1
|
Zhang L, Xu F, Hou L. IL-6 and diabetic kidney disease. Front Immunol 2024; 15:1465625. [PMID: 39749325 PMCID: PMC11693507 DOI: 10.3389/fimmu.2024.1465625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Diabetic kidney disease (DKD) is a severe microvascular complication of diabetes associated with high mortality and disability rates. Inflammation has emerged as a key pathological mechanism in DKD, prompting interest in novel therapeutic approaches targeting inflammatory pathways. Interleukin-6 (IL-6), a well-established inflammatory cytokine known for mediating various inflammatory responses, has attracted great attention in the DKD field. Although multiple in vivo and in vitro studies highlight the potential of targeting IL-6 in DKD treatment, its exact roles in the disease remains unclear. This review presents the roles of IL-6 in the pathogenesis of DKD, including immunoinflammation, metabolism, hemodynamics, and ferroptosis. In addition, we summarize the current status of IL-6 inhibitors in DKD-related clinical trials and discuss the potential of targeting IL-6 for treating DKD in the clinic.
Collapse
Affiliation(s)
- Lei Zhang
- Pharmacy Department, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| | - Futian Xu
- Logistics Management Department, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| | - Liyan Hou
- Pharmacy Department, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| |
Collapse
|
2
|
Bale S, Verma P, Yalavarthi B, Bajželj M, Hasan SA, Silverman JN, Broderick K, Shah KA, Hamill T, Khanna D, Sigalov AB, Bhattacharyya S, Varga J. Inhibiting triggering receptor expressed on myeloid cells 1 signaling to ameliorate skin fibrosis. JCI Insight 2024; 9:e176319. [PMID: 39418109 PMCID: PMC11623937 DOI: 10.1172/jci.insight.176319] [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: 10/25/2023] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
Systemic sclerosis (SSc) is characterized by immune system failure, vascular insult, autoimmunity, and tissue fibrosis. TGF-β is a crucial mediator of persistent myofibroblast activation and aberrant extracellular matrix production in SSc. The factors responsible for this are unknown. By amplifying pattern recognition receptor signaling, triggering receptor expressed on myeloid cells 1 (TREM-1) is implicated in multiple inflammatory conditions. In this study, we used potentially novel ligand-independent TREM-1 inhibitors in preclinical models of fibrosis and explanted SSc skin fibroblasts in order to investigate the pathogenic role of TREM-1 in SSc. Selective pharmacological TREM-1 blockade prevented and reversed skin fibrosis induced by bleomycin in mice and mitigated constitutive collagen synthesis and myofibroblast features in SSc fibroblasts in vitro. Our results implicate aberrantly activated TREM-1 signaling in SSc pathogenesis, identify a unique approach to TREM-1 blockade, and suggest a potential therapeutic benefit for TREM-1 inhibition.
Collapse
Affiliation(s)
- Swarna Bale
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Priyanka Verma
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Bharath Yalavarthi
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Matija Bajželj
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Syed A.M. Hasan
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jenna N. Silverman
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Katherine Broderick
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kris A. Shah
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Timothy Hamill
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Dinesh Khanna
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Swati Bhattacharyya
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - John Varga
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Samulevich ML, Carman LE, Aneskievich BJ. Critical Analysis of Cytoplasmic Progression of Inflammatory Signaling Suggests Potential Pharmacologic Targets for Wound Healing and Fibrotic Disorders. Biomedicines 2024; 12:2723. [PMID: 39767629 PMCID: PMC11726985 DOI: 10.3390/biomedicines12122723] [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: 10/30/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Successful skin wound healing is dependent on an interplay between epidermal keratinocytes and dermal fibroblasts as they react to local extracellular factors (DAMPs, PAMPs, cytokines, etc.) surveyed from that environment by numerous membrane receptors (e.g., TLRs, cytokine receptors, etc.). In turn, those receptors are the start of a cytoplasmic signaling pathway where balance is key to effective healing and, as needed, cell and matrix regeneration. When directed through NF-κB, these signaling routes lead to transient responses to the benefit of initiating immune cell recruitment, cell replication, local chemokine and cytokine production, and matrix protein synthesis. The converse can also occur, where ongoing canonical NF-κB activation leads to chronic, hyper-responsive states. Here, we assess three key players, TAK1, TNFAIP3, and TNIP1, in cytoplasmic regulation of NF-κB activation, which, because of their distinctive and yet inter-related functions, either promote or limit that activation. Their balanced function is integral to successful wound healing, given their significant control over the expression of inflammation-, fibrosis-, and matrix remodeling-associated genes. Intriguingly, these three proteins have also been emphasized in dysregulated NF-κB signaling central to systemic sclerosis (SSc). Notably, diffuse SSc shares some tissue features similar to an excessive inflammatory/fibrotic wound response without eventual resolution. Taking a cue from certain instances of aberrant wound healing and SSc having some shared aspects, e.g., chronic inflammation and fibrosis, this review looks for the first time, to our knowledge, at what those pathologies might have in common regarding the cytoplasmic progression of NF-κB-mediated signaling. Additionally, while TAK1, TNFAIP3, and TNIP1 are often investigated and reported on individually, we propose them here as three proteins whose consequences of function are very highly interconnected at the signaling focus of NF-κB. We thus highlight the emerging promise for the eventual clinical benefit derived from an improved understanding of these integral signal progression modulators. Depending on the protein, its indirect or direct pharmacological regulation has been reported. Current findings support further intensive studies of these points in NF-κB regulation both for their basic function in healthy cells as well as with the goal of targeting them for translational benefit in multiple cutaneous wound healing situations, whether stemming from acute injury or a dysregulated inflammatory/fibrotic response.
Collapse
Affiliation(s)
- Michael L. Samulevich
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (M.L.S.); (L.E.C.)
| | - Liam E. Carman
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (M.L.S.); (L.E.C.)
| | - Brian J. Aneskievich
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
4
|
Zinellu A, Mangoni AA. Vascular endothelial growth factor as a potential biomarker in systemic sclerosis: a systematic review and meta-analysis. Front Immunol 2024; 15:1442913. [PMID: 39669565 PMCID: PMC11634811 DOI: 10.3389/fimmu.2024.1442913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024] Open
Abstract
Introduction Systemic sclerosis (SSc), a chronic autoimmune condition, is characterized by microvascular dysfunction, ineffective angiogenesis, and fibrosis. The identification of robust biomarkers reflecting these processes may assist in clinical management and lead to the discovery of new therapies. We sought to address this issue by conducting a systematic review and meta-analysis of studies investigating one such biomarker, vascular endothelial growth factor (VEGF), in SSc patients and healthy controls and in SSc patients with localized or diffuse disease, different video capillaroscopy patterns (early, active, or late), and presence or absence of complications. Methods We searched PubMed, Scopus, and Web of Science from inception to 15 May 2024. We assessed the risk of bias and the certainty of evidence using the JBI checklist for analytical studies and GRADE, respectively. Results In 42 eligible studies, compared to controls, patients with SSc had significantly higher plasma or serum VEGF concentrations (standard mean difference, SMD=0.93, 95% CI 0.71 to 1.15, p<0.001; moderate certainty). In further analyses, VEGF concentrations were significantly higher in SSc patients with diffused disease than those with localized disease (SMD=0.30, 95% CI 0.01 to 0.59, p=0.046; very low certainty), in patients with late vs. active video capillaroscopy pattern (SMD=0.35, 95% CI 0.09 to 0.61, p=0.008; very low certainty), and in patients with pulmonary hypertension than those without (SMD=0.93, 95% CI 0.34 to 1.53, p=0.002; very low certainty). By contrast, no significant differences were observed between SSc patients with and without digital ulcers, interstitial lung disease, and telangiectasias, whereas limited evidence was available for alveolitis. Meta-regression and subgroup analysis of studies investigating VEGF in SSc patients and controls showed no significant associations between the effects size and various patient and study characteristics, including SSc duration and use of corticosteroids, immunosuppressors and vasodilators. By contrast, significant associations were observed with the geographical location where the study was conducted. Discussion The results of this systematic review and meta-analysis suggest that VEGF can be useful in the assessment and management of SSc and in the identification of novel therapeutic strategies in this patient group. Systematic Review Registration https://www.crd.york.ac.uk/prospero, identifier CRD42024552925.
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A. Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
- Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Bedford Park, SA, Australia
| |
Collapse
|
5
|
Ono Y, Mogami A, Saito R, Seki N, Ishigaki S, Takei H, Yoshimoto K, Chiba K, Takeuchi T, Kaneko Y. Interleukin-17A is a potential therapeutic target predicted by proteomics for systemic sclerosis patients at high risk of pulmonary arterial hypertension. Sci Rep 2024; 14:29484. [PMID: 39604413 PMCID: PMC11603215 DOI: 10.1038/s41598-024-76987-6] [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: 07/24/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
We explored effective therapeutic targets for systemic sclerosis (SSc) patients with high risk for pulmonary arterial hypertension (PAH) by plasma proteomics analysis. A total of fifty-seven patients with SSc were enrolled in the study and the prevalence of PAH was 19.3%. On the other hand, 75.4% of SSc patients showed the ratio of forced vital capacity percentage/diffusing capacity of the lungs for carbon monoxide percentage> 1.6 and met criteria for high risk of PAH. Identification of elevated plasma proteins in SSc patients with high risk of PAH, followed by upstream regulator analysis, predicted interleukin (IL)-17A as a major upstream molecule. Furthermore, we performed in vitro neutralization study using MT-6194, a bispecific antibody targeting both IL-17A and IL-6 receptor. We found that MT-6194 broadly suppressed the increased expression of downstream molecules of IL-17A including IL-17A-related cytokines/chemokines, IL-17A-driven NFκB pathway and IL-6-driven JAK/STAT pathway which were shown to be increased in SSc patients with high risk of PAH by the proteomics. Consequently, it is revealed that IL-17A is a promising target for early intervention in SSc patients with high risk for PAH.
Collapse
Affiliation(s)
- Yuuichi Ono
- Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Kanagawa, 227-0033, Japan
| | - Akira Mogami
- Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Kanagawa, 227-0033, Japan
| | - Ryuta Saito
- Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Kanagawa, 227-0033, Japan
| | - Noriyasu Seki
- Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Kanagawa, 227-0033, Japan
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Sho Ishigaki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hiroshi Takei
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Keiko Yoshimoto
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kenji Chiba
- Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Kanagawa, 227-0033, Japan.
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Saitama Medical University, Iruma-gun, Saitama, 350-0495, Japan
| | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, 160-8582, Japan
| |
Collapse
|
6
|
Meng Y, Han X, Ji Q, Zhang A, Zhan Y, Chen X. Trends of mortality from chronic respiratory diseases by sex and ethnicity in the USA: a secular analysis from 1979 to 2021 using data from death certificates. BMJ Open Respir Res 2024; 11:e002296. [PMID: 39581694 PMCID: PMC11590864 DOI: 10.1136/bmjresp-2024-002296] [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: 01/06/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND Over the past decades, the prevalence of chronic respiratory diseases (CRDs) has undergone significant changes. Analysing long-term mortality trends by sex and ethnicity can inform more targeted public health interventions. METHODS Using data from death certificates, we calculated age-standardised mortality rates of chronic obstructive pulmonary disease (COPD), interstitial lung disease (ILD), asthma and pneumoconiosis among the US population from 1979 to 2021, stratified by sex and ethnicity. Trend analyses of mortality rates were conducted using joinpoint regression models. RESULTS Mortality trends varied significantly by demographic groups and disease types. Specifically, COPD mortality exhibited an overall increasing trend among females and white individuals throughout the study period while remaining stable among males and black individuals. ILD mortality showed an overall increasing trends for both males and females as well as individuals of white ethnicity. Asthma mortality showed an increase before 1996 followed by a consistent decline across all populations from about 1996 to 2021. In contrast, pneumoconiosis mortality has risen among white individuals since 2001 with a notable increase among females after 2009. CONCLUSION Our analysis reveals fluctuating trends in CRD mortality rates from 1979 to 2021 with varying patterns across sex, ethnicity and disease types. These findings underscore the need for ongoing research and tailored interventions to address these disparities.
Collapse
Affiliation(s)
- Yaxian Meng
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Xiaojie Han
- Department of Chronic Disease Control, Guangming Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Qianqian Ji
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Aijie Zhang
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yiqiang Zhan
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Xiaoliang Chen
- Department of Chronic Disease Control, Guangming Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Chen S, Fan Y, Wu Q, Zhang G, Wang Y, Li W, Yang S, Matucci-Cerinic M, Furst DE. Integrative Transcriptomic Analysis of Peripheral Blood Monocytes in Systemic Sclerosis and Shared Pathogenic Pathways in Autoimmune Diseases. Arch Med Res 2024; 56:103072. [PMID: 39208548 DOI: 10.1016/j.arcmed.2024.103072] [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: 03/30/2024] [Revised: 07/04/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Systemic sclerosis (SSc) is an autoimmune disease (AD), that receives less attention compared to rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and primary Sjögren's syndrome (pSS). This study aims to analyze transcriptional profiles and immune cell composition in peripheral blood mononuclear cells (PBMC) from SSc patients compared to other ADs. METHODS RNA-seq data from 119 untreated patients (eight with SSc, 42 with RA, 41 with pSS, 28 with SLE) and 20 healthy controls were analyzed. Bioinformatics tools were employed to identify differentially expressed genes (DEGs), biological functions and immune cell profiles unique to SSc and shared with other ADs. RESULTS 1,148 DEGs were found in SSc, with upregulated genes associated with megakaryocyte processes and downregulated genes associated with neutrophil function and immune response. DEGs, including ALDH1A1 and MEGF9, were associated with neutropenia. Upregulated transcription factors (TFs) were linked to embryonic hematopoiesis and downregulated TFs were involved in leukocyte differentiation and immune regulation. Comparative analysis with other ADs revealed common pathogenic pathways, emphasizing megakaryocyte proliferation. Neutrophils count was significantly decreased in ADs (p < 0.001) compared to healthy controls. Comparative analysis highlighted common pathways, particularly in megakaryocyte proliferation, and unique genes (MEGF9, MMP8, and KRT family members) in SSc, suggesting roles in neutrophil function, skin integrity, and fibrosis. CONCLUSIONS This study identifies dysregulated gene expression (KRT and MMP8) associated with neutrophil function and increased megakaryocytes in SSc, highlighting common patterns across autoimmune diseases. These findings offer new insights into the potential pathogenesis of SSc, and help to explore new targets for the treatment.
Collapse
Affiliation(s)
- Shaoqi Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yu Fan
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Qiulin Wu
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yukai Wang
- Department of Rheumatology and Immunology, Shantou Central Hospital, Shantou, Guangdong, China
| | - Weiping Li
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| | - Shengli Yang
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China; Shanghai Academician Consulting and Academic Activities Center of Chinese Academy of Engineering, Shanghai, China.
| | - Marco Matucci-Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, San Raffaele Hospital, Milan, Italy
| | - Daniel E Furst
- Division of Rheumatology, School of Medicine, University of California at Los Angeles, California, USA
| |
Collapse
|
8
|
Ananyeva LP, Garzanova LA, Desinova OV, Shayakhmetova RU, Starovoitova MN, Koneva OA, Ovsyannikova OB, Glukhova SI, Nasonov EL. The Use of "Acellbia"-A Biosimilar of Rituximab in Systemic Sclerosis. DOKL BIOCHEM BIOPHYS 2024; 517:140-147. [PMID: 38861146 DOI: 10.1134/s1607672924700844] [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: 04/03/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 06/12/2024]
Abstract
The possibilities of modern therapy for systemic sclerosis (SSc) remains limited, since most of the used drugs do not have a disease-modifying effect. This encourages the study of new approaches that potentially affect the fundamental pathological processes underlying the disease. One example is anti-B-cell therapy, in particular rituximab (RTX). Until now RTX does not have a registration for the treatment of SSc, but there is a large positive experience of its use, which is reflected in recent meta-analyses and clinical recommendations. Complicated and expensive methods for obtaining genetically engineered biological drugs (biologics) have contributed to the emergence of more accessible biosimilars, one of which is the RTX biosimilar, Acellbia (Biocad, Russian Federation). The ''biosimilar'' versions of RTX might reduce the cost of therapy and increase patients accessibility to this treatment option. The RTX biosimilar Acellbia (ACB) has received approval in Russian Federation in 2014 for all indications held by reference RTX (including rheumatoid arthritis and ANCA-associated vasculitis).
Collapse
Affiliation(s)
- L P Ananyeva
- Nasonova Research Institute of Rheumatology, Moscow, Russia.
| | - L A Garzanova
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - O V Desinova
- Nasonova Research Institute of Rheumatology, Moscow, Russia.
| | | | | | - O A Koneva
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | | | - S I Glukhova
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - E L Nasonov
- Nasonova Research Institute of Rheumatology, Moscow, Russia
- Sechenov First Moscow State Medical University of the Ministry of Health Care of Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
9
|
Garzanova LA, Ananyeva LP, Koneva OA, Desinova OV, Starovoytova MN, Ovsyannikova OB, Shayakhmetova RU, Glukhova SI. Safety and Tolerability of Rituximab in the Treatment of Systemic Sclerosis. DOKL BIOCHEM BIOPHYS 2024; 517:156-165. [PMID: 38861145 DOI: 10.1134/s1607672924700856] [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: 04/03/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 06/12/2024]
Abstract
Rituximab (RTX) has been used for the treatment of systemic sclerosis (SSс) for a long time and has shown good efficacy for skin fibrosis and interstitial lung disease (ILD). However, data on tolerability and long-term adverse events (AEs) during RTX therapy in SSc are insufficient. The objective of this study was to assess the tolerability and safety of RTX in patients with SSс in a long-term prospective follow-up. Our open-label prospective study included 151 SSс patients who received at least one RTX infusion. The mean age of the patients was 47.9 ± 13.4 years; the majority of them were women (83%). The mean disease duration was 6.4 ± 5.8 years. The mean follow-up period after the first RTX infusion was 5.6 ± 2.6 years (845.6 patient-years (PY)). All patients received RTX in addition to ongoing therapy with prednisone and/or immunosuppressants. AEs were assessed and recorded by a doctor in the hospital immediately after RTX infusion and then by patient's reported outcome during the observation period. All causes of death were considered, regardless of treatment. A total of 85 AEs (56%) were registered, the overall incidence of AEs was 10/100 PY (95% confidence interval (CI) 8-12). The highest frequency of all AEs was observed in the first 2-6 months after the first course of RTX, however, these were mainly mild and moderate AEs (71%). The most frequent AEs were infections, they were observed in 40% of cases, with no serious opportunistic infections reported. The overall incidence of all infections was 7.1/100 PY (95% CI 5.5-9), serious infections-1.5/100 PY (95% CI 0.9-2.6). Infusion reactions occurred in 8% of patients. Other AEs were noted in 3% (0.6/100 PY, 95% CI 0.3-1.4). The overall incidence of serious AEs was 18%-3.2/100 PY (95% CI 2.2-4.6). There was a significant decrease of the immunoglobulin G (Ig G) during follow-up; however, its average values remained within normal limits. There were 17 deaths (11%) (2/100 PY, 95% CI 1.3-3.2). In most cases, patients died from the progression of the major organ failure, which arose before RTX treatment. In our study, the safety profile of RTX in SSс was assessed as favorable. It was similar to the AE profile in other autoimmune diseases treated with RTX. With an increase in the cumulative dose of RTX, no increase in AEs was observed. The mortality is comparable to the other severe autoimmune diseases in observational studies. Monitoring of IgG may be useful for patients with SSс on RTX therapy for early detection of the risk of developing infectious complications. RTX could be considered as a relatively safe drug for the complex therapy of SSс when standard therapy is ineffective or impossible.
Collapse
Affiliation(s)
- L A Garzanova
- Nasonova Research Institute of Rheumatology, Moscow, Russia.
| | - L P Ananyeva
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - O A Koneva
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | - O V Desinova
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| | | | | | | | - S I Glukhova
- Nasonova Research Institute of Rheumatology, Moscow, Russia
| |
Collapse
|
10
|
Parikh A, Francis AJ, Sreenivasan C, Pandey M, AlQassab O, Kanthajan T, AlEdani EM. Efficacy of Rituximab Versus Cyclophosphamide and Mycophenolate for the Treatment of Interstitial Lung Disease in Systemic Sclerosis: A Systematic Review. Cureus 2024; 16:e68279. [PMID: 39350831 PMCID: PMC11441839 DOI: 10.7759/cureus.68279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Interstitial lung disease (ILD) is a common complication of systemic sclerosis (SSc), contributing to significant morbidity and mortality in affected individuals. The optimal treatment approach for SSc-associated ILD remains uncertain, with rituximab, cyclophosphamide, and mycophenolate among potential therapeutic options. This systematic review aims to evaluate and synthesize the existing evidence on the efficacy of rituximab compared to cyclophosphamide and mycophenolate for the treatment of ILD in patients with systemic sclerosis. A comprehensive search of the following electronic databases, PubMed, Science Direct, Google Scholar, and Cochrane Library, has been conducted to identify relevant studies, including randomized controlled trials, systematic review and meta-analysis, prospective cohort studies, and retrospective cohort studies. Data on study characteristics, participant demographics, interventions, outcomes, and key findings have been extracted and synthesized. The risk of bias in the included studies has been assessed using appropriate tools such as the Cochrane Bias assessment tool for randomized controlled trials, the New Castle Ottawa tool for cohort studies, and the AMSTAR checklist for systematic reviews and meta-analysis. The research team ultimately selected 15 high-quality studies for review. Rituximab demonstrated similar efficacy to cyclophosphamide and mycophenolate in improving lung function (forced vital capacity (FVC) and diffusing capacity of the lung for carbon monoxide (DLCO)), with fewer severe adverse events. Cyclophosphamide, while effective, had higher toxicity, leading to more frequent adverse events such as leukopenia and infections. Mycophenolate showed comparable efficacy to cyclophosphamide but with fewer side effects, making it a well-tolerated alternative. The findings of this systematic review will provide valuable insights into the comparative efficacy of rituximab, cyclophosphamide, and mycophenolate in the management of ILD in systemic sclerosis, informing clinical decision-making and guiding future research in this area.
Collapse
Affiliation(s)
- Aneri Parikh
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Aida J Francis
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Chithra Sreenivasan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Manorama Pandey
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Osamah AlQassab
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Tatchaya Kanthajan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Esraa M AlEdani
- Dermatology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
11
|
Kudsi M, Tarcha R, Khalayli N. Nintedanib in systemic sclerosis treatment: a case report. J Med Case Rep 2024; 18:110. [PMID: 38388392 PMCID: PMC10885361 DOI: 10.1186/s13256-024-04433-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/10/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Nintedanib was approved for the treatment of scleroderma and scleroderma-related interstitial lung disease, as it decrease the forced expiratory volume. CASE PRESENTATION A 48-year-old Asian female patient with systemic scleroderma 6 years ago developed breathlessness, nausea, heart palpation, and sudden severe occipital headache over the preceding week. She was receiving aspirin 81 mg/day and amlodipine 5 mg/day. Her diagnosis was diffuse scleroderma with pulmonary hypertension, interstitial lung involvement, and renal crisis. The modified Rodnan score was 18. We begin captopril at a dose of 12.5 mg, progressively escalating to 200 mg/day, and oral nintedanib was started at 150 mg. A total of 12 months after initiation of treatment, the patient's kidney function was normal. The pulmonary function tests improved. The modified Rodnan score was reduced to 10. We did not encounter any side effects in our case due to nintedanib treatment. CONCLUSION Treatment with nintedanib is crucial for slowing lung function decline. Diarrhea was the most common adverse event. Scleroderma renal crisis occurs in 10% of patients and typically presents with an abrupt onset of hypertension and kidney failure. The optimal antihypertensive agent for scleroderma renal crisis is an ACE inhibitor. The mainstay of therapy in scleroderma renal crisis has been shown to improve or stabilize renal function in approximately 70% of patients and improve survival in nearly 80% at 1 year. Nintedanib may be effective, and fairly safe to use. Further exploration is anticipated to advance a new period of systemic sclerosis treatment.
Collapse
Affiliation(s)
- Maysoun Kudsi
- Department of Rheumatology, Faculty of Medicine, Damascus University, Al-Mazzeh, Damascus, Syria
| | - Raghad Tarcha
- Department of Rheumatology, Faculty of Medicine, Damascus University, Al-Mazzeh, Damascus, Syria.
| | - Naram Khalayli
- Faculty of Medicine, Damascus University, Damascus, Syria
| |
Collapse
|
12
|
Windirsch K, Jordan S, Becker MO, Bruni C, Dobrota R, Elhai M, Garaiman IA, Mihai CM, Iudici M, Hasler P, Ribi C, Maurer B, Gabrielli A, Hoffmann-Vold AM, Distler O. Therapeutic management of fibrosis in systemic sclerosis patients - an analysis from the Swiss EUSTAR cohort. Swiss Med Wkly 2024; 154:3630. [PMID: 38579324 DOI: 10.57187/s.3630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024] Open
Abstract
OBJECTIVES Systemic sclerosis is a chronic autoimmune connective tissue disease leading to microvascular and fibrotic manifestations in multiple organs. Several treatment options and recommendations from different European countries are available. In this study, for which the ambit is Switzerland specifically, we aim to describe the treatment patterns of systemic sclerosis patients with fibrotic manifestations. METHODS Systemic sclerosis patients were selected from six Swiss tertiary centres recorded in the multicentre, prospective European Scleroderma Trials and Research (EUSTAR) registry. Patients fulfilling the 2013 ACR/EULAR systemic sclerosis classification criteria at baseline were included. To determine the differences in treatment of varying degrees of fibrosis, four groups were identified: (1) patients with a modified Rodnan skin score (mRSS) >0; (2) those with mRSS ≥7; (3) those with interstitial lung disease (SSc-ILD), diagnosed by either chest X-Ray or high-resolution computed tomography; and (4) patients fulfilling one of the additional criteria for extensive interstitial lung disease, defined as interstitial lung disease involvement of >20% in high-resolution computed tomography, dyspnea NYHA-stage 3/4, or a predicted forced vital capacity (FVC) of <70%. RESULTS A total of 590 patients with systemic sclerosis fulfilled the inclusion criteria. In this cohort, 421 (71.4%) had mRSS >0, of whom 195 (33.1%) had mRSS ≥7; interstitial lung disease was diagnosed in 198 of 456 (43.4%), of whom 106 (18.0 %) showed extensive interstitial lung disease. Regarding non-biologic disease-modifying medications (DMARDs), the most frequently prescribed was methotrexate, followed by hydroxychloroquine and mycophenolate mofetil. Rituximab and tocilizumab were most frequently used among the biologic DMARDs. Specifically, 148/372 (39.8%) of treated patients with skin fibrosis received methotrexate, mycophenolate mofetil or rituximab, and 80/177 (45.2%) with interstitial lung disease received cyclophosphamide, mycophenolate mofetil, tocilizumab or rituximab. Most patients received a proton-pump inhibitor, and few patients underwent hematopoietic stem cell transplantation. CONCLUSION Overall, in Switzerland, a wide range of medications is prescribed for systemic sclerosis patients. This includes modern, targeted treatments for which randomised controlled clinical trial have been recently reported.
Collapse
Affiliation(s)
- Kevin Windirsch
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Suzana Jordan
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mike Oliver Becker
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cosimo Bruni
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Rucsandra Dobrota
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Muriel Elhai
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ion-Alexandru Garaiman
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Carmen-Marina Mihai
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michele Iudici
- Division of Rheumatology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Paul Hasler
- Department of Rheumatology,Kantonsspital Aarau, Aarau, Switzerland
| | - Camillo Ribi
- Department of Clinical Immunology and Allergy, Centre hospitalier universitaire vaudois (CHUV), Lausanne, Switzerland
| | - Britta Maurer
- Department of Rheumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Armando Gabrielli
- Marche Polytechnic University, Institute of Clinical Medicine, University of Ancona, Ancona, Italy
| | | | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Cho A, Paulitschke V, Knobler R. Mode of action, indications and recommendations on extracorporeal photopheresis (ECP). J Dtsch Dermatol Ges 2023; 21:1369-1380. [PMID: 37723908 DOI: 10.1111/ddg.15167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/02/2023] [Indexed: 09/20/2023]
Abstract
Extracorporeal photopheresis (ECP) has gained importance in the treatment of several diseases. Initially introduced as a new therapeutic modality for the treatment of patients with cutaneous T-cell lymphoma, the indications for the use of ECP have expanded to include hematology and transplantation immunology. Extracorporeal photopheresis has found its place in the treatment plan of cutaneous T-cell lymphoma, systemic sclerosis, graft-versus-host disease, organ transplantation such as heart and lung, sometimes as first-line therapy and very often in combination with various systemic immunosuppressive therapies. The procedure basically consists of three steps: leukapheresis, photoactivation and reinfusion. The following article presents possible theories about the mechanism of action, which is not yet fully understood, and discusses the five most common indications for ECP treatment with corresponding therapy recommendations.
Collapse
Affiliation(s)
- Ara Cho
- University Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Verena Paulitschke
- University Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Robert Knobler
- University Department of Dermatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Cho A, Paulitschke V, Knobler R. Wirkweise, Indikationen und Therapieempfehlungen der extrakorporalen Photopherese (ECP). J Dtsch Dermatol Ges 2023; 21:1369-1381. [PMID: 37946642 DOI: 10.1111/ddg.15167_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/02/2023] [Indexed: 11/12/2023]
Abstract
ZusammenfassungDie extrakorporale Photopherese (ECP) hat in jüngster Zeit bei der Behandlung verschiedener Krankheiten an Bedeutung gewonnen. Ursprünglich als neue Therapie zur Behandlung von Patienten mit kutanem T‐Zell‐Lymphom vorgestellt, hat sich der Indikationsbereich für die ECP auf Hämatologie und Transplantationsimmunologie erweitert. Die ECP hat ihren festen Platz im Therapieplan bei kutanen T‐Zell‐Lymphomen, systemischer Sklerose, Graft‐versus‐Host‐Erkrankung, Organtransplantationen wie Herz und Lunge, teilweise als Erstlinientherapie und sehr häufig in Kombination mit verschiedenen systemischen immunsuppressiven Therapien. Das Verfahren besteht im Wesentlichen aus drei Schritten: Leukapherese, Photoaktivierung und Reinfusion. Im folgenden Artikel werden die noch nicht vollständig verstandenen Wirkmechanismen dargestellt, die fünf häufigsten Indikationen für die Behandlung mit ECP diskutiert und Therapieempfehlungen für die jeweilige Indikation gegeben.
Collapse
Affiliation(s)
- Ara Cho
- Universitätsklinik für Dermatologie, Medizinische Universität Wien, Wien, Österreich
| | - Verena Paulitschke
- Universitätsklinik für Dermatologie, Medizinische Universität Wien, Wien, Österreich
| | - Robert Knobler
- Universitätsklinik für Dermatologie, Medizinische Universität Wien, Wien, Österreich
| |
Collapse
|
15
|
Wang Q, Li CL, Wu L, Hu JY, Yu Q, Zhang SX, He PF. Distinct molecular subtypes of systemic sclerosis and gene signature with diagnostic capability. Front Immunol 2023; 14:1257802. [PMID: 37849750 PMCID: PMC10577296 DOI: 10.3389/fimmu.2023.1257802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023] Open
Abstract
Background As Systemic Sclerosis (SSc) is a connective tissue ailment that impacts various bodily systems. The study aims to clarify the molecular subtypes of SSc, with the ultimate objective of establishing a diagnostic model that can inform clinical treatment decisions. Methods Five microarray datasets of SSc were retrieved from the GEO database. To eliminate batch effects, the combat algorithm was applied. Immune cell infiltration was evaluated using the xCell algorithm. The ConsensusClusterPlus algorithm was utilized to identify SSc subtypes. Limma was used to determine differential expression genes (DEGs). GSEA was used to determine pathway enrichment. A support vector machine (SVM), Random Forest(RF), Boruta and LASSO algorithm have been used to select the feature gene. Diagnostic models were developed using SVM, RF, and Logistic Regression (LR). A ROC curve was used to evaluate the performance of the model. The compound-gene relationship was obtained from the Comparative Toxicogenomics Database (CTD). Results The identification of three immune subtypes in SSc samples was based on the expression profiles of immune cells. The utilization of 19 key intersectional DEGs among subtypes facilitated the classification of SSc patients into three robust subtypes (gene_ClusterA-C). Gene_ClusterA exhibited significant enrichment of B cells, while gene_ClusterC showed significant enrichment of monocytes. Moderate activation of various immune cells was observed in gene_ClusterB. We identified 8 feature genes. The SVM model demonstrating superior diagnostic performance. Furthermore, correlation analysis revealed a robust association between the feature genes and immune cells. Eight pertinent compounds, namely methotrexate, resveratrol, paclitaxel, trichloroethylene, formaldehyde, silicon dioxide, benzene, and tetrachloroethylene, were identified from the CTD. Conclusion The present study has effectively devised an innovative molecular subtyping methodology for patients with SSc and a diagnostic model based on machine learning to aid in clinical treatment. The study has identified potential molecular targets for therapy, thereby offering novel perspectives for the treatment and investigation of SSc.
Collapse
Affiliation(s)
- Qi Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, Taiyuan, China
| | - Chen-Long Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, Taiyuan, China
| | - Li Wu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Department of Anesthesiology , Shanxi Provincial People’s Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Jing-Yi Hu
- School of Management, Shanxi Medical University, Taiyuan, China
| | - Qi Yu
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, Taiyuan, China
- School of Management, Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Pei-Feng He
- Shanxi Key Laboratory of Big Data for Clinical Decision Research, Taiyuan, China
- School of Management, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
16
|
Kudsi M, Khalayli N, Tarcha R, Al-Darwish L. Tocilizumab in systemic sclerosis treatment: a case report. Ann Med Surg (Lond) 2023; 85:4586-4588. [PMID: 37663690 PMCID: PMC10473287 DOI: 10.1097/ms9.0000000000000969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/10/2023] [Indexed: 09/05/2023] Open
Abstract
Introduction In the United States, tocilizumab was approved for the treatment of scleroderma and scleroderma-related interstitial lung disease because it inhibited the decrease in forced expiratory volume, so scleroderma treatment is entering a new era. Case presentation A 44-year-old female patient with systemic scleroderma, diagnosed 6 years ago, presented with breathlessness over the last week. The modified Rodnan's score was 18. Tocilizumab 162 mg subcutaneously once every 2 weeks was prescribed. After 4 weeks, a decrease in Rodnan score was observed. Tocilizumab was stopped after 6 months without any side effects due to treatment. Discussion Treatment with tocilizumab may maintain lung function nearly unchanged. Its effect on perfecting skin fibrosis seems promising. Tocilizumab may be fairly safe to use. Conclusion Tocilizumab may be effective and fairly safe to use. Further exploration is anticipated to advance a new period of systemic sclerosis treatment.
Collapse
Affiliation(s)
| | | | | | - Lama Al-Darwish
- Faculty of Pharmacy, Al-Sham Private University (ASPU), Damascus, Syria
| |
Collapse
|
17
|
Ortega-Ferreira C, Soret P, Robin G, Speca S, Hubert S, Le Gall M, Desvaux E, Jendoubi M, Saint-Paul J, Chadli L, Chomel A, Berger S, Nony E, Neau B, Fould B, Licznar A, Levasseur F, Guerrier T, Elouej S, Courtade-Gaïani S, Provost N, Nguyen TQ, Verdier J, Launay D, De Ceuninck F. Antibody-mediated neutralization of galectin-3 as a strategy for the treatment of systemic sclerosis. Nat Commun 2023; 14:5291. [PMID: 37652913 PMCID: PMC10471779 DOI: 10.1038/s41467-023-41117-9] [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: 10/28/2022] [Accepted: 08/22/2023] [Indexed: 09/02/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune, inflammatory and fibrotic disease with limited treatment options. Developing new therapies is therefore crucial to address patient needs. To this end, we focused on galectin-3 (Gal-3), a lectin known to be associated with several pathological processes seen in SSc. Using RNA sequencing of whole-blood samples in a cross-sectional cohort of 249 patients with SSc, Gal-3 and its interactants defined a strong transcriptomic fingerprint associated with disease severity, pulmonary and cardiac malfunctions, neutrophilia and lymphopenia. We developed new Gal-3 neutralizing monoclonal antibodies (mAb), which were then evaluated in a mouse model of hypochlorous acid (HOCl)-induced SSc. We show that two of these antibodies, D11 and E07, reduced pathological skin thickening, lung and skin collagen deposition, pulmonary macrophage content, and plasma interleukin-5 and -6 levels. Moreover, E07 changed the transcriptional profiles of HOCl-treated mice, resulting in a gene expression pattern that resembled that of control mice. Similarly, pathological pathways engaged in patients with SSc were counteracted by E07 in mice. Collectively, these findings demonstrate the translational potential of Gal-3 blockade as a therapeutic option for SSc.
Collapse
Affiliation(s)
- Céline Ortega-Ferreira
- Servier R&D Center, Biomarker Assay Development, Translational Medicine, Gif-sur-Yvette, France
| | - Perrine Soret
- Servier R&D Center, Biomarker Biostatistics, Gif-sur-Yvette, France
| | | | - Silvia Speca
- U1286 INFINITE, Institute for Translational Research in Inflammation, Lille University, Gif-sur-Yvette, France
- Inserm, Lille, France
| | - Sandra Hubert
- Servier R&D Center, Neurosciences and Immuno-inflammation Therapeutic Area, Gif-sur-Yvette, France
| | | | - Emiko Desvaux
- Servier R&D Center, Neurosciences and Immuno-inflammation Therapeutic Area, Gif-sur-Yvette, France
| | - Manel Jendoubi
- U1286 INFINITE, Institute for Translational Research in Inflammation, Lille University, Gif-sur-Yvette, France
- Inserm, Lille, France
| | | | - Loubna Chadli
- Servier R&D Center, Clinical Biomarker Development, Translational Medicine, Gif-sur-Yvette, France
| | - Agnès Chomel
- Servier R&D Center, Protein Sciences, Gif-sur-Yvette, France
| | - Sylvie Berger
- Servier R&D Center, Structural Sciences, Gif-sur-Yvette, France
| | - Emmanuel Nony
- Servier R&D Center, Protein Sciences, Gif-sur-Yvette, France
| | - Béatrice Neau
- Servier R&D Center, Preclinical Biostatistics, Quantitative Pharmacology, Gif-sur-Yvette, France
| | - Benjamin Fould
- Servier R&D Center, Protein Sciences, Gif-sur-Yvette, France
| | - Anne Licznar
- Servier R&D Center, DMPK Department, Translational Medicine, Gif-sur-Yvette, France
| | - Franck Levasseur
- Servier R&D Center, DMPK Department, Translational Medicine, Gif-sur-Yvette, France
| | - Thomas Guerrier
- U1286 INFINITE, Institute for Translational Research in Inflammation, Lille University, Gif-sur-Yvette, France
- Inserm, Lille, France
| | - Sahar Elouej
- Servier R&D Center, Computational Medicine, Gif-sur-Yvette, France
| | | | - Nicolas Provost
- Servier R&D Center, Molecular Genomics, Gif-sur-Yvette, France
| | | | - Julien Verdier
- Servier R&D Center, Neurosciences and Immuno-inflammation Therapeutic Area, Gif-sur-Yvette, France
| | - David Launay
- U1286 INFINITE, Institute for Translational Research in Inflammation, Lille University, Gif-sur-Yvette, France
- Inserm, Lille, France
- Lille University Hospital, Department of Internal Medicine and Clinical Immunology, Reference Center for Rare Systemic Autoimmune Diseases, North and North-West France (CeRAINO), Lille, France
| | - Frédéric De Ceuninck
- Servier R&D Center, Neurosciences and Immuno-inflammation Therapeutic Area, Gif-sur-Yvette, France.
| |
Collapse
|
18
|
Moingeon P. Artificial intelligence-driven drug development against autoimmune diseases. Trends Pharmacol Sci 2023; 44:411-424. [PMID: 37268540 DOI: 10.1016/j.tips.2023.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 06/04/2023]
Abstract
Artificial intelligence (AI)-based predictive models are being used to foster a precision medicine approach to treat complex chronic diseases such as autoimmune and autoinflammatory disorders (AIIDs). In the past few years the first models of systemic lupus erythematosus (SLE), primary Sjögren syndrome (pSS), and rheumatoid arthritis (RA) have been produced by molecular profiling of patients using omic technologies and integrating the data with AI. These advances have confirmed a complex pathophysiology involving multiple proinflammatory pathways and also provide evidence for shared molecular dysregulation across different AIIDs. I discuss how models are used to stratify patients, assess causality in pathophysiology, design drug candidates in silico, and predict drug efficacy in virtual patients. By relating individual patient characteristics to the predicted properties of millions of drug candidates, these models can improve the management of AIIDs through more personalized treatments.
Collapse
Affiliation(s)
- Philippe Moingeon
- Research and Development, Servier Laboratories, 50 Rue Carnot, 92150 Suresnes, France; French Academy of Pharmacy, 4 Avenue de l'Observatoire, 75006 Paris, France.
| |
Collapse
|
19
|
Colic J, Campochiaro C, Hughes M, Matucci Cerinic M, Dagna L. Investigational drugs for the treatment of scleroderma: what's new? Expert Opin Investig Drugs 2023; 32:601-614. [PMID: 37526079 DOI: 10.1080/13543784.2023.2242762] [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: 05/17/2023] [Accepted: 07/26/2023] [Indexed: 08/02/2023]
Abstract
INTRODUCTION Systemic sclerosis (SSc) is an orphan, chronic, autoimmune, fibrotic disease with unknown etiology characterized by progressive fibrosis of the skin and internal organs. SSc has the highest mortality, the deadliest among the connective tissue diseases, despite the introduction of new treatment options in the past decades. AREAS COVERED The aim of the current systematic review was to investigate new targeted therapy and their impact on disease progression, mainly focusing on phase I and II clinical trials within the past three years. EXPERT OPINION Despite recent groundbreaking advancements in understanding SSc pathophysiology, early diagnosis and early introduction of effective targeted treatments within the optimal window of opportunity to prevent irreversible disease damage still represents a significant clinical challenge. Ongoing significant research for new molecular and epigenetics pathways is of fundamental importance to offer new perspectives on disease phenotype and for the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Jelena Colic
- Department of Rheumatology, Institute of Rheumatology, Belgrade, Serbia
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| | - Michael Hughes
- Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, England
| | - Marco Matucci Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milano, Italy
- Division of Rheumatology, Department of Experimental and Clinical Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC) and Denothe Centre, University of Florence, Florence, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milano, Italy
- Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
20
|
Madsen SF, Sand JMB, Juhl P, Karsdal M, Thudium CS, Siebuhr AS, Bay-Jensen AC. Fibroblasts are not just fibroblasts: clear differences between dermal and pulmonary fibroblasts' response to fibrotic growth factors. Sci Rep 2023; 13:9411. [PMID: 37296166 PMCID: PMC10256773 DOI: 10.1038/s41598-023-36416-6] [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: 02/20/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
Systemic Sclerosis (SSc) hallmark is skin fibrosis, but up to 80% of the patients have fibrotic involvement in the pulmonary system. Antifibrotic drugs which have failed in a general SSc population have now been approved in patients with SSc-associated interstitial lung disease (ILD). This indicates that the fibrotic progression and regulation of fibroblasts likely depend on local factors specific to the tissue type. This study investigated the difference between dermal and pulmonary fibroblasts in a fibrotic setting, mimicking the extracellular matrix. Primary healthy fibroblasts were grown in a crowded environment and stimulated with TGF-β1 and PDGF-AB. The viability, morphology, migration capacity, extracellular matrix formation, and gene expression were assessed: TGF-β1 only increased the viability in the dermal fibroblasts. PDGF-AB increased the migration capacity of dermal fibroblasts while the pulmonary fibroblasts fully migrated. The morphology of the fibroblasts was different without stimulation. TGF-β1 increased the formation of type III collagen in pulmonary fibroblasts, while PDGF-AB increased it in dermal fibroblasts. The gene expression trend of type VI collagen was the opposite after PDGF-AB stimulation. The fibroblasts exhibit different response profiles to TGF-β1 and PDGF-AB; this suggests that drivers of fibrosis are tissue-dependent, which needs to be considered in drug development.
Collapse
Affiliation(s)
- Sofie Falkenløve Madsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Immunoscience, Nordic Bioscience, Herlev, Denmark.
| | | | | | | | | | | | | |
Collapse
|
21
|
Richter P, Cardoneanu A, Dima N, Bratoiu I, Rezus C, Burlui AM, Costin D, Macovei LA, Rezus E. Interstitial Lung Disease in Systemic Lupus Erythematosus and Systemic Sclerosis: How Can We Manage the Challenge? Int J Mol Sci 2023; 24:9388. [PMID: 37298342 PMCID: PMC10253395 DOI: 10.3390/ijms24119388] [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: 05/03/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Interstitial lung disease (ILD) is a severe and frequent manifestation of connective tissue diseases (CTD). Due to its debilitating potential, it requires serious evaluation and treatment. The prevalence of ILD in systemic lupus erythematosus (SLE) is still controversial. Therefore, in order to establish the diagnosis of ILD, an overlap syndrome must be excluded. Increasing the identification of SLE-associated ILD cases should become a target. To treat this complication, various therapies are now being proposed. To date, no placebo-controlled studies were conducted. Regarding another CTD, systemic sclerosis (SSc), SSc-associated ILD is considered one of the leading causes of mortality. The incidence of ILD varies among disease subtypes, being influenced by diagnostic method, but also by disease duration. Due to the high prevalence of this complication, all SSc patients should be investigated for ILD at the time of SSc diagnosis and during the course of the disease. Fortunately, progress was made in terms of treatment. Nintedanib, a tyrosine kinases inhibitor, showed promising results. It appeared to decrease the rate of progression of ILD compared to placebo. This review aimed to provide up-to-date findings related to SLE-associated ILD and SSc-associated ILD, in order to raise awareness of their diagnosis and management.
Collapse
Affiliation(s)
- Patricia Richter
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Anca Cardoneanu
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- “Sf. Spiridon” Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- “Sf. Spiridon” Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Alexandra Maria Burlui
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Damiana Costin
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Luana Andreea Macovei
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology, “Grigore T Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
22
|
Zhuang M, Li CC, Chen SY, Tu XH, Liu L, Chen XL, Xu CW, Wang J. Development and validation of a Systemic Sclerosis Health Literacy Scale. Front Public Health 2023; 11:1038019. [PMID: 36908416 PMCID: PMC9996225 DOI: 10.3389/fpubh.2023.1038019] [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: 09/06/2022] [Accepted: 01/31/2023] [Indexed: 02/25/2023] Open
Abstract
Background and aim Health literacy levels are strongly associated with clinical outcomes and quality of life in patients with chronic diseases, and patients with limited health literacy often require more medical care and achieve poorer clinical outcomes. Among the large number of studies on health literacy, few studies have focused on the health literacy of people with systemic sclerosis (SSc), and there is no specific tool to measure health literacy in this group. Therefore, this study plans to develop a health literacy scale for patients with SSc. Methods This study included 428 SSc patients from the outpatient and inpatient departments of the Department of Rheumatology and Immunology, the first affiliated Hospital of Anhui Medical University and the first affiliated Hospital of University of Science and Technology of China. The formulation of the scale was completed by forming the concept of health literacy of SSc patients, establishing the item pool, screening items, and evaluating reliability and validity. Classical measurement theory was used to screen items, factor analysis was used to explore the construct validity of the scale, and Cronbach's alpha coefficient was used to assess the internal consistency. Results Our study population was predominantly middle-aged women, with a male to female ratio of 1:5.7 and a mean age of 51.57 ± 10.99. A SSc Health Literacy scale with 6 dimensions and 30 items was developed. The six dimensions are clinic ability, judgment/evaluation information ability, access to information ability, social support, treatment compliance and application information ability. The Cronbach's alpha coefficient of the scale is 0.960, retest reliability is 0.898, split-half reliability is 0.953, content validity is 0.983, which has good reliability and validity. Conclusion The Systemic Sclerosis Health Literacy Scale may become a valid tool to evaluate the health literacy level of patients with SSc.
Collapse
Affiliation(s)
- Meng Zhuang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Medical Data Processing Center, Anhui Medical University, Hefei, China
| | - Cheng-Cheng Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Medical Data Processing Center, Anhui Medical University, Hefei, China
| | - Shan-Yu Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin-Hua Tu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Medical Data Processing Center, Anhui Medical University, Hefei, China
| | - Lian Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Medical Data Processing Center, Anhui Medical University, Hefei, China
| | - Xi-Lai Chen
- Department of Statistics, College of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu, China
| | - Cheng-Wei Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Medical Data Processing Center, Anhui Medical University, Hefei, China
| | - Jing Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Medical Data Processing Center, Anhui Medical University, Hefei, China
| |
Collapse
|
23
|
Bissenova S, Ellis D, Mathieu C, Gysemans C. Neutrophils in autoimmunity: when the hero becomes the villain. Clin Exp Immunol 2022; 210:128-140. [PMID: 36208466 PMCID: PMC9750832 DOI: 10.1093/cei/uxac093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/20/2022] [Accepted: 10/06/2022] [Indexed: 01/25/2023] Open
Abstract
Neutrophils were long considered to be a short-lived homogenous cell population, limited to their role as first responders in anti-bacterial and -fungal immunity. While it is true that neutrophils are first to infiltrate the site of infection to eliminate pathogens, growing evidence suggests their functions could extend beyond those of basic innate immune cells. Along with their well-established role in pathogen elimination, utilizing effector functions such as phagocytosis, degranulation, and the deployment of neutrophil extracellular traps (NETs), neutrophils have recently been shown to possess antigen-presenting capabilities. Moreover, the identification of different subtypes of neutrophils points to a multifactorial heterogeneous cell population with great plasticity in which some subsets have enhanced pro-inflammatory characteristics, while others seem to behave as immunosuppressors. Interestingly, the aberrant presence of activated neutrophils with a pro-inflammatory profile in several systemic and organ-specific autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc), multiple sclerosis (MS), and type 1 diabetes (T1D) could potentially be exploited in novel therapeutic strategies. The full extent of the involvement of neutrophils, and more specifically that of their various subtypes, in the pathophysiology of autoimmune diseases is yet to be elucidated.
Collapse
Affiliation(s)
- Samal Bissenova
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Darcy Ellis
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Conny Gysemans
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Li JX. Secular Trends in Systemic Sclerosis Mortality in the United States from 1981 to 2020. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15088. [PMID: 36429809 PMCID: PMC9690027 DOI: 10.3390/ijerph192215088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Systemic sclerosis (SSc) has the highest mortality rate among autoimmune disorders. Individuals with SSc frequently die from complications or infections related to SSc. Nonetheless, the sex-age-period interaction of SSc is complex and remains unclear. The study aims to analyze the secular trend of SSc mortality based on data regarding underlying cause of death (UCD) and multiple causes of death (MCD) and clarify the sex-age interaction with time. METHODS The multiple-cause mortality statistics provided by the National Center for Health Statistics were used to identify all deaths in the United States from 1981 to 2020 in which SSc was indicated anywhere on the death certificates. The age-standardized mortality rate (ASMR) was determined for both sexes, as well as the variations in these rates. Joinpoint regression analysis was utilized to determine the annual percentage change (APC) of ASMR. RESULTS A total of 44,672 and 66,259 individuals who died between 1981 and 2020 were identified based on the UCD and MCD data, respectively. According to the UCD data, SSc-related AMSR (SSc-ASMR) of the male and female decedents, respectively, declined from 5.01 and 1.94 in 1981-1990 to 4.77 and 1.32 in 2011-2020, respectively (mortality rate ratio 0.95, 95% confidence interval 0.92-0.98). From 1986 to 1999, the APC of SSc-ASMR in female decedents decreased except for those aged 45-64 years (APC 2.1%, p = 0.002). For MCD analysis, in trend 1, only APC of SSc-ASMR in male decedents aged 45-64 years decreased. The SSc-ASMR of both male and female decedents fell on trend 2 arm. In 2011-2020, the ratio of UCD to MCD increased across all age groups for both sexes compared to 1981-1990. Overall, compared to the male decedents, the SSc-ASMR in female decedents increased significantly before 1999, peaked in 1999, followed by continuous decrease until 2020 according to UCD and MCD statistics. CONCLUSIONS Over the past four decades, the SSc deaths based on the MCD data were 1.48 times more than the UCD data, and the proportion of UCD over MCD increased over time. The SSc-ASMRs in all the sex-age groups significantly decreased over the past two decades. Notably, the mortality rate ratio of women to men with SSc increased in the past four decades.
Collapse
Affiliation(s)
- Jing-Xing Li
- Department of General Medicine, China Medical University Hospital, No. 2, Yude Rd., North District, Taichung 404327, Taiwan; ; Tel.: +886-905-770-531
- School of Medicine, China Medical University, Taichung 404327, Taiwan
- Graduate Institute of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei 100225, Taiwan
| |
Collapse
|