 is potentially involved in early SSc-ILD. More research is needed to confirm whether CXCL10 can be classified as a prospective biomarker to detect patients with SSc at higher risk of developing new onset ILD. FUNDING This collaborative project is co-financed by the Ministry of Economic Affairs and Climate Policy of the Netherlands utilizing the PPP-allowance made available by the Top Sector Life Sciences & Health to stimulate public-private partnerships (PPP-2019_007). Part of this study is financially supported by Sanofi Genzyme (NL8921).
Collapse
Affiliation(s)
- Yehya Al-Adwi
- University of Groningen, University Medical Centre Groningen, Department of Internal Medicine, Division of Vascular Medicine, Groningen, the Netherlands.
| | - Isabella Maria Atzeni
- University of Groningen, University Medical Centre Groningen, Department of Internal Medicine, Division of Vascular Medicine, Groningen, the Netherlands
| | - Berber Doornbos-van der Meer
- University of Groningen, University Medical Centre Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Marcel John van der Leij
- University of Groningen, University Medical Centre Groningen, Department of Laboratory Medicine, Groningen, the Netherlands
| | | | - Bart-Jan Kroesen
- University of Groningen, University Medical Centre Groningen, Department of Laboratory Medicine, Groningen, the Netherlands
| | - Alja Stel
- University of Groningen, University Medical Centre Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Wim Timens
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Christiaan Tji Gan
- University of Groningen, University Medical Centre Groningen, Department of Pulmonary Diseases and Tuberculosis, Groningen, the Netherlands
| | - Harry van Goor
- Department of Endocrinology, University Medical Centre Groningen, Groningen, the Netherlands
| | - Johanna Westra
- University of Groningen, University Medical Centre Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Douwe Johannes Mulder
- University of Groningen, University Medical Centre Groningen, Department of Internal Medicine, Division of Vascular Medicine, Groningen, the Netherlands
| |
Collapse
|
15
|
Petelytska L, Bonomi F, Cannistrà C, Fiorentini E, Peretti S, Torracchi S, Bernardini P, Coccia C, De Luca R, Economou A, Levani J, Matucci-Cerinic M, Distler O, Bruni C. Heterogeneity of determining disease severity, clinical course and outcomes in systemic sclerosis-associated interstitial lung disease: a systematic literature review. RMD Open 2023; 9:e003426. [PMID: 37940340 PMCID: PMC10632935 DOI: 10.1136/rmdopen-2023-003426] [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: 06/23/2023] [Accepted: 10/02/2023] [Indexed: 11/10/2023] Open
Abstract
Objective The course of systemic sclerosis-associated interstitial lung disease (SSc-ILD) is highly variable and different from continuously progressive idiopathic pulmonary fibrosis (IPF). Most proposed definitions of progressive pulmonary fibrosis or SSc-ILD severity are based on the research data from patients with IPF and are not validated for patients with SSc-ILD. Our study aimed to gather the current evidence for severity, progression and outcomes of SSc-ILD.Methods A systematic literature review to search for definitions of severity, progression and outcomes recorded for SSc-ILD was performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines in Medline, Embase, Web of Science and Cochrane Library up to 1 August 2023.Results A total of 9054 papers were reviewed and 342 were finally included. The most frequent tools used for the definition of SSc-ILD progression and severity were combined changes of carbon monoxide diffusing capacity (DLCO) and forced vital capacity (FVC), isolated FVC or DLCO changes, high-resolution CT (HRCT) extension and composite algorithms including pulmonary function test, clinical signs and HRCT data. Mortality was the most frequently reported long-term event, both from all causes or ILD related.Conclusions The studies presenting definitions of SSc-ILD 'progression', 'severity' and 'outcome' show a large heterogeneity. These results emphasise the need for developing a standardised, consensus definition of severe SSc-ILD, to link a disease specific definition of progression as a surrogate outcome for clinical trials and clinical practice.PROSPERO registration number CRD42022379254.Cite Now.
Collapse
Affiliation(s)
- Liubov Petelytska
- Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department Internal Medicine #3, Bogomolets National Medical University, Kiiv, Ukraine
| | - Francesco Bonomi
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Carlo Cannistrà
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Elisa Fiorentini
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Silvia Peretti
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Sara Torracchi
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Pamela Bernardini
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Carmela Coccia
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Riccardo De Luca
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Alessio Economou
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Juela Levani
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, San Raffaele Hospital, Milan, Italy
| | - Oliver Distler
- Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cosimo Bruni
- Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence - Careggi University Hospital, Florence, Italy
| |
Collapse
|
16
|
Amati F, Spagnolo P, Ryerson CJ, Oldham JM, Gramegna A, Stainer A, Mantero M, Sverzellati N, Lacedonia D, Richeldi L, Blasi F, Aliberti S. Walking the path of treatable traits in interstitial lung diseases. Respir Res 2023; 24:251. [PMID: 37872563 PMCID: PMC10594881 DOI: 10.1186/s12931-023-02554-8] [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: 08/25/2023] [Accepted: 10/05/2023] [Indexed: 10/25/2023] Open
Abstract
Interstitial lung diseases (ILDs) are complex and heterogeneous diseases. The use of traditional diagnostic classification in ILD can lead to suboptimal management, which is worsened by not considering the molecular pathways, biological complexity, and disease phenotypes. The identification of specific "treatable traits" in ILDs, which are clinically relevant and modifiable disease characteristics, may improve patient's outcomes. Treatable traits in ILDs may be classified into four different domains (pulmonary, aetiological, comorbidities, and lifestyle), which will facilitate identification of related assessment tools, treatment options, and expected benefits. A multidisciplinary care team model is a potential way to implement a "treatable traits" strategy into clinical practice with the aim of improving patients' outcomes. Multidisciplinary models of care, international registries, and the use of artificial intelligence may facilitate the implementation of the "treatable traits" approach into clinical practice. Prospective studies are needed to test potential therapies for a variety of treatable traits to further advance care of patients with ILD.
Collapse
Affiliation(s)
- Francesco Amati
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Christopher J Ryerson
- Department of Medicine, University of British Columbia and Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Justin M Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrea Gramegna
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Anna Stainer
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Marco Mantero
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Nicola Sverzellati
- Unit of Scienze Radiologiche, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Donato Lacedonia
- Department of Medical and Occupational Sciences, Institute of Respiratory Disease, Università degli Studi di Foggia, Foggia, Italy
| | - Luca Richeldi
- Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Blasi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy.
- IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| |
Collapse
|
17
|
Martín-López M, Carreira PE. The Impact of Progressive Pulmonary Fibrosis in Systemic Sclerosis-Associated Interstitial Lung Disease. J Clin Med 2023; 12:6680. [PMID: 37892818 PMCID: PMC10607647 DOI: 10.3390/jcm12206680] [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: 09/04/2023] [Revised: 10/08/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune connective tissue disease characterized by immune dysregulation and progressive fibrosis, typically affecting the skin, with variable internal organ involvement. Interstitial lung disease (ILD), with a prevalence between 35 and 75%, is the leading cause of death in patients with SSc, indicating that all newly diagnosed patients should be screened for this complication. Some patients with SSc-ILD experience a progressive phenotype, which is characterized by worsening fibrosis on high-resolution computed tomography (HRCT), a decline in lung function, and premature mortality. To assess progression and guide therapeutic decisions, regular monitoring is essential and should include pulmonary function testing (PFT), symptom assessment, and repeat HRCT imaging when indicated. Multidisciplinary discussion allows a comprehensive evaluation of the available information and its consequences for management. There has been a shift in the approach to managing SSc-ILD, which includes the addition of targeted biologic and antifibrotic therapies to standard immunosuppressive therapy (particularly mycophenolate mofetil or cyclophosphamide), with autologous hematopoietic stem-cell transplantation and lung transplantation reserved for refractory cases.
Collapse
Affiliation(s)
- María Martín-López
- Department of Rheumatology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
- Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Patricia E. Carreira
- Department of Rheumatology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
- Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| |
Collapse
|
18
|
Amati F, Bongiovanni G, Tonutti A, Motta F, Stainer A, Mangiameli G, Aliberti S, Selmi C, De Santis M. Treatable Traits in Systemic Sclerosis. Clin Rev Allergy Immunol 2023; 65:251-276. [PMID: 37603199 DOI: 10.1007/s12016-023-08969-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2023] [Indexed: 08/22/2023]
Abstract
Systemic sclerosis (SSc) is a chronic systemic disease within the spectrum of connective tissue diseases, specifically characterized by vascular abnormalities and inflammatory and fibrotic involvement of the skin and internal organs resulting in high morbidity and mortality. The clinical phenotype of SSc is heterogeneous, and serum autoantibodies together with the extent of skin involvement have a predictive value in the risk stratification. Current recommendations include an organ-based management according to the predominant involvement with only limited individual factors included in the treatment algorithm. Similar to what has been proposed for other chronic diseases, we hypothesize that a "treatable trait" approach based on relevant phenotypes and endotypes could address the unmet needs in SSc stratification and treatment to maximize the outcomes. We provide herein a comprehensive review and a critical discussion of the literature regarding potential treatable traits in SSc, focusing on established and candidate biomarkers, with the purpose of setting the bases for a precision medicine-based approach. The discussion, structured based on the organ involvement, allows to conjugate the pathogenetic mechanisms of tissue injury with the proposed predictors, particularly autoantibodies and other serum biomarkers. Ultimately, we are convinced that precision medicine is the ideal guide to manage a complex condition such as SSc for which available treatments are largely unsatisfactory.
