1
|
Mota PC, Soares ML, Ferreira AC, Santos RF, Rufo JC, Vasconcelos D, Carvalho A, Guimarães S, Vasques-Nóvoa F, Cardoso C, Melo N, Alexandre AT, Coelho D, Novais-Bastos H, Morais A. Polymorphisms and haplotypes of TOLLIP and MUC5B are associated with susceptibility and survival in patients with fibrotic hypersensitivity pneumonitis. Pulmonology 2025; 31:2416788. [PMID: 38309995 DOI: 10.1016/j.pulmoe.2024.01.002] [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: 11/15/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 02/05/2024] Open
Abstract
INTRODUCTION AND OBJECTIVES Hypersensitivity pneumonitis (HP) is an interstitial lung disease with diverse clinical features that can present a fibrotic phenotype similar to idiopathic pulmonary fibrosis (IPF) in genetically predisposed individuals. While several single nucleotide polymorphisms (SNPs) have been associated with IPF, the genetic factors contributing to fibrotic HP (fHP) remain poorly understood. This study investigated the association of MUC5B and TOLLIP variants with susceptibility, clinical presentation and survival in Portuguese patients with fHP. MATERIAL AND METHODS A case-control study was undertaken with 97 fHP patients and 112 controls. Six SNPs residing in the MUC5B and TOLLIP genes and their haplotypes were analyzed. Associations with risk, survival, and clinical, radiographic, and pathological features of fHP were probed through comparisons among patients and controls. RESULTS MUC5B rs35705950 and three neighboring TOLLIP variants (rs3750920, rs111521887, and rs5743894) were associated with increased susceptibility to fHP. Minor allele frequencies were greater among fHP patients than in controls (40.7% vs 12.1%, P<0.0001; 52.6% vs 40.2%, P = 0.011; 22.7% vs 13.4%, P = 0.013; and 23.2% vs 12.9%, P = 0.006, respectively). Haplotypes formed by these variants were also linked to fHP susceptibility. Moreover, carriers of a specific haplotype (G-T-G-C) had a significant decrease in survival (adjusted hazard ratio 6.92, 95% CI 1.73-27.64, P = 0.006). Additional associations were found between TOLLIP rs111521887 and rs5743894 variants and decreased lung function at baseline, and the MUC5B SNP and radiographic features, further highlighting the influence of genetic factors in fHP. CONCLUSION These findings suggest that TOLLIP and MUC5B variants and haplotypes may serve as valuable tools for risk assessment and prognosis in fibrotic hypersensitivity pneumonitis, potentially contributing to its patient stratification, and offer insights into the genetic factors influencing the clinical course of the condition.
Collapse
Affiliation(s)
- P C Mota
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - M L Soares
- Laboratório de Apoio à Investigação em Medicina Molecular (LAIMM), Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Portugal
- LAIMM, Núcleo de Recursos Laboratoriais, Unidade de Gestão de Conhecimento, Departamento de Recursos Comuns, Faculdade de Medicina da Universidade do Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A C Ferreira
- Laboratório de Apoio à Investigação em Medicina Molecular (LAIMM), Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - R F Santos
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Escola Superior de Saúde - Instituto Politécnico do Porto, Portugal
| | - J C Rufo
- Indoor Air Quality and Respiratory Health Lab, Laboratory for Integrative and Translational Research in Population Health (ITR), Porto, Portugal
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Center for Translational Health and Medical Biotechnology Research (T.Bio), Escola Superior de Saúde, Instituto Politécnico do Porto, Porto, Portugal
| | - D Vasconcelos
- Laboratório de Apoio à Investigação em Medicina Molecular (LAIMM), Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A Carvalho
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Departamento de Radiologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
| | - S Guimarães
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Departamento de Anatomia Patológica, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
| | - F Vasques-Nóvoa
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Departamento de Medicina Interna, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- UnIC@RISE, Department of Surgery and Physiology, Portugal
| | - C Cardoso
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - N Melo
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
| | - A T Alexandre
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
| | - D Coelho
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - H Novais-Bastos
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - A Morais
- Departamento de Pneumologia, Centro Hospitalar Universitário de São João, EPE, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
2
|
Hindré R, Uzunhan Y. [Antifibrotic therapies: Where do we stand 10years later?]. Rev Mal Respir 2025:S0761-8425(25)00168-8. [PMID: 40268574 DOI: 10.1016/j.rmr.2025.04.002] [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: 01/26/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
INTRODUCTION Fibrosing interstitial lung diseases (ILD) are severe respiratory conditions that can lead to respiratory failure and death. Over the past decade, antifibrotic therapies have represented a significant therapeutic advancement and are now widely used. STATE OF THE ART Pirfenidone and nintedanib have been approved for the treatment of idiopathic pulmonary fibrosis (IPF), while only nintedanib has been approved for systemic sclerosis-related ILD and progressive pulmonary fibrosis (PPF). Both drugs help to reduce the decline in forced vital capacity (FVC) characterizing these three indications and to decrease mortality, acute exacerbations, and quality of life impairment in patients with IPF and PPF. PERSPECTIVES Tolerance to these treatments remains a major challenge, prompting evaluation of alternative administration routes, such as inhalation. Numerous ongoing clinical trials and encouraging results from phase 3 studies are expected to lead to the approval of new antifibrotic molecules. CONCLUSIONS Antifibrotic therapies have proven to be crucial in the management of IPF and PPF. Prescription should be a shared decision with the patient and may be considered at an early stage, even in elderly individuals, provided that dedicated support is avaialble.