Collapse
Affiliation(s)
- Francesco Amati
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Gabriele Bongiovanni
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Antonio Tonutti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesca Motta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Anna Stainer
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Giuseppe Mangiameli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Thoracic Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | - Maria De Santis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
19
|
Volkmann ER, Wilhalme H, Assassi S, Kim GHJ, Goldin J, Kuwana M, Tashkin DP, Roth MD. Combining Clinical and Biological Data to Predict Progressive Pulmonary Fibrosis in Patients With Systemic Sclerosis Despite Immunomodulatory Therapy. ACR Open Rheumatol 2023; 5:547-555. [PMID: 37592449 PMCID: PMC10570669 DOI: 10.1002/acr2.11598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/04/2023] [Accepted: 07/13/2023] [Indexed: 08/19/2023] Open
Abstract
OBJECTIVE Progressive pulmonary fibrosis (PPF) is the leading cause of death in systemic sclerosis (SSc). This study aimed to develop a clinical prediction nomogram using clinical and biological data to assess risk of PPF among patients receiving treatment of SSc-related interstitial lung disease (SSc-ILD). METHODS Patients with SSc-ILD who participated in the Scleroderma Lung Study II (SLS II) were randomized to treatment with either mycophenolate mofetil (MMF) or cyclophosphamide (CYC). Clinical and biological parameters were analyzed using univariable and multivariable logistic regression, and a nomogram was created to assess the risk of PPF and validated by bootstrap resampling. RESULTS Among 112 participants with follow-up data, 22 (19.6%) met criteria for PPF between 12 and 24 months. An equal proportion of patients randomized to CYC (n = 11 of 56) and mycophenolate mofetil (n = 11 of 56) developed PPF. The baseline severity of ILD was similar for patients who did, compared to those who did not, experience PPF in terms of their baseline forced vital capacity percent predicted, diffusing capacity for carbon monoxide percent predicted, and quantitative radiological extent of ILD. Predictors in the nomogram included sex, baseline CXCL4 level, and baseline gastrointestinal reflux score. The nomogram demonstrated moderate discrimination in estimating the risk of PPF, with a C-index of 0.72 (95% confidence interval 0.60-0.84). CONCLUSION The SLS II data set provided a unique opportunity to investigate predictors of PPF and develop a nomogram to help clinicians identify patients with SSc-ILD who require closer monitoring while on therapy and potentially an alternative treatment approach. This nomogram warrants external validation in other SSc-ILD cohorts to confirm its predictive power.
Collapse
Affiliation(s)
| | - Holly Wilhalme
- University of California, Los Angeles David Geffen School of Medicine
| | | | - Grace Hyun J. Kim
- University of California, Los Angeles David Geffen School of Medicine
| | - Jonathan Goldin
- University of California, Los Angeles David Geffen School of Medicine
| | | | - Donald P. Tashkin
- University of California, Los Angeles David Geffen School of Medicine
| | - Michael D. Roth
- University of California, Los Angeles David Geffen School of Medicine
| |
Collapse
|
20
|
Piroozmand A, Zamani B, Haddad Kashani H, Amini Mahabadi J. Serum interleukin-6 level and its association with pulmonary involvement in progressive systemic sclerosis; a case-control study. Clin Mol Allergy 2023; 21:7. [PMID: 37670355 PMCID: PMC10478355 DOI: 10.1186/s12948-023-00188-1] [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: 06/26/2022] [Accepted: 07/26/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Primary Systemic Sclerosis (PSS) is a connective tissue disorder characterized by excessive collagen deposition in the skin and internal organs. Interstitial lung disease (ILD) is a late demonstration of PSS and cytokines can contribute to the disease pathology. The purpose of the current study was to determine the association between serum interleukin-6 level and pulmonary involvement in progressive systemic sclerosis. METHODS AND MATERIALS Demographic data and serum interleukin-6 levels were measured for 30 PSS patients with pulmonary involvement (case group) and 30 PSS patients without pulmonary involvement (control group) following informed consent. The disease duration and activity, C-reactive protein (CRP), chest x-ray and highresolution CT scan (HRCT) findings, ejection fraction (EF) and echocardiography findings, and pulmonary artery pressure (PAP) were also determined in both groups. RESULTS The age of patients in case and control groups was 52.5 ± 9.3 and 43.9 ± 9.7 years, respectively (p = 0.001). No significant difference was found between serum levels of IL-6 in case and control groups (73.1 ± 95.4 vs 46.7 ± 83.6 pg/ml, p = 0.267). However, IL-6 level was significantly higher in male case patients compared to male controls (p = 0.007). The duration of PSS was 11.6 ± 6.4 and 7.4 ± 4.2 years in case and control groups, respectively (p = 0.002). The quantitative CRP and PAP was also significantly higher in case patients (p = 0.01 and p < 0.001, respectively). There was found reticulonodular pattern in 20 (66.7%) of the cases, whereas 28 (93.3%) of the controls had normal Chest X-rays (CXR) (p < 0.001). EF was significantly lower in case patients compared to control patients (p = 0.001). CONCLUSION The serum level of IL-6 did not appear to have a relationship with pulmonary involvement, hence it could not be regarded as a potential therapeutic target.
Collapse
Affiliation(s)
- Ahmad Piroozmand
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Batool Zamani
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hamed Haddad Kashani
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
- Gametogenesis Research Center, Institute for Basic Sciences, Kashan University of Medical Science, Kashan, Iran
| | - Javad Amini Mahabadi
- Gametogenesis Research Center, Institute for Basic Sciences, Kashan University of Medical Science, Kashan, Iran
| |
Collapse
|
21
|
Cerro-Chiang G, Ayres M, Rivas A, Romero T, Parker SJ, Mastali M, Elashoff D, Chen P, Van Eyk JE, Wolters PJ, Boin F, Zaman T. Protein biomarkers of disease progression in patients with systemic sclerosis associated interstitial lung disease. Sci Rep 2023; 13:8645. [PMID: 37244972 PMCID: PMC10224939 DOI: 10.1038/s41598-023-35840-y] [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] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Systemic sclerosis is a rare connective tissue disease; and interstitial lung disease (SSc-ILD) is associated with significant morbidity and mortality. There are no clinical, radiologic features, nor biomarkers that identify the specific time when patients are at risk for progression at which the benefits from treatment outweigh the risks. Our study aimed to identify blood protein biomarkers associated with progression of interstitial lung disease in patients with SSc-ILD using an unbiased, high-throughput approach. We classified SSc-ILD as progressive or stable based on change in forced vital capacity over 12 months or less. We profiled serum proteins by quantitative mass spectrometry and analyzed the association between protein levels and progression of SSc-ILD via logistic regression. The proteins associated with at a p value of < 0.1 were queried in the ingenuity pathway analysis (IPA) software to identify interaction networks, signaling, and metabolic pathways. Through principal component analysis, the relationship between the top 10 principal components and progression was evaluated. Unsupervised hierarchical clustering with heatmapping was done to define unique groups. The cohort consisted of 72 patients, 32 with progressive SSc-ILD and 40 with stable disease with similar baseline characteristics. Of a total of 794 proteins, 29 were associated with disease progression. After adjusting for multiple testing, these associations did not remain significant. IPA identified five upstream regulators that targeted proteins associated with progression, as well as a canonical pathway with a higher signal in the progression group. Principal component analysis showed that the ten components with the highest Eigenvalues represented 41% of the variability of the sample. Unsupervised clustering analysis revealed no significant heterogeneity between the subjects. We identified 29 proteins associated with progressive SSc-ILD. While these associations did not remain significant after accounting for multiple testing, some of these proteins are part of pathways relevant to autoimmunity and fibrogenesis. Limitations included a small sample size and a proportion of immunosuppressant use in the cohort, which could have altered the expression of inflammatory and immunologic proteins. Future directions include a targeted evaluation of these proteins in another SSc-ILD cohort or application of this study design to a treatment naïve population.
Collapse
Affiliation(s)
- Giuliana Cerro-Chiang
- Division of Pulmonary and Critical Care Medicine, Cedars Sinai Medical Center, 8700 Beverly Blvd., South Tower Room 6723, Los Angeles, CA, 90048, USA.
| | - Matthew Ayres
- Advanced Clinical Biosystems Institute Biomedical Sciences, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Alejandro Rivas
- Advanced Clinical Biosystems Institute Biomedical Sciences, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Tahmineh Romero
- Division of General Internal Medicine and Health Services Research, University of California Los Angeles, Los Angeles, CA, USA
| | - Sarah J Parker
- Advanced Clinical Biosystems Institute Biomedical Sciences, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Mitra Mastali
- Advanced Clinical Biosystems Institute Biomedical Sciences, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - David Elashoff
- Division of General Internal Medicine and Health Services Research, University of California Los Angeles, Los Angeles, CA, USA
| | - Peter Chen
- Division of Pulmonary and Critical Care Medicine, Cedars Sinai Medical Center, 8700 Beverly Blvd., South Tower Room 6723, Los Angeles, CA, 90048, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Institute Biomedical Sciences, The Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine., University of California, San Francisco, San Francisco, CA, USA
| | - Francesco Boin
- Division of Rheumatology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Tanzira Zaman
- Division of Pulmonary and Critical Care Medicine, Cedars Sinai Medical Center, 8700 Beverly Blvd., South Tower Room 6723, Los Angeles, CA, 90048, USA
| |
Collapse
|
22
|
Pugashetti JV, Khanna D, Kazerooni EA, Oldham J. Clinically Relevant Biomarkers in Connective Tissue Disease-Associated Interstitial Lung Disease. Immunol Allergy Clin North Am 2023; 43:411-433. [PMID: 37055096 PMCID: PMC10584384 DOI: 10.1016/j.iac.2023.01.012] [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: 03/06/2023]
Abstract
Interstitial lung disease (ILD) complicates connective tissue disease (CTD) with variable incidence and is a leading cause of death in these patients. To improve CTD-ILD outcomes, early recognition and management of ILD is critical. Blood-based and radiologic biomarkers that assist in the diagnosis CTD-ILD have long been studied. Recent studies, including -omic investigations, have also begun to identify biomarkers that may help prognosticate such patients. This review provides an overview of clinically relevant biomarkers in patients with CTD-ILD, highlighting recent advances to assist in the diagnosis and prognostication of CTD-ILD.
Collapse
Affiliation(s)
- Janelle Vu Pugashetti
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan.
| | - Dinesh Khanna
- Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan
| | - Ella A Kazerooni
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan; Division of Cardiothoracic Radiology, Department of Radiology, University of Michigan
| | - Justin Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan; Department of Epidemiology, University of Michigan
| |
Collapse
|
23
|
Aversa Z, Atkinson EJ, Carmona EM, White TA, Heeren AA, Jachim SK, Zhang X, Cummings SR, Chiarella SE, Limper AH, LeBrasseur NK. Biomarkers of cellular senescence in idiopathic pulmonary fibrosis. Respir Res 2023; 24:101. [PMID: 37029417 PMCID: PMC10080755 DOI: 10.1186/s12931-023-02403-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/19/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Cellular senescence is a cell fate in response to diverse forms of age-related damage and stress that has been implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF). The associations between circulating levels of candidate senescence biomarkers and disease outcomes have not been specifically studied in IPF. In this study we assessed the circulating levels of candidate senescence biomarkers in individuals affected by IPF and controls and evaluated their ability to predict disease outcomes. METHODS We measured the plasma concentrations of 32 proteins associated with senescence in Lung Tissue Research Consortium participants and studied their relationship with the diagnosis of IPF, parameters of pulmonary and physical function, health-related quality of life, mortality, and lung tissue expression of P16, a prototypical marker of cellular senescence. A machine learning approach was used to evaluate the ability of combinatorial biomarker signatures to predict disease outcomes. RESULTS The circulating levels of several senescence biomarkers were significantly elevated in persons affected by IPF compared to controls. A subset of biomarkers accurately classified participants as having or not having the disease and was significantly correlated with measures of pulmonary function, health-related quality of life and, to an extent, physical function. An exploratory analysis revealed senescence biomarkers were also associated with mortality in IPF participants. Finally, the plasma concentrations of several biomarkers were associated with their expression levels in lung tissue as well as the expression of P16. CONCLUSIONS Our results suggest that circulating levels of candidate senescence biomarkers are informative of disease status, pulmonary and physical function, and health-related quality of life. Additional studies are needed to validate the combinatorial biomarkers signatures that emerged using a machine learning approach.
Collapse
Affiliation(s)
- Zaira Aversa
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | | | - Eva M Carmona
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Thomas A White
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Amanda A Heeren
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Sarah K Jachim
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Steven R Cummings
- Departments of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| | | | - Andrew H Limper
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
24
|
Pope JE, Denton CP, Johnson SR, Fernandez-Codina A, Hudson M, Nevskaya T. State-of-the-art evidence in the treatment of systemic sclerosis. Nat Rev Rheumatol 2023; 19:212-226. [PMID: 36849541 PMCID: PMC9970138 DOI: 10.1038/s41584-023-00909-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2023] [Indexed: 03/01/2023]
Abstract
Systemic sclerosis (SSc) is a rare autoimmune connective tissue disease with multi-organ involvement, fibrosis and vasculopathy. Treatment in SSc, including early diffuse cutaneous SSc (dcSSc) and the use of organ-specific therapies, has improved, as evident from randomized clinical trials. Treatments for early dcSSc include immunosuppressive agents such as mycophenolate mofetil, methotrexate, cyclophosphamide, rituximab and tocilizumab. Patients with rapidly progressive early dcSSc might be eligible for autologous haematopoietic stem cell transplantation, which can improve survival. Morbidity from interstitial lung disease and pulmonary arterial hypertension is improving with the use of proven therapies. Mycophenolate mofetil has surpassed cyclophosphamide as the initial treatment for SSc-interstitial lung disease. Nintedanib and possibly perfinidone can be considered in SSc pulmonary fibrosis. Pulmonary arterial hypertension is frequently treated with initial combination therapy (for example, with phosphodiesterase 5 inhibitors and endothelin receptor antagonists) and, if necessary, the addition of a prostacyclin analogue. Raynaud phenomenon and digital ulcers are treated with dihydropyridine calcium channel blockers (especially nifedipine), then phosphodiesterase 5 inhibitors or intravenous iloprost. Bosentan can reduce the development of new digital ulcers. Trial data for other manifestations are mostly lacking. Research is needed to develop targeted and highly effective treatments, best practices for organ-specific screening and early intervention, and sensitive outcome measurements.
Collapse
Affiliation(s)
- Janet E Pope
- Division of Rheumatology, St Joseph's Health Care, London, ON, Canada.
- Department of Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.
| | | | - Sindhu R Johnson
- Toronto Scleroderma Program, Toronto Western Hospital, Mount Sinai Hospital, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Andreu Fernandez-Codina
- Division of Rheumatology, St Joseph's Health Care, London, ON, Canada
- General Internal Medicine, Windsor Regional Hospital, Windsor, ON, Canada
- Critical Care, Emergency and Systemic Autoimmune Diseases, Hospital Clinic, Barcelona, Spain
| | - Marie Hudson
- Department of Medicine, McGill University, Montreal, QC, Canada
- Division of Rheumatology and Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Tatiana Nevskaya
- Division of Rheumatology, St Joseph's Health Care, London, ON, Canada
| |
Collapse
|
25
|
Abstract
Systemic sclerosis, also known as scleroderma, is a rare and complex autoimmune connective-tissue disease. Once considered an untreatable and unpredictable condition, research advancements have improved our understanding of its disease pathogenesis and clinical phenotypes and expanded our treatment armamentarium. Early and accurate diagnosis is essential, while ongoing efforts to risk stratify patients have a central role in predicting both organ involvement and disease progression. A holistic approach is required when choosing the optimal therapeutic strategy, balancing the side-effect profile with efficacy and tailoring the treatment according to the goals of care of the patient. This Seminar reviews the multiple clinical dimensions of systemic sclerosis, beginning at a precursor very early stage of disease, with a focus on timely early detection of organ involvement. This Seminar also summarises management considerations according to the pathological hallmarks of systemic sclerosis (eg, inflammation, fibrosis, and vasculopathy) and highlights unmet needs and opportunities for future research and discovery.
Collapse
Affiliation(s)
- Elizabeth R Volkmann
- Department of Medicine, Division of Rheumatology, University of California, Los Angeles, CA, USA; David Geffen School of Medicine, Los Angeles, CA, USA.
| | | | - Vanessa Smith
- Department of Internal Medicine and Department of Rheumatology, Ghent University (Hospital), Ghent, Belgium; Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Centre, Ghent, Belgium
| |
Collapse
|
26
|
Cerro Chiang G, Parimon T. Understanding Interstitial Lung Diseases Associated with Connective Tissue Disease (CTD-ILD): Genetics, Cellular Pathophysiology, and Biologic Drivers. Int J Mol Sci 2023; 24:ijms24032405. [PMID: 36768729 PMCID: PMC9917355 DOI: 10.3390/ijms24032405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
Connective tissue disease-associated interstitial lung disease (CTD-ILD) is a collection of systemic autoimmune disorders resulting in lung interstitial abnormalities or lung fibrosis. CTD-ILD pathogenesis is not well characterized because of disease heterogeneity and lack of pre-clinical models. Some common risk factors are inter-related with idiopathic pulmonary fibrosis, an extensively studied fibrotic lung disease, which includes genetic abnormalities and environmental risk factors. The primary pathogenic mechanism is that these risk factors promote alveolar type II cell dysfunction triggering many downstream profibrotic pathways, including inflammatory cascades, leading to lung fibroblast proliferation and activation, causing abnormal lung remodeling and repairs that result in interstitial pathology and lung fibrosis. In CTD-ILD, dysregulation of regulator pathways in inflammation is a primary culprit. However, confirmatory studies are required. Understanding these pathogenetic mechanisms is necessary for developing and tailoring more targeted therapy and provides newly discovered disease biomarkers for early diagnosis, clinical monitoring, and disease prognostication. This review highlights the central CTD-ILD pathogenesis and biological drivers that facilitate the discovery of disease biomarkers.
Collapse
Affiliation(s)
- Giuliana Cerro Chiang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Correspondence:
| | - Tanyalak Parimon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
27
|
Beesley CF, Goldman NR, Taher TE, Denton CP, Abraham DJ, Mageed RA, Ong VH. Dysregulated B cell function and disease pathogenesis in systemic sclerosis. Front Immunol 2023; 13:999008. [PMID: 36726987 PMCID: PMC9885156 DOI: 10.3389/fimmu.2022.999008] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/22/2022] [Indexed: 01/18/2023] Open
Abstract
Systemic sclerosis (SSc) is a complex, immune-mediated rheumatic disease characterised by excessive extracellular matrix deposition in the skin and internal organs. B cell infiltration into lesional sites such as the alveolar interstitium and small blood vessels, alongside the production of defined clinically relevant autoantibodies indicates that B cells play a fundamental role in the pathogenesis and development of SSc. This is supported by B cell and fibroblast coculture experiments revealing that B cells directly enhance collagen and extracellular matrix synthesis in fibroblasts. In addition, B cells from SSc patients produce large amounts of profibrotic cytokines such as IL-6 and TGF-β, which interact with other immune and endothelial cells, promoting the profibrotic loop. Furthermore, total B cell counts are increased in SSc patients compared with healthy donors and specific differences can be found in the content of naïve, memory, transitional and regulatory B cell compartments. B cells from SSc patients also show differential expression of activation markers such as CD19 which may shape interactions with other immune mediators such as T follicular helper cells and dendritic cells. The key role of B cells in SSc is further supported by the therapeutic benefit of B cell depletion with rituximab in some patients. It is notable also that B cell signaling is impaired in SSc patients, and this could underpin the failure to induce tolerance in B cells as has been shown in murine models of scleroderma.
Collapse
Affiliation(s)
- Claire F. Beesley
- Centre for Rheumatology, Division of Medicine, University College London, London, United Kingdom
| | - Nina R. Goldman
- Centre for Rheumatology, Division of Medicine, University College London, London, United Kingdom
| | - Taher E. Taher
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Christopher P. Denton
- Centre for Rheumatology, Division of Medicine, University College London, London, United Kingdom
| | - David J. Abraham
- Centre for Rheumatology, Division of Medicine, University College London, London, United Kingdom
| | - Rizgar A. Mageed
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Voon H. Ong
- Centre for Rheumatology, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
28
|
Kamenova A, Tzouvelekis A, Margaritopoulos GA. Recent advances in the treatment of systemic sclerosis associated interstitial lung disease. Front Med (Lausanne) 2023; 10:1155771. [PMID: 37035331 PMCID: PMC10079888 DOI: 10.3389/fmed.2023.1155771] [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: 01/31/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Connective tissue diseases (CTDs) are a heterogenous group of systemic inflammatory disorders. The development of connective tissue disease-associated interstitial lung disease (CTD-ILD) is a key complication associated with significant morbidity and mortality. The aim of this review is to explore the pathogenesis of CTD-ILD and summarize the recent evidence from clinical trials for novel treatment options, including the role of antifibrotics and immunomodulatory therapies with a focus on systemic sclerosis associated ILD. Further clinical trials are ongoing to explore combination therapies and more targeted therapeutic options. Clinicians remain faced with the difficult challenge of appropriately selecting patients who will benefit from the available therapies and timing the start of therapy at the most suitable part of the disease course.
Collapse
Affiliation(s)
- Antoniya Kamenova
- Interstitial Lung Disease Unit, London North West University Hospital HT, London, United Kingdom
| | - Argyris Tzouvelekis
- Respiratory Medicine Department, University of Patras, Patras, Greece
- *Correspondence: Argyris Tzouvelekis,
| | | |
Collapse
|
29
|
Muntyanu A, Le M, Ridha Z, O’Brien E, Litvinov IV, Lefrançois P, Netchiporouk E. Novel role of long non-coding RNAs in autoimmune cutaneous disease. J Cell Commun Signal 2022; 16:487-504. [PMID: 34346026 PMCID: PMC9733767 DOI: 10.1007/s12079-021-00639-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/22/2021] [Indexed: 12/13/2022] Open
Abstract
Systemic autoimmune rheumatic diseases (SARDs) are a heterogeneous group of chronic multisystem inflammatory disorders that are thought to have a complex pathophysiology, which is not yet fully understood. Recently, the role of non-coding RNAs, including long non-coding RNA (lncRNA), has been of particular interest in the pathogenesis of SARDs. We aimed to summarize the potential roles of lncRNA in SARDs affecting the skin including, systemic sclerosis (SSc), dermatomyositis (DM) and cutaneous lupus erythematosus (CLE). We conducted a narrative review summarizing original articles published until July 19, 2021, regarding lncRNA associated with SSc, DM, and CLE. Several lncRNAs were hypothesized to play an important role in disease pathogenesis of SSc, DM and CLE. In SSc, Negative Regulator of IFN Response (NRIR) was thought to modulate Interferon (IFN) response in monocytes, anti-sense gene to X-inactivation specific transcript (TSIX) to regulate increased collagen stability, HOX transcript antisense RNA (HOTAIR) to increase numbers of myofibroblasts, OTUD6B-Anti-Sense RNA 1 to decrease fibroblast apoptosis, ncRNA00201 to regulate pathways in SSc pathogenesis and carcinogenesis, H19X potentiating TGF-β-driven extracellular matrix production, and finally PSMB8-AS1 potentiates IFN response. In DM, linc-DGCR6-1 expression was hypothesized to target the USP18 protein, a type 1 IFN-inducible protein that is considered a key regulator of IFN signaling. Additionally, AL136018.1 is suggested to regulate the expression Cathepsin G, which increases the permeability of vascular endothelial cells and the chemotaxis of inflammatory cells in peripheral blood and muscle tissue in DM. Lastly, lnc-MIPOL1-6 and lnc-DDX47-3 in discoid CLE were thought to be associated with the expression of chemokines, which are significant in Th1 mediated disease. In this review, we summarize the key lncRNAs that may drive pathogenesis of these connective tissue diseases and could potentially serve as therapeutic targets in the future.
Collapse
Affiliation(s)
- Anastasiya Muntyanu
- Division Dermatology, McGill University Health Centre, 1650 Cedar Ave, Montreal, QC H3G 1A4 Canada
| | - Michelle Le
- Division Dermatology, McGill University Health Centre, 1650 Cedar Ave, Montreal, QC H3G 1A4 Canada
| | - Zainab Ridha
- Faculty of Medicine, Université de Laval, Québec, QC Canada
| | - Elizabeth O’Brien
- Division Dermatology, McGill University Health Centre, 1650 Cedar Ave, Montreal, QC H3G 1A4 Canada
| | - Ivan V. Litvinov
- Division Dermatology, McGill University Health Centre, 1650 Cedar Ave, Montreal, QC H3G 1A4 Canada
| | - Philippe Lefrançois
- Division Dermatology, McGill University Health Centre, 1650 Cedar Ave, Montreal, QC H3G 1A4 Canada
| | - Elena Netchiporouk
- Division Dermatology, McGill University Health Centre, 1650 Cedar Ave, Montreal, QC H3G 1A4 Canada
| |
Collapse
|
30
|
Zhu W, Wang Y, Liu C, Wu Y, Li Y, Wang Y. Connective tissue disease-related interstitial lung disease is alleviated by tripterine through inhibition of the PI3K/Akt, apoptosis, and TNF-α signalling pathways. Front Pharmacol 2022; 13:990760. [DOI: 10.3389/fphar.2022.990760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022] Open
Abstract
Background: Interstitial lung disease (ILD) is the major cause of morbidity and mortality in patients with various rheumatic diseases. However, more interventions need to be sought. Tripterine, an extract of Tripterygium wilfordii Hook. F, has been widely studied for its powerful anti-inflammatory effect. However, its mechanism of action in treating connective tissue disease-related (CTD)-ILD remains unclear.Purpose: To investigate the mechanism of tripterine in CTD-ILD treatment by combining network pharmacology and an in vivo experiment.Methods: The related targets of tripterine were obtained after searching the Traditional Chinese Medicine System Pharmacology Database and Analysis Platform, Comparative Toxicogenomics Database, GeneCards, Search Tool for Interacting Chemicals database, and SymMap database. Following this, Online Mendelian Inheritance in Man, GeneCards, Genebank, and DrugBank were used to screen the targets of CTD-ILD. A target-signalling pathway network was constructed using Cytoscape. Additionally, topological analysis was performed. Protein interaction analysis was performed using the STRING online analysis platform. Following this, Gene Ontology (GO) and the Kyoto Encyclopaedia of Genes and Genomes (KEGG) signalling pathway enrichment analyses were performed. Subsequently, the molecular docking between tripterine and the core targets was verified. Finally, experimental verification was performed in bleomycin-induced model mice.Results: A total of 134 common targets and 10 core targets of tripterine, including signal transducer and activator of transcription 3, tumour necrosis factor (TNF), v-rel avian reticuloendotheliosis viral oncogene homolog A, protein kinase B (Akt) α (Akt1), mitogen-activated protein kinase (MAPK) 1, Jun transcription factor family, tumour protein 53, MAPK3, nuclear factor kappa B subunit 1, and caspase 8, were obtained. GO enrichment analysis revealed that, while treating CTD-ILD, tripterine was mainly involved in cytokine receptor binding, receptor-ligand activity, signal receptor activation, cytokine activity, protein ubiquitination, deoxyribonucleic acid transcriptase activity, etc. The KEGG pathway enrichment analysis revealed that the most significant signalling pathways were multiple viral infections and the phosphatidylinositol-3-kinase (PI3K)/Akt, TNF, and apoptosis signalling pathways. Molecular docking results revealed that tripterine had good docking activity with the core targets. Experimental studies also demonstrated that tripterine could inhibit the activation of PI3K/Akt, apoptosis, and TNF-α signalling pathways in lung tissue and significantly improve lung pathology and collagen deposition in the model mice.Conclusions: This study preliminarily revealed the potential molecular biological mechanism of tripterine while treating CTD-ILD might be related to inhibiting the PI3K/Akt, apoptosis, and TNF-α signalling pathways. Tripterygium wilfordii Hook. F. and its extract could be used clinically for treating CTD-ILD.
Collapse
|
31
|
Kuster S, Jordan S, Elhai M, Held U, Steigmiller K, Bruni C, Cacciapaglia F, Vettori S, Siegert E, Rednic S, Codullo V, Airo P, Braun-Moscovici Y, Hunzelmann N, Joao Salvador M, Riccieri V, Gheorghiu AM, Alegre Sancho JJ, Romanowska-Prochnicka K, Castellví I, Kötter I, Truchetet ME, López-Longo FJ, Novikov PI, Giollo A, Shirai Y, Belloli L, Zanatta E, Hachulla E, Smith V, Denton C, Ionescu RM, Schmeiser T, Distler JHW, Gabrielli A, Hoffmann-Vold AM, Kuwana M, Allanore Y, Distler O. Effectiveness and safety of tocilizumab in patients with systemic sclerosis: a propensity score matched controlled observational study of the EUSTAR cohort. RMD Open 2022; 8:e002477. [PMID: 36328401 PMCID: PMC9639158 DOI: 10.1136/rmdopen-2022-002477] [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: 05/18/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES Tocilizumab showed trends for improving skin fibrosis and prevented progression of lung fibrosis in systemic sclerosis (SSc) in randomised controlled clinical trials. We aimed to assess safety and effectiveness of tocilizumab in a real-life setting using the European Scleroderma Trial and Research (EUSTAR) database. METHODS Patients with SSc fulfilling the American College of Rheumatology (ACR)/EULAR 2013 classification criteria, with baseline and follow-up visits at 12±3 months, receiving tocilizumab or standard of care as the control group, were selected. Propensity score matching was applied. Primary endpoints were the modified Rodnan skin score (mRSS) and FVC at 12±3 months compared between the groups. Secondary endpoints were the percentage of progressive/regressive patients for skin and lung at 12±3 months. RESULTS Ninety-three patients with SSc treated with tocilizumab and 3180 patients with SSc with standard of care fulfilled the inclusion criteria. Comparison between groups did not show significant differences, but favoured tocilizumab across all predefined primary and secondary endpoints: mRSS was lower in the tocilizumab group (difference -1.0, 95% CI -3.7 to 1.8, p=0.48). Similarly, FVC % predicted was higher in the tocilizumab group (difference 1.5 (-6.1 to 9.1), p=0.70). The percentage of progressive/regressive patients favoured tocilizumab over controls. These results were robust regarding the sensitivity analyses. Safety analysis confirmed previously reported adverse event profiles. CONCLUSION Although this large, observational, controlled, real-life EUSTAR study did not show significant effectiveness of tocilizumab on skin and lung fibrosis, the consistency of direction of all predefined endpoints generates hypothesis for potential effectiveness in a broader SSc population.
Collapse
Affiliation(s)
- Simon Kuster
- Department of Rheumatology; University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Suzana Jordan
- Department of Rheumatology; University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Muriel Elhai
- Department of Rheumatology; University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Rheumatology, Paris Descartes University, Cochin Hospital, APHP, Paris, France
| | - Ulrike Held
- Department of Biostatistics at Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Klaus Steigmiller
- Department of Biostatistics at Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Cosimo Bruni
- Department of Rheumatology; University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence, Italy
| | - Fabio Cacciapaglia
- Rheumatology Unit, DETO, University Hospital Polyclinic of Bari, Bari, Italy
| | - Serena Vettori
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Elise Siegert
- Clinic for Rheumatology and Immunology, Bad Bramstedt, Germany
- Berlin Institute of Health at Charité, Berlin, Germany
| | - Simona Rednic
- Clinica Reumatologie, University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Veronica Codullo
- Department of Rheumatology, IRCCS Foundation Policlinico San Matteo, Pavia, Italy
| | - Paolo Airo
- UOC Rheumatology and Clinical Immunology, Spedali Civili di Brescia, Brescia, Italy
| | | | | | - Maria Joao Salvador
- Department of Rheumatology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Valeria Riccieri
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Ana-Maria Gheorghiu
- Department of Internal Medicine and Rheumatology, Cantacuzino Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Katarzyna Romanowska-Prochnicka
- Department of Biophysics, Physiology and Pathophysiology, Medical University of Warsaw, Warsaw, Poland
- Department of Connective Tissues Diseases, Institute of Rheumatology, Warsaw, Poland
| | - Ivan Castellví
- Department of Rheumatology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ina Kötter
- Clinic for Rheumatology and Immunology, Bad Bramstedt, Germany
- University Hospital Hamburg-Eppendorf, Division of Rheumatology and Systemic Inflammatory Diseases, Hamburg, Germany
| | - Marie-Elise Truchetet
- Department of Rheumatology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - F J López-Longo
- Department of Rheumatology, National Reference Center for Systemic Autoimmune Rare Diseases, Hopital Pellegrin, Bordeaux, France
| | - Pavel I Novikov
- Department of Rheumatology, National Reference Center for Systemic Autoimmune Rare Diseases, Hopital Pellegrin, Bordeaux, France
| | - Alessandro Giollo
- Rheumatology Section, Department of Medicine, University of Verona, Verona, Italy
| | - Yuichiro Shirai
- Department of Allergy and Rheumatology, Nippon Medical School, Tokyo, Japan
| | - Laura Belloli
- Rheumatology Unit, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Elisabetta Zanatta
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Padova, Padova, Italy
| | - Eric Hachulla
- Referral Centre for Rare Systemic Auto-immune Diseases for North and North-West of France, Department of Internal Medicine, Claude Huriez Hospital, University of Lille, Lille, France
| | - Vanessa Smith
- Department of Internal Medicine, Department of Rheumatology, Ghent University Hospital, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Centre (IRC), Ghent, Belgium
| | - Chris Denton
- Centre for Rheumatology, University College London Medical School-Royal Free Campus, London, UK
| | - Ruxandra M Ionescu
- Internal Medicine and Rheumatology Department-St. Maria Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Joerg H W Distler
- Department of Internal Medicine III, Erlangen University Hospital, Erlangen, Germany
| | - Armando Gabrielli
- Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | | | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School, Tokyo, Japan
| | - Yannick Allanore
- Department of Rheumatology, Paris Descartes University, Cochin Hospital, APHP, Paris, France
| | - Oliver Distler
- Department of Rheumatology; University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Kurche JS, Stancil IT, Michalski JE, Yang IV, Schwartz DA. Dysregulated Cell-Cell Communication Characterizes Pulmonary Fibrosis. Cells 2022; 11:3319. [PMID: 36291184 PMCID: PMC9600037 DOI: 10.3390/cells11203319] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/27/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease of older adults characterized by fibrotic replacement of functional gas exchange units in the lung. The strongest risk factor for IPF is a genetic variantin the promoter region of the gel-forming mucin, MUC5B. To better understand how the MUC5B variant influences development of fibrosis, we used the NicheNet R package and leveraged publicly available single-cell RNA sequencing data to identify and evaluate how epithelia participating in gas exchange are influenced by ligands expressed in control, MUC5B variant, and fibrotic environments. We observed that loss of type-I alveolar epithelia (AECI) characterizes the single-cell RNA transcriptome in fibrotic lung and validated the pattern of AECI loss using single nuclear RNA sequencing. Examining AECI transcriptomes, we found enrichment of transcriptional signatures for IL6 and AREG, which we have previously shown to mediate aberrant epithelial fluidization in IPF and murine bleomycin models. Moreover, we found that the protease ADAM17, which is upstream of IL6 trans-signaling, was enriched in control MUC5B variant donors. We used immunofluorescence to validate a role for enhanced expression of ADAM17 among MUC5B variants, suggesting involvement in IPF pathogenesis and maintenance.
Collapse
Affiliation(s)
- Jonathan S. Kurche
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| | - Ian T. Stancil
- Program in Cellular Biology and Biophysics, Graduate School, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jacob E. Michalski
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ivana V. Yang
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David A. Schwartz
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| |
Collapse
|
33
|
Jasmer KJ, Muñoz Forti K, Woods LT, Cha S, Weisman GA. Therapeutic potential for P2Y 2 receptor antagonism. Purinergic Signal 2022:10.1007/s11302-022-09900-3. [PMID: 36219327 DOI: 10.1007/s11302-022-09900-3] [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: 08/26/2022] [Accepted: 09/22/2022] [Indexed: 10/17/2022] Open
Abstract
G protein-coupled receptors are the target of more than 30% of all FDA-approved drug therapies. Though the purinergic P2 receptors have been an attractive target for therapeutic intervention with successes such as the P2Y12 receptor antagonist, clopidogrel, P2Y2 receptor (P2Y2R) antagonism remains relatively unexplored as a therapeutic strategy. Due to a lack of selective antagonists to modify P2Y2R activity, studies using primarily genetic manipulation have revealed roles for P2Y2R in a multitude of diseases. These include inflammatory and autoimmune diseases, fibrotic diseases, renal diseases, cancer, and pathogenic infections. With the advent of AR-C118925, a selective and potent P2Y2R antagonist that became commercially available only a few years ago, new opportunities exist to gain a more robust understanding of P2Y2R function and assess therapeutic effects of P2Y2R antagonism. This review discusses the characteristics of P2Y2R that make it unique among P2 receptors, namely its involvement in five distinct signaling pathways including canonical Gαq protein signaling. We also discuss the effects of other P2Y2R antagonists and the pivotal development of AR-C118925. The remainder of this review concerns the mounting evidence implicating P2Y2Rs in disease pathogenesis, focusing on those studies that have evaluated AR-C118925 in pre-clinical disease models.
Collapse
Affiliation(s)
- Kimberly J Jasmer
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Kevin Muñoz Forti
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Lucas T Woods
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Seunghee Cha
- Department of Oral and Maxillofacial Diagnostic Sciences, Center for Orphaned Autoimmune Disorders, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Gary A Weisman
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA. .,Department of Biochemistry, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
34
|
Cole A, Denton CP. Biomarkers in Systemic Sclerosis Associated Interstitial Lung Disease (SSc-ILD). CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2022. [DOI: 10.1007/s40674-022-00196-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Abstract
Purpose of Review
Interstitial lung disease (ILD) is the leading cause of mortality in systemic sclerosis, a rare autoimmune disease characterised by fibrosis and vasculopathy. The variety of phenotypes in SSc-ILD have inspired multiple studies aimed at the identification of biomarkers which can provide disease-specific information but due to the complex pathogenesis of SSc-ILD, it has been challenging to validate such markers. We provide a comprehensive update on those most studied along with emerging biomarkers.
Recent Findings
We review the up-to-date findings with regard to the use of well-studied molecular biomarkers in SSc-ILD along with novel biomarkers offering promise as prognostic markers such as IGFBP-2 and IGFBP-7, the adipokine CTRP9, endothelial progenitor cells, and cellular markers such as CD21lo/neg B cells. Expression profiling data is being used in SSc patients to determine genetic and epigenetic clusters which shed further light on mechanisms involved in the pathogenesis of SSc-ILD and are likely to uncover novel biomarkers.
Summary
With the exception of autoantibodies, there are no routinely measured biomarkers in SSc-ILD and reliable validation of the many potential biomarkers is lacking. Identifying biomarkers which can offer diagnostic and prognostic certainty may help patients to receive preventative treatment as part of a personalised medicine approach.
Collapse
|
35
|
Bellocchi C, Chung A, Volkmann ER. Predicting the Progression of Very Early Systemic Sclerosis: Current Insights. Open Access Rheumatol 2022; 14:171-186. [PMID: 36133926 PMCID: PMC9484572 DOI: 10.2147/oarrr.s285409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/06/2022] [Indexed: 11/28/2022] Open
Abstract
Systemic sclerosis (SSc) is a complex autoimmune connective tissue disease with distinct pathological hallmarks (ie, inflammation, vasculopathy, fibrosis) that may predominate at different stages in the disease course with varying severity. Initial efforts to classify patients with SSc identified a subset of patients with very early SSc. These patients possessed signs of SSc (eg, Raynaud phenomenon, SSc specific autoantibodies and/or nailfold capillary abnormalities) without fulfilling complete SSc classification criteria. Recognizing the inherent value in early diagnosis and intervention in SSc, researchers have endeavored to identify risk factors for progression from very early SSc to definite SSc. The present review summarizes the clinical phenotype of patients with very early and early SSc. Through a scoping review of recent literature, this review also describes risk factors for progression to definite SSc with a focus on the specific clinical features that arise early in the SSc disease course (eg, diffuse cutaneous sclerosis, interstitial lung disease, esophageal dysfunction, renal crisis, cardiac involvement). In addition to clinical risk factors, this review provides evidence for how biological data (ie, serological, genomic, proteomic profiles, skin bioengineering methods) can be integrated into risk assessment models in the future. Furthering our understanding of biological features of very early SSc will undoubtedly provide novel insights into SSc pathogenesis and may illuminate new therapeutic targets to prevent progression of SSc.
Collapse
Affiliation(s)
- Chiara Bellocchi
- Scleroderma Unit, Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, University of Milan, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Augustine Chung
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Elizabeth R Volkmann
- Division of Rheumatology, Department of Medicine, University of California, David Geffen School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
36
|
Chikhoune L, Brousseau T, Morell-Dubois S, Farhat MM, Maillard H, Ledoult E, Lambert M, Yelnik C, Sanges S, Sobanski V, Hachulla E, Launay D. Association between Routine Laboratory Parameters and the Severity and Progression of Systemic Sclerosis. J Clin Med 2022; 11:jcm11175087. [PMID: 36079017 PMCID: PMC9457158 DOI: 10.3390/jcm11175087] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Systemic sclerosis (SSc) is a heterogeneous connective tissue disease with a high mortality and morbidity rate. Identification of biomarkers that can predict the evolution of SSc is a key factor in the management of patients. The aim of this study was to assess the association of routine laboratory parameters, widely used in practice and easily available, with the severity and progression of SSc. (2) Methods: In this retrospective monocentric cohort study, 372 SSc patients were included. We gathered clinical and laboratory data including routine laboratory parameters: C-reactive-protein (CRP), erythrocyte sedimentation rate (ESR), complete blood count, serum sodium and potassium levels, creatinin, urea, ferritin, albumin, uric acid, N-terminal pro-brain natriuretic peptide (NTproBNP), serum protein electrophoresis, and liver enzymes. Associations between these routine laboratory parameters and clinical presentation and outcome were assessed. (3) Results: Median (interquartile range) age was 59.0 (50.0; 68.0) years. White blood cell, monocyte, and neutrophil absolute counts were significantly higher in patients with diffuse cutaneous SSc and with interstitial lung disease (ILD) (p < 0.001). CRP was significantly higher in patients with ILD (p < 0.001). Hemoglobin and ferritin were significantly lower in patients with pulmonary hypertension (PH) including pulmonary arterial hypertension and ILD associated PH (p = 0.016 and 0.046, respectively). Uric acid and NT pro BNP were significantly higher in patients with PH (<0.001). Monocyte count was associated with ILD progression over time. (4) Conclusions: Overall, our study highlights the association of routine laboratory parameters used in current practice with the severity and progression of SSc.
Collapse
Affiliation(s)
- Liticia Chikhoune
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Thierry Brousseau
- CHU Lille, Service de Biochimie Automatisée Protéines, F-59000 Lille, France
| | - Sandrine Morell-Dubois
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Meryem Maud Farhat
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - Helene Maillard
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Emmanuel Ledoult
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - Marc Lambert
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Cecile Yelnik
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Sebastien Sanges
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - Vincent Sobanski
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - Eric Hachulla
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - David Launay
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
- Correspondence: ; Tel.: +33-3-2044-4433
| |
Collapse
|
37
|
Shi S, Chen L, Liu X, Yu M, Wu C, Xiao Y. Development of a scoring system with multidimensional markers for fibrosing interstitial lung disease. Sci Rep 2022; 12:14217. [PMID: 35987772 PMCID: PMC9392719 DOI: 10.1038/s41598-022-16382-1] [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: 03/03/2022] [Accepted: 07/08/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractFibrosing interstitial lung disease (ILD) can cause high mortality and sensitive evaluation of fibrosing ILD could be critical. The aim of this study is to develop a scoring system to predict prognosis of fibrosing ILD. 339 patients with fibrosing ILD were enrolled as a derivation cohort. Cox multiple regression analysis indicated that smoking history (HR = 3.826, p = 0.001), age(HR = 1.043, p = 0.015), CEA(HR = 1.059, p = 0.049),CYFRA21-1(HR = 1.177, p = 0.004) and DLCO% predicted (HR = 0.979, p = 0.032) were independent prognostic factors for fibrosing ILD. The clinical scoring system for fibrosing ILD was established based on the clinical variables (age [A], CEA and CYFRA21-1 [C], DLCO% predicted [D], and smoking history [S]; ACDS). The area under the receiver operating characteristic curve (AUROC) of the scoring system for predicting prognosis of fibrosing ILD was 0.90 (95%CI: 0.87–0.94, p < 0.001). The cutoff value was 2.5 with their corresponding specificity (90.7%) and sensitivity (78.8%). To validate the value of ACDS score levels to predict the survival of patients with fibrosing ILD, 98 additional fibrosing ILD patients were included as a validation cohort. The log-rank test showed a significant difference in survival between the two groups(ACDS score < 2.5 and ACDS score ≥ 2.5) in validation cohort. The independent risk factors for mortality in patients with fibrosing ILD are higher CEA, higher CYFRA21-1, smoking history, lower DLCO%predicted at baseline and older age. ACDS is a simple and feasible clinical model for predicting survival of fibrosing ILD.
Collapse
|
38
|
Dutt N, Shishir S, Chauhan NK, Jalandra R, kuwal A, Garg P, Kumar D, Vishwajeet V, Chakraborti A, Deokar K, Asfahan S, Babu A, bajad P, Gupta N, Khurana A, Garg MK. Mortality and Its Predictors in COVID-19 Patients With Pre-existing Interstitial Lung Disease. Cureus 2022; 14:e27759. [PMID: 36106257 PMCID: PMC9448685 DOI: 10.7759/cureus.27759] [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] [Accepted: 08/07/2022] [Indexed: 11/05/2022] Open
Abstract
Background The data on the impact of coronavirus disease 2019 (COVID-19) on interstitial lung disease (ILD) is still limited. To the best of our knowledge, there has been no study from India to date to assess the impact of COVID-19 in patients with preexisting ILD. We undertook this study to assess the clinical outcome of ILD patients admitted to our hospital with COVID-19. Methods In this retrospective observational study, records of reverse transcription polymerase chain reaction (RT-PCR)-confirmed COVID-19 patients with preexisting ILD who were admitted to the hospital in the period from May 1, 2020, to April 30, 2021, were obtained from the hospital database. The clinical outcomes of the patients were recorded. Univariate analysis was performed to find relation between various predetermined risk factors for mortality and those with significant p values (p<0.05) were subjected to multiple logistic regression to determine independent risk factors. Results In our study of 28 patients, the overall mortality was 35.7%. On comparing the parameters associated with increased mortality, there was no effect of age, gender, comorbidities, type of ILD, CT thorax findings on diagnosis, use of corticosteroids and antifibrotics in the past, spirometric findings on mortality. On multivariate analysis, the significant parameters were interleukin 6 (IL-6), p=0.02, OR=1.020 (1.006-1.043) and D-dimer, p=0.04, OR=2.14 (5.55-1.14). Conclusion COVID-19 in patients with pre-existing ILD has a comparatively higher mortality. D-dimer and IL-6 are significant predictors of mortality in ILD patients infected with COVID-19.
Collapse
|
39
|
Assessment of disease outcome measures in systemic sclerosis. Nat Rev Rheumatol 2022; 18:527-541. [PMID: 35859133 DOI: 10.1038/s41584-022-00803-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 01/08/2023]
Abstract
The assessment of disease activity in systemic sclerosis (SSc) is challenging owing to its heterogeneous manifestations across multiple organ systems, the variable rate of disease progression and regression, and the relative paucity of patients in early-phase therapeutic trials. Despite some recent successes, most clinical trials have failed to show efficacy, underscoring the need for improved outcome measures linked directly to disease pathogenesis, particularly applicable for biomarker studies focused on skin disease. Current outcome measures in SSc-associated interstitial lung disease and SSc skin disease are largely adequate, although advancing imaging technology and the incorporation of skin mRNA biomarkers might provide opportunities for earlier detection of the therapeutic effect. Biomarkers can further inform pathogenesis, enabling early phase trials to act as reverse translational studies through the incorporation of routine high-throughput sequencing.
Collapse
|
40
|
Treatment approach to connective tissue disease-associated interstitial lung disease. Curr Opin Pharmacol 2022; 65:102245. [PMID: 35662004 DOI: 10.1016/j.coph.2022.102245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/21/2022]
Abstract
Interstitial lung disease (ILD) is a common manifestation in connective tissue diseases (CTD), such as rheumatoid arthritis (RA), systemic sclerosis (SSc), and inflammatory myositis (IM). ILD is associated with significant morbidity and mortality in nearly all CTD highlighting the critical need for effective treatment strategies in this patient population. In this review, we will summarize the approach to treatment when there is concern for CTD-ILD and highlight recent advancements in therapeutics within various forms of CTD-ILD.
Collapse
|
41
|
Liu Y, Cheng L, Zhan H, Li H, Li X, Huang Y, Li Y. The Roles of Noncoding RNAs in Systemic Sclerosis. Front Immunol 2022; 13:856036. [PMID: 35464474 PMCID: PMC9024074 DOI: 10.3389/fimmu.2022.856036] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Noncoding RNAs (ncRNAs) constitute more than 90% of the RNAs in the human genome. In the past decades, studies have changed our perception of ncRNAs from “junk” transcriptional products to functional regulatory molecules that mediate critical processes, including chromosomal modifications, mRNA splicing and stability, and translation, as well as key signaling pathways. Emerging evidence suggests that ncRNAs are abnormally expressed in not only cancer but also autoimmune diseases, such as systemic sclerosis (SSc), and may serve as novel biomarkers and therapeutic targets for the diagnosis and treatment of SSc. However, the functions and underlying mechanisms of ncRNAs in SSc remain incompletely understood. In this review, we discuss the current findings on the biogenetic processes and functions of ncRNAs, including microRNAs and long noncoding RNAs, as well as explore emerging ncRNA-based diagnostics and therapies for SSc.
Collapse
Affiliation(s)
- Yongmei Liu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linlin Cheng
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haoting Zhan
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haolong Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaomeng Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuan Huang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Yongzhe Li,
| |
Collapse
|
42
|
Huang B, Li J, Zhao J. Screening and identification of potential biomarkers and therapeutic targets for systemic sclerosis-associated interstitial lung disease. Arch Rheumatol 2022; 36:548-559. [PMID: 35382367 PMCID: PMC8957772 DOI: 10.46497/archrheumatol.2021.8625] [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: 12/05/2020] [Accepted: 02/18/2021] [Indexed: 11/03/2022] Open
Abstract
Objectives This study aims to analyze gene expression in lung tissue and lung fibroblasts of patients with systemic sclerosis-associated interstitial lung disease (SSc-ILD) to identify potential biomarkers and therapeutic targets and to examine its possible role in the pathogenesis of SSc-ILD. Patients and methods We obtained datasets from Gene Expression Omnibus (GEO) database, and used Robust Rank Aggregation to calculate the co-expressed differentially-expressed-genes (DEGs) in three chips, then analyzed the function, signaling pathways and the protein-protein interaction network of the DEGs. Finally, we verified the DEGs related to SSc-ILD by three databases of Comparative Toxicogenomics Database (CTD), GENE, and DisGeNET, respectively. Results There were 16 co-expressed DEGs related to SSc-ILD in three GEO series, of which six genes were upregulated, and 10 genes were downregulated. The CTD included 29,936 genes related to SSc, and the GENE and DisGeNET databases had 429 genes related to SSc. Conclusion The results of gene differential expression analysis suggest that interleukin-6, chemokine ligand 2, intercellular adhesion molecule 1, tumor necrosis factor alpha-induced protein 3, pentraxin 3, and cartilage oligomeric matrix protein may be implicated in the pathogenesis of SSc-ILD and are expected to be potential biomarkers and therapeutic targets for SSc-ILD.
Collapse
Affiliation(s)
- Biqing Huang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College Hospital, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College Hospital, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
43
|
Stephens MR, Moore DF, Dau J, Jobbagy S, Neel VA, Bolster MB, Fedeles F. A case of generalized morphea profunda following SARS-CoV-2 infection. JAAD Case Rep 2022; 23:20-23. [PMID: 35308063 PMCID: PMC8917007 DOI: 10.1016/j.jdcr.2022.02.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Michael R. Stephens
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Duncan F. Moore
- Division of Rheumatology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Soma Jobbagy
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Victor A. Neel
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marcy B. Bolster
- Division of Rheumatology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Flavia Fedeles
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Correspondence to: Flavia Fedeles, MD, Dermatology Department, Massachusetts General Hospital, 50 Staniford Street, Suite 200, Boston, MA 02114-2517.
| |
Collapse
|
44
|
Aragona CO, Versace AG, Ioppolo C, La Rosa D, Lauro R, Tringali MC, Tomeo S, Ferlazzo G, Roberts WN, Bitto A, Irrera N, Bagnato G. Emerging Evidence and Treatment Perspectives from Randomized Clinical Trials in Systemic Sclerosis: Focus on Interstitial Lung Disease. Biomedicines 2022; 10:504. [PMID: 35203713 PMCID: PMC8962255 DOI: 10.3390/biomedicines10020504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/04/2022] Open
Abstract
Systemic sclerosis (SSc) is a complex rare autoimmune disease with heterogeneous clinical manifestations. Currently, interstitial lung disease (ILD) and cardiac involvement (including pulmonary arterial hypertension) are recognized as the leading causes of SSc-associated mortality. New molecular targets have been discovered and phase II and phase III clinical trials published in the last 5 years on SSc-ILD will be discussed in this review. Details on the study design; the drug tested and its dose; the inclusion and exclusion criteria of the study; the concomitant immunosuppression; the outcomes and the duration of the study were reviewed. The two most common drugs used for the treatment of SSc-ILD are cyclophosphamide and mycophenolate mofetil, both supported by randomized controlled trials. Additional drugs, such as nintedanib and tocilizumab, have been approved to slow pulmonary function decline in SSc-ILD. In this review, we discuss the therapeutic alternatives for SSc management, offering the option to customize the design of future studies to stratify SSc patients and provide a patient-specific treatment according to the new emerging pathogenic features of SSc-ILD.
Collapse
Affiliation(s)
- Caterina Oriana Aragona
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| | - Antonio Giovanni Versace
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| | - Carmelo Ioppolo
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| | - Daniela La Rosa
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| | - Rita Lauro
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| | - Maria Concetta Tringali
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| | - Simona Tomeo
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| | - Guido Ferlazzo
- Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy;
| | | | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| | - Gianluca Bagnato
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (C.O.A.); (A.G.V.); (C.I.); (D.L.R.); (R.L.); (M.C.T.); (S.T.); (A.B.); (N.I.)
| |
Collapse
|
45
|
iNKT cells can effectively inhibit IL-6 production by B cells in systemic sclerosis. Cytotherapy 2022; 24:482-488. [PMID: 35181242 DOI: 10.1016/j.jcyt.2021.12.002] [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: 07/17/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is a connective tissue disease with poorly understood pathogenesis and limited treatment options. Patient mortality is rooted predominantly in the development of pulmonary and cardiac complications. The overactivated immune system is assumed to sustain the inflammatory signature of this autoimmune disease. Here, we investigate the potential of immunoregulatory invariant natural killer T (iNKT) cells to inhibit proinflammatory B cell responses in an in vitro model of inflammation. METHODS B cells from healthy volunteers (n = 17) and patients with SSc (n = 15) were used for functional testing upon lipopolysaccharide (LPS) stimulation in a co-culture system with third-party iNKT cells. Cytokine production was measured with antibody-based immunoassays (ELISA) and intracellular cytokine staining. RESULTS iNKT cells strongly inhibited the production of proinflammatory interleukin-6 by B cells upon stimulation with LPS in both healthy volunteers and patients with SSc. In a Transwell assay, cell contact between B cells and iNKT cells proved necessary for this inhibitory effect. Similarly, blocking of CD1d on the surface of B cells abolished the immunoregulatory effect of iNKT cells on B cells. B cell subsets with higher expression of CD1d, namely unswitched memory B cells, were more susceptible to iNKT cell inhibition. CONCLUSION Our in vitro data underline the potential of iNKT cells in the control of SSc and provide a rationale for the use of novel iNKT cell-based therapeutic strategies in the context of autoimmune diseases.
Collapse
|
46
|
Cardoneanu A, Burlui AM, Macovei LA, Bratoiu I, Richter P, Rezus E. Targeting Systemic Sclerosis from Pathogenic Mechanisms to Clinical Manifestations: Why IL-6? Biomedicines 2022; 10:biomedicines10020318. [PMID: 35203527 PMCID: PMC8869570 DOI: 10.3390/biomedicines10020318] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/04/2022] Open
Abstract
Systemic sclerosis (SS) is a chronic autoimmune disorder, which has both cutaneous and systemic clinical manifestations. The disease pathogenesis includes a triad of manifestations, such as vasculopathy, autoimmunity, and fibrosis. Interleukin-6 (IL-6) has a special role in SS development, both in vascular damage and in the development of fibrosis. In the early stages, IL-6 participates in vascular endothelial activation and apoptosis, leading to the release of damage-associated molecular patterns (DAMPs), which maintain inflammation and autoimmunity. Moreover, IL-6 plays an important role in the development of fibrotic changes by mediating the transformation of fibroblasts into myofibroblasts. All of these are associated with disabling clinical manifestations, such as skin thickening, pulmonary fibrosis, pulmonary arterial hypertension (PAH), heart failure, and dysphagia. Tocilizumab is a humanized monoclonal antibody that inhibits IL-6 by binding to the specific receptor, thus preventing its proinflammatory and fibrotic actions. Anti-IL-6 therapy with Tocilizumab is a new hope for SS patients, with data from clinical trials supporting the favorable effect, especially on skin and lung damage.
Collapse
Affiliation(s)
- Anca Cardoneanu
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
- Correspondence:
| | - Alexandra Maria Burlui
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| | - Luana Andreea Macovei
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| | - Patricia Richter
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
47
|
Benfaremo D, Svegliati S, Paolini C, Agarbati S, Moroncini G. Systemic Sclerosis: From Pathophysiology to Novel Therapeutic Approaches. Biomedicines 2022; 10:biomedicines10010163. [PMID: 35052842 PMCID: PMC8773282 DOI: 10.3390/biomedicines10010163] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/30/2022] Open
Abstract
Systemic sclerosis (SSc) is a systemic, immune-mediated chronic disorder characterized by small vessel alterations and progressive fibrosis of the skin and internal organs. The combination of a predisposing genetic background and triggering factors that causes a persistent activation of immune system at microvascular and tissue level is thought to be the pathogenetic driver of SSc. Endothelial alterations with subsequent myofibroblast activation, excessive extracellular matrix (ECM) deposition, and unrestrained tissue fibrosis are the pathogenetic steps responsible for the clinical manifestations of this disease, which can be highly heterogeneous according to the different entity of each pathogenic step in individual subjects. Although substantial progress has been made in the management of SSc in recent years, disease-modifying therapies are still lacking. Several molecular pathways involved in SSc pathogenesis are currently under evaluation as possible therapeutic targets in clinical trials. These include drugs targeting fibrotic and metabolic pathways (e.g., TGF-β, autotaxin/LPA, melanocortin, and mTOR), as well as molecules and cells involved in the persistent activation of the immune system (e.g., IL4/IL13, IL23, JAK/STAT, B cells, and plasma cells). In this review, we provide an overview of the most promising therapeutic targets that could improve the future clinical management of SSc.
Collapse
Affiliation(s)
- Devis Benfaremo
- Clinica Medica, Department of Internal Medicine, Ospedali Riuniti “Umberto I-G.M. Lancisi-G. Salesi”, 60126 Ancona, Italy;
| | - Silvia Svegliati
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (C.P.); (S.A.)
| | - Chiara Paolini
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (C.P.); (S.A.)
| | - Silvia Agarbati
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (C.P.); (S.A.)
| | - Gianluca Moroncini
- Clinica Medica, Department of Internal Medicine, Ospedali Riuniti “Umberto I-G.M. Lancisi-G. Salesi”, 60126 Ancona, Italy;
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, 60126 Ancona, Italy; (S.S.); (C.P.); (S.A.)
- Correspondence:
| |
Collapse
|
48
|
Khanna D, Lin CJF, Furst DE, Wagner B, Zucchetto M, Raghu G, Martinez FJ, Goldin J, Siegel J, Denton CP. Long-Term Safety and Efficacy of Tocilizumab in Early Systemic Sclerosis-Interstitial Lung Disease: Open Label Extension of a Phase 3 Randomized Controlled Trial. Am J Respir Crit Care Med 2021; 205:674-684. [PMID: 34851799 DOI: 10.1164/rccm.202103-0714oc] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Tocilizumab, an anti-interleukin-6 receptor antibody, had no statistically significant effect on skin sclerosis but preserved lung function over 48 weeks in patients with early systemic sclerosis-associated interstitial lung disease in a phase 3 randomized controlled trial. OBJECTIVES Assess long-term safety and efficacy of tocilizumab. METHODS Adults with diffuse cutaneous systemic sclerosis for ≤60 months and elevated acute-phase reactants, including those with interstitial lung disease, received weekly placebo or tocilizumab 162 mg subcutaneously in the 48-week, double-blind period then open-label tocilizumab from weeks 48 to 96 (placebo-tocilizumab; continuous-tocilizumab). MEASUREMENTS AND MAIN RESULTS Eighty-two of 107 placebo-tocilizumab and 85 of 105 continuous-tocilizumab patients completed 96 weeks. Mean age and disease duration were 48 years and 23 months; high-resolution computed tomography revealed interstitial lung disease in 61%. Mean (95% CI) change in modified Rodnan skin score from baseline to week 96 was -8.4 (-10.0, -6.8) for placebo-tocilizumab and -9.6 (-10.9, -8.4) for continuous-tocilizumab. Mean (95% CI) change in forced vital capacity (percent-predicted) from baseline to week 96 was -3.3 (-5.1, -1.5) for placebo-tocilizumab and -0.5 (-2.4, 1.3) for continuous-tocilizumab among completers and, in post hoc analysis, -4.1 (-6.7, -1.6) and -0.6 (-3.1, 2.0), respectively, among completers with interstitial lung disease (mean [95% CI] change from weeks 48 to 96: 0.9 [-0.8, 2.7] and -0.4 [-2.3, 1.5], respectively). Rates per 100 patient-years of serious adverse events from weeks 48 to 96 were 14.8 for placebo-tocilizumab and 15.8 for continuous-tocilizumab. CONCLUSIONS Tocilizumab preserved lung function, slowing decline in forced vital capacity, in patients with systemic sclerosis, including those with interstitial lung disease. Long-term safety was consistent with the known safety profile of tocilizumab. TRIAL REGISTRATION NUMBER ClinicalTrials.gov, NCT01791153.
Collapse
Affiliation(s)
- Dinesh Khanna
- University of Michigan, Ann Arbor, Michigan, United States;
| | - Celia J F Lin
- Genentech Inc, 7412, South San Francisco, California, United States
| | - Daniel E Furst
- University of California Los Angeles, 8783, Los Angeles, California, United States
| | - Bridget Wagner
- Genentech Inc, 7412, South San Francisco, California, United States
| | | | - Ganesh Raghu
- University of Washington, 7284, Seattle, Washington, United States
| | | | - Jonathan Goldin
- UCLA School Of Medicine, Los Angeles, California, United States
| | - Jeffrey Siegel
- Genentech Inc, 7412, South San Francisco, California, United States
| | - Christopher P Denton
- University College London, 4919, London, United Kingdom of Great Britain and Northern Ireland
| | | |
Collapse
|
49
|
Hinchcliff M, Garcia-Milian R, Di Donato S, Dill K, Bundschuh E, Galdo FD. Cellular and Molecular Diversity in Scleroderma. Semin Immunol 2021; 58:101648. [PMID: 35940960 DOI: 10.1016/j.smim.2022.101648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With the increasing armamentarium of high-throughput tools available at manageable cost, it is attractive and informative to determine the molecular underpinnings of patient heterogeneity in systemic sclerosis (SSc). Given the highly variable clinical outcomes of patients labelled with the same diagnosis, unravelling the cellular and molecular basis of disease heterogeneity will be crucial to predicting disease risk, stratifying management and ultimately informing a patient-centered precision medicine approach. Herein, we summarise the findings of the past several years in the fields of genomics, transcriptomics, and proteomics that contribute to unraveling the cellular and molecular heterogeneity of SSc. Expansion of these findings and their routine integration with quantitative analysis of histopathology and imaging studies into clinical care promise to inform a scientifically driven patient-centred personalized medicine approach to SSc in the near future.
Collapse
Affiliation(s)
- Monique Hinchcliff
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA.
| | | | - Stefano Di Donato
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK
| | | | - Elizabeth Bundschuh
- Yale School of Medicine, Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, USA
| | - Francesco Del Galdo
- Raynaud's and Scleroderma Programme, Leeds Institute of Rheumatic and Musculoskeletal Medicine and NIHR Biomedical Research Centre, University of Leeds, UK.
| |
Collapse
|
50
|
Sen’kova AV, Savin IA, Brenner EV, Zenkova MA, Markov AV. Core genes involved in the regulation of acute lung injury and their association with COVID-19 and tumor progression: A bioinformatics and experimental study. PLoS One 2021; 16:e0260450. [PMID: 34807957 PMCID: PMC8608348 DOI: 10.1371/journal.pone.0260450] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is a specific form of lung damage caused by different infectious and non-infectious agents, including SARS-CoV-2, leading to severe respiratory and systemic inflammation. To gain deeper insight into the molecular mechanisms behind ALI and to identify core elements of the regulatory network associated with this pathology, key genes involved in the regulation of the acute lung inflammatory response (Il6, Ccl2, Cat, Serpine1, Eln, Timp1, Ptx3, Socs3) were revealed using comprehensive bioinformatics analysis of whole-genome microarray datasets, functional annotation of differentially expressed genes (DEGs), reconstruction of protein-protein interaction networks and text mining. The bioinformatics data were validated using a murine model of LPS-induced ALI; changes in the gene expression patterns were assessed during ALI progression and prevention by anti-inflammatory therapy with dexamethasone and the semisynthetic triterpenoid soloxolone methyl (SM), two agents with different mechanisms of action. Analysis showed that 7 of 8 revealed ALI-related genes were susceptible to LPS challenge (up-regulation: Il6, Ccl2, Cat, Serpine1, Eln, Timp1, Socs3; down-regulation: Cat) and their expression was reversed by the pre-treatment of mice with both anti-inflammatory agents. Furthermore, ALI-associated nodal genes were analysed with respect to SARS-CoV-2 infection and lung cancers. The overlap with DEGs identified in postmortem lung tissues from COVID-19 patients revealed genes (Saa1, Rsad2, Ifi44, Rtp4, Mmp8) that (a) showed a high degree centrality in the COVID-19-related regulatory network, (b) were up-regulated in murine lungs after LPS administration, and (c) were susceptible to anti-inflammatory therapy. Analysis of ALI-associated key genes using The Cancer Genome Atlas showed their correlation with poor survival in patients with lung neoplasias (Ptx3, Timp1, Serpine1, Plaur). Taken together, a number of key genes playing a core function in the regulation of lung inflammation were found, which can serve both as promising therapeutic targets and molecular markers to control lung ailments, including COVID-19-associated ALI.
Collapse
Affiliation(s)
- Aleksandra V. Sen’kova
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Innokenty A. Savin
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgenyi V. Brenner
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Marina A. Zenkova
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Andrey V. Markov
- Laboratory of Nucleic Acids Biochemistry, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|