Collapse
Affiliation(s)
- R Hindré
- Service de pneumologie, Centre de référence constitutif des maladies pulmonaires rares, AP-HP, hôpital Avicenne, 93000 Bobigny, France; Inserm UMR 1272, Université Sorbonne Paris Nord, Bobigny, France.
| | - Y Uzunhan
- Service de pneumologie, Centre de référence constitutif des maladies pulmonaires rares, AP-HP, hôpital Avicenne, 93000 Bobigny, France; Inserm UMR 1272, Université Sorbonne Paris Nord, Bobigny, France
| |
Collapse
|
3
|
Meadows I, Mvungi H, Salim K, Kaswaga O, Mbelele P, Liyoyo A, Semvua H, Ngoma A, Heysell SK, Mpagama SG. N-Acetylcysteine to Reduce Kidney and Liver Injury Associated with Drug-Resistant Tuberculosis Treatment. Pharmaceutics 2025; 17:516. [PMID: 40284511 PMCID: PMC12030172 DOI: 10.3390/pharmaceutics17040516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Background: New drug classes and regimens have shortened the treatment duration for drug-resistant tuberculosis, but adverse events (AEs) and organ toxicity remain unacceptably common. N-acetylcysteine (NAC) has demonstrated potential in reducing kidney and liver toxicity in other clinical settings, but efficacy in drug-resistant tuberculosis treatment has not been rigorously evaluated. Method: A randomized controlled trial was conducted at Kibong'oto Infectious Diseases Hospital in Tanzania to assess the efficacy of NAC in reducing AEs in patients undergoing rifampin-resistant pulmonary tuberculosis treatment. Participants received an all-oral standardized rifampin-resistant regimen alone, with NAC 900 mg daily, or NAC 900 mg twice daily for 6 months. AEs, severe AEs, and renal and liver toxicity were monitored monthly and classified according to the Risk, Injury, Failure, Loss, and End-stage kidney disease criteria and National Cancer Institute Common Terminology Criteria for Adverse Events. Incident ratios and Kaplan-Meier curves were employed to compare group event occurrences. Results: A total of 66 patients (mean age 47 ± 12 years; 80% male) were randomized into three groups of 22. One hundred and fifty-eight AEs were recorded: 52 (33%) in the standard treatment group, 55 (35%) in the NAC 900 mg daily group, and 51 (32%) in the NAC 900 mg twice-daily group (p > 0.99). Severe AEs were observed in four patients in the standard group, two in the NAC 900 mg daily group, and three in the NAC 900 mg twice-daily group. Renal toxicity was more prevalent in the standard treatment group compared to those that received NAC (45% vs. 23%; p = 0.058), with a shorter onset of time to toxicity (χ2 = 3.199; p = 0.074). Liver injury events were rare across all groups. Conclusion: Among Tanzanian adults receiving rifampin-resistant tuberculosis treatment, NAC did not significantly reduce overall AEs but demonstrated important trends in reducing renal toxicity.
Collapse
Affiliation(s)
- Idu Meadows
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22903, USA; (I.M.); (S.K.H.)
| | - Happiness Mvungi
- Kibong’oto Infectious Disease Hospital, Siha 25102, Tanzania; (H.M.); (K.S.); (O.K.); (P.M.)
- Kilimanjaro Christian Medical College, Moshi 25212, Tanzania;
| | - Kassim Salim
- Kibong’oto Infectious Disease Hospital, Siha 25102, Tanzania; (H.M.); (K.S.); (O.K.); (P.M.)
| | - Oscar Kaswaga
- Kibong’oto Infectious Disease Hospital, Siha 25102, Tanzania; (H.M.); (K.S.); (O.K.); (P.M.)
| | - Peter Mbelele
- Kibong’oto Infectious Disease Hospital, Siha 25102, Tanzania; (H.M.); (K.S.); (O.K.); (P.M.)
| | - Alphonce Liyoyo
- Kibong’oto Infectious Disease Hospital, Siha 25102, Tanzania; (H.M.); (K.S.); (O.K.); (P.M.)
| | - Hadija Semvua
- Kilimanjaro Christian Medical College, Moshi 25212, Tanzania;
| | - Athumani Ngoma
- Kibong’oto Infectious Disease Hospital, Siha 25102, Tanzania; (H.M.); (K.S.); (O.K.); (P.M.)
| | - Scott K. Heysell
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22903, USA; (I.M.); (S.K.H.)
| | - Stellah G. Mpagama
- Kibong’oto Infectious Disease Hospital, Siha 25102, Tanzania; (H.M.); (K.S.); (O.K.); (P.M.)
- Kilimanjaro Christian Medical College, Moshi 25212, Tanzania;
| |
Collapse
|
4
|
Vasarmidi E, Worrell JC, Mahmutovic Persson I, Yaqub N, Miądlikowska E, Barnig C, Boots A, Reynaert NL, Cuevas Ocaña S. Insights into interstitial lung disease pathogenesis. Breathe (Sheff) 2025; 21:240261. [PMID: 40365095 PMCID: PMC12070197 DOI: 10.1183/20734735.0261-2024] [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: 12/07/2024] [Accepted: 03/11/2025] [Indexed: 05/15/2025] Open
Abstract
This review summarises some of the key features of interstitial lung diseases (ILDs) from a translational science point of view and brings insights into potential therapeutic options. Genetic predisposition and environmental factors like smoking, pollution and infections significantly impact the onset, progression and treatment response in ILDs, highlighting the need for personalised management. Fibroblasts are central to ILD pathology, influencing the tissue microenvironment, immune cell interactions and extracellular matrix (ECM) production, making them critical therapeutic targets. Monocyte-derived M2 macrophages drive fibrosis in idiopathic pulmonary fibrosis by secreting cytokines and remodelling the ECM. Understanding macrophage subtypes and their dynamics offers new therapeutic possibilities. Chronic type 2 immunity contributes to fibrosis, emphasising the need to enhance protective markers in order to even out the balance shift of pathological immune responses in ILD treatments. Serum biomarkers like Krebs von den Lungen-6 (KL-6), surfactant protein (SFTP) D, matrix metalloproteinase-7 (MMP-7), and C-C motif chemokine ligand (CCL)-18 are valuable for diagnosing and predicting ILD progression, although more research is needed for clinical application. Animal models, especially bleomycin-based models, offer insights into ILD pathology, but challenges like lung hyperinflation highlight the need for careful model selection and translational research to bridge preclinical and clinical findings.
Collapse
Affiliation(s)
- Eirini Vasarmidi
- Department of Respiratory Medicine, Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, Heraklion, Greece
- These authors contributed equally
| | - Julie C. Worrell
- Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
- These authors contributed equally
| | - Irma Mahmutovic Persson
- Respiratory Immunopharmacology, Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
- Lund University BioImaging Centre (LBIC), Faculty of Medicine, Lund University, Lund, Sweden
- These authors contributed equally
| | - Naheem Yaqub
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Ewa Miądlikowska
- Department of Pneumology, Medical University of Lodz, Lodz, Poland
| | - Cindy Barnig
- Université de Franche-Comté, CHU Besançon, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Agnes Boots
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Niki L. Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Sara Cuevas Ocaña
- Biodiscovery Institute, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
5
|
Karampitsakos T, Tourki B, Herazo-Maya JD. The Dawn of Precision Medicine in Fibrotic Interstitial Lung Disease. Chest 2025; 167:1120-1132. [PMID: 39521375 PMCID: PMC12001815 DOI: 10.1016/j.chest.2024.10.042] [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: 04/08/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
TOPIC IMPORTANCE Interstitial lung diseases (ILDs) represent a broad group of heterogeneous parenchymal lung diseases. Some ILDs progress, causing architectural distortion and pulmonary fibrosis, and thus are called fibrotic ILDs. Recent studies have shown a beneficial effect of antifibrotic therapy in fibrotic ILDs other than idiopathic pulmonary fibrosis (IPF) that manifest progressive pulmonary fibrosis (PPF). However, it remains challenging to predict which patients with fibrotic ILDs will demonstrate PPF. Precision medicine approaches could identify patients at risk for progression and guide treatment in patients with IPF or PPF. REVIEW FINDINGS Multiple biomarkers able to highlight disease susceptibility risk, to provide an accurate diagnosis, and to prognosticate or assess treatment response have been identified. Advances in precision medicine led to the identification of endotypes that could discriminate patients with different fibrotic ILDs or patients with different disease courses. Importantly, recent studies have shown that particular compounds were efficacious only in particular endotypes. The aforementioned findings are promising. However, implementation in clinical practice remains an unmet need. SUMMARY Substantial progress has been observed in the context of precision medicine approaches in fibrotic ILDs in recent years. Nonetheless, infrastructure, financial, regulatory, and ethical challenges remain before precision medicine can be implemented in clinical practice. Overcoming such barriers and moving from a one-size-fits-all approach to patient-centered care could improve patient quality of life and survival substantially.
Collapse
Affiliation(s)
- Theodoros Karampitsakos
- Division of Pulmonary, Critical Care and Sleep Medicine, Ubben Center for Pulmonary Fibrosis Research, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Bochra Tourki
- Division of Pulmonary, Critical Care and Sleep Medicine, Ubben Center for Pulmonary Fibrosis Research, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Jose D Herazo-Maya
- Division of Pulmonary, Critical Care and Sleep Medicine, Ubben Center for Pulmonary Fibrosis Research, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL.
| |
Collapse
|
6
|
Liu H, Shen J, He C. Advances in idiopathic pulmonary fibrosis diagnosis and treatment. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2025; 3:12-21. [PMID: 40226606 PMCID: PMC11993042 DOI: 10.1016/j.pccm.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Indexed: 04/15/2025]
Abstract
Significant advances have been made in diagnosing and treating idiopathic pulmonary fibrosis (IPF) in the last decade. The incidence and prevalence of IPF are increasing, and morbidity and mortality remain high despite the two Food and Drug Administration (FDA)-approved medications, pirfenidone and nintedanib. Hence, there is an urgent need to develop new diagnostic tools and effective therapeutics to improve early, accurate diagnosis of IPF and halt or reverse the progression of fibrosis with a better safety profile. New diagnostic tools such as transbronchial cryobiopsy and genomic classifier require less tissue and generally have good safety profiles, and they have been increasingly utilized in clinical practice. Advances in artificial intelligence-aided diagnostic software are promising, but challenges remain. Both pirfenidone and nintedanib focus on growth factor-activated pathways to inhibit fibroblast activation. Novel therapies targeting different pathways and cell types (immune and epithelial cells) are being investigated. Biomarker-based personalized medicine approaches are also in clinical trials. This review aims to summarize recent diagnostic and therapeutic development in IPF.
Collapse
Affiliation(s)
- Hongli Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jiaxi Shen
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chao He
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX 77024, USA
| |
Collapse
|
7
|
Valand A, Rajasekar P, Wain LV, Clifford RL. Interplay between genetics and epigenetics in lung fibrosis. Int J Biochem Cell Biol 2025; 180:106739. [PMID: 39848439 DOI: 10.1016/j.biocel.2025.106739] [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: 07/25/2024] [Revised: 12/15/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
Lung fibrosis, including idiopathic pulmonary fibrosis (IPF), is a complex and devastating disease characterised by the progressive scarring of lung tissue leading to compromised respiratory function. Aberrantly activated fibroblasts deposit extracellular matrix components into the surrounding lung tissue, impairing lung function and capacity for gas exchange. Both genetic and epigenetic factors have been found to play a role in the pathogenesis of lung fibrosis, with emerging evidence highlighting the interplay between these two regulatory mechanisms. This review provides an overview of the current understanding of the interplay between genetics and epigenetics in lung fibrosis. We discuss the genetic variants associated with susceptibility to lung fibrosis and explore how epigenetic modifications such as DNA methylation, histone modifications, and non-coding RNA expression contribute to disease. Insights from genome-wide association studies (GWAS) and epigenome-wide association studies (EWAS) are integrated to explore the molecular mechanisms underlying lung fibrosis pathogenesis. We also discuss the potential clinical implications of genetics and epigenetics in lung fibrosis, including the development of novel therapeutic targets. Overall, this review highlights the importance of considering both genetic and epigenetic factors in the understanding and management of lung fibrosis.
Collapse
Affiliation(s)
- Anita Valand
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, University of Nottingham, UK; Nottingham NIHR Biomedical Research Centre, Nottingham, UK; Biodiscovery Institute, University Park, University of Nottingham, UK
| | - Poojitha Rajasekar
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, University of Nottingham, UK; Nottingham NIHR Biomedical Research Centre, Nottingham, UK; Biodiscovery Institute, University Park, University of Nottingham, UK
| | - Louise V Wain
- Department of Population Health Sciences, University of Leicester, Leicester, UK; NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Rachel L Clifford
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, University of Nottingham, UK; Nottingham NIHR Biomedical Research Centre, Nottingham, UK; Biodiscovery Institute, University Park, University of Nottingham, UK.
| |
Collapse
|
8
|
Lewandowska KB, Lechowicz U, Roży A, Falis M, Błasińska K, Jakubowska L, Franczuk M, Żołnowska B, Gryczka-Wróbel J, Radwan-Rohrenschef P, Lewandowska A, Witczak-Jankowska O, Sobiecka M, Szturmowicz M, Tomkowski WZ. MUC5B Polymorphism in Patients with Idiopathic Pulmonary Fibrosis-Does It Really Matter? Int J Mol Sci 2025; 26:2218. [PMID: 40076835 PMCID: PMC11900561 DOI: 10.3390/ijms26052218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a rare disorder concerning elderly people, predominantly men, active or former smokers, with a progressive nature and leading to premature mortality. The cause of the disease is unknown. However, there are some risk factors, among which genetic predisposition plays a role. The aim of our single-centered observational study was to assess the correlation between single nucleotide polymorphism (SNP) of the MUC5B gene (rs35705950) and the disease course, antifibrotic treatment effect, and survival in patients with IPF. A total of 93 patients entered the study, of whom 88 were treated with either nintedanib or pirfenidone. The GG genotype was found in 28 (30.1%) subjects, while the GT or TT genotypes were found in the remaining 65 (63.4%) and 6 (6.5%) patients, respectively. The T allele minor allele frequency (MAF) accounted for 38.2% of the whole group. Patients with different genotypes did not differ significantly regarding age, sex, pulmonary function tests' results, response to the antifibrotic treatment, or survival. However, we found a survival advantage in female patients and patients with higher pre-treatment TL,co. Treatment with antifibrotics significantly decreased the magnitude of FVC and TL,co decline compared to the time before treatment initiation, regardless of MUC5B status. In conclusion, we found high prevalence of T allele of MUC5B gene in patients with IPF; however, it showed no influence on disease trajectory, survival, or antifibrotic treatment effect in the presented cohort.
Collapse
Affiliation(s)
- Katarzyna B. Lewandowska
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (U.L.); (A.R.)
| | - Adriana Roży
- Department of Genetics and Clinical Immunology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (U.L.); (A.R.)
| | - Maria Falis
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Katarzyna Błasińska
- Department of Radiology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (K.B.); (L.J.)
| | - Lilia Jakubowska
- Department of Radiology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (K.B.); (L.J.)
| | - Monika Franczuk
- Department of Respiratory Physiopathology, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland;
| | - Beata Żołnowska
- Outpatient Clinic, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland;
| | - Justyna Gryczka-Wróbel
- 2nd Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland;
| | - Piotr Radwan-Rohrenschef
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Anna Lewandowska
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Olimpia Witczak-Jankowska
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Małgorzata Sobiecka
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Monika Szturmowicz
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| | - Witold Z. Tomkowski
- First Department of Lung Diseases, National Research Institute of Tuberculosis and Lung Diseases, Płocka 26, 01-138 Warsaw, Poland; (M.F.); (P.R.-R.); (A.L.); (O.W.-J.); (M.S.); (M.S.); (W.Z.T.)
| |
Collapse
|
9
|
Liu H, Cui H, Liu G. The Intersection between Immune System and Idiopathic Pulmonary Fibrosis-A Concise Review. FIBROSIS (HONG KONG, CHINA) 2025; 3:10004. [PMID: 40124525 PMCID: PMC11928166 DOI: 10.70322/fibrosis.2025.10004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is marked by progressive alveolar destruction, impaired tissue regeneration, and relentless fibrogenesis, culminating in respiratory failure and death. A diverse array of resident and non-resident cells within the lung contribute to disease pathogenesis. Notably, immune cells, both resident and recruited, respond to cues from sites of lung injury by undergoing phenotypic transitions and producing a wide range of mediators that influence, initiate, or dictate the function, or dysfunction, of key effector cells in IPF pathology, such as alveolar epithelial cells, lung fibroblasts, and capillary endothelial cells. The role of the immune system in IPF has undergone an interesting evolution, oscillating from initial enthusiasm to skepticism, and now to a renewed focus. This shift reflects both the past failures of immune-targeting therapies for IPF and the unprecedented insights into immune cell heterogeneity provided by emerging technologies. In this article, we review the historical evolution of perspectives on the immune system's role in IPF pathogenesis and examine the lessons learned from previous therapeutic failures targeting immune responses. We discuss the major immune cell types implicated in IPF progression, highlighting their phenotypic transitions and mechanisms of action. Finally, we identify key knowledge gaps and propose future directions for research on the immune system in IPF.
Collapse
Affiliation(s)
- Hongli Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
10
|
Cerri S, Manzini E, Nori O, Pacchetti L, Rossi L, Turchiano MG, Samarelli AV, Raineri G, Andrisani D, Gozzi F, Beghè B, Clini E, Tonelli R. Genetic Risk Factors in Idiopathic and Non-Idiopathic Interstitial Lung Disease: Similarities and Differences. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1967. [PMID: 39768847 PMCID: PMC11677115 DOI: 10.3390/medicina60121967] [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: 10/30/2024] [Revised: 11/16/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
Recent advances in genetics and epigenetics have provided critical insights into the pathogenesis of both idiopathic and non-idiopathic interstitial lung diseases (ILDs). Mutations in telomere-related genes and surfactant proteins have been linked to familial pulmonary fibrosis, while variants in MUC5B and TOLLIP increase the risk of ILD, including idiopathic pulmonary fibrosis and rheumatoid arthritis-associated ILD. Epigenetic mechanisms, such as DNA methylation, histone modifications, and non-coding RNAs such as miR-21 and miR-29, regulate fibrotic pathways, influencing disease onset and progression. Although no standardized genetic panel for ILD exists, understanding the interplay of genetic mutations and epigenetic alterations could aid in the development of personalized therapeutic approaches. This review highlights the genetic and epigenetic factors driving ILD, emphasizing their potential for refining diagnosis and treatment.
Collapse
Affiliation(s)
- Stefania Cerri
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Elisa Manzini
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
- Respiratory Disease Unit, Hospital of Sassuolo, 41049 Sassuolo, Italy
| | - Ottavia Nori
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
- U.O. Pneumologia, Presidio Ospedaliero di Arco, APSS Provincia Autonoma di Trento, 38062 Trento, Italy
| | - Lucia Pacchetti
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
- Division of Pneumology, MultiMedica IRCCS, 20099 Milan, Italy
| | - Laura Rossi
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
- Respiratory Disease Unit, Arcispedale Santa Maria Nuova, 42123 Reggio Emilia, Italy
| | - Maria Giulia Turchiano
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Post Doctoral School in Respiratory Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy; (E.M.); (O.N.); (L.P.); (L.R.)
| | - Anna Valeria Samarelli
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Giulia Raineri
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Dario Andrisani
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Filippo Gozzi
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Bianca Beghè
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
| | - Enrico Clini
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| | - Roberto Tonelli
- Respiratory Disease Unit, University Hospital of Modena, 41124 Modena, Italy; (S.C.); (M.G.T.); (D.A.); (F.G.); (B.B.); (E.C.)
- Laboratory of Experimental Pneumology, Department of Surgical and Medical Science, University of Modena and Reggio Emilia, 41124 Modena, Italy;
- Center for Rare Lung Diseases, University Hospital of Modena, 41124 Modena, Italy
| |
Collapse
|
11
|
Li X, Cui B, Jiang L. Associations between genetic variants of Toll-interacting proteins and interstitial lung diseases: a systematic review and meta-analysis. Orphanet J Rare Dis 2024; 19:432. [PMID: 39578840 PMCID: PMC11583435 DOI: 10.1186/s13023-024-03410-8] [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: 12/19/2023] [Accepted: 10/13/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Genetic polymorphisms in Toll-interacting protein (TOLLIP) have been documented in relation to clinical manifestations of interstitial lung disease (ILD). Nevertheless, the findings across studies present inconsistencies. The present meta-analysis endeavors to elucidate the nexus between genetic variations in TOLLIP and the onset and prognosis of interstitial lung disease (ILD), with the overarching aim of providing insight into the pathophysiological underpinnings of ILD. METHOD This systematic review was registered in PROSPERO. The OVID MEDLINE, OVID EMBASE, and Web of Science electronic databases were searched. RESULTS Fourteen studies with a total of 4821 cases and 9765 controls were examined. The final TOLLIP variants to be included in this meta-analysis were rs5743890, rs111521887, and rs3750920. There were significantly fewer TOLLIP rs5743890 minor allele C carriers among individuals with interstitial lung disease (ILD) than among those without this condition (11.42% vs. 18.92%). Conversely, patients with ILD exhibited higher frequencies of rs111521887 minor allele G carriers (28.92% vs. 22.44%) and rs3750920 minor allele T carriers (40.06% vs. 34.00%). A potential association between rs5743890_C and a reduced incidence of ILD was plausible (p = 0.04, OR = 0.72, 95% CI = 0.53-0.99). Furthermore, a stratified analysis revealed that rs5743890_C was significantly associated with a decreased risk of IPF (p = 0.004, OR = 0.62, 95% CI = 0.44-0.86). There was a significant correlation between susceptibility to ILD and rs111521887 G (p < 0.00001, OR = 1.48, 95% CI = 1.33-1.65) and rs3750920 T (p < 0.00001, OR = 1.34, 95% CI = 1.26-1.44). The survival of IPF patients was correlated with the TOLLIP rs5743890 SNP, and patients with the rs5743890_C genotype had worse survival (p = 0.02, HR = 1.59, 95% CI = 1.07-2.36). CONCLUSION This study showed that rs5743890_C was associated with a lower incidence of ILD and a worse survival rate in patients with IPF. Rs111521887_G and rs3750920_T were found to be associated with an elevated risk of ILD incidence, while no significant association was observed with ILD prognosis. Furthermore, studies are warranted to validate our results and assess the effects of TOLLIP genetic variants on ILD.
Collapse
Affiliation(s)
- Xiaoyuan Li
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Beibei Cui
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Lili Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Monteleone G, ILDs Study Group SIP/IRS, Bergantini L, D’Alessandro M, Pianigiani T, Simonetti J, Iovene B, Varone F, Sgalla G, Richeldi L, Bargagli E, Cameli P. The management of Familial Pulmonary Fibrosis in different medical settings: Where does that leave us? An Italian nationwide survey. SARCOIDOSIS, VASCULITIS, AND DIFFUSE LUNG DISEASES : OFFICIAL JOURNAL OF WASOG 2024; 41:e2024047. [PMID: 39315977 PMCID: PMC11472680 DOI: 10.36141/svdld.v41i3.15744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND AND AIM Familial Pulmonary Fibrosis (FPF) is an emerging group of interstitial lung diseases (ILDs) caused by mutations mainly involving "telomere-related genes" and "surfactant-related genes". Although, in 2023, European Respiratory Society proposed a statement for FPFs management, these still remain a burden. Our work aimed to evaluate the management and impact of FPF in three Italian different medical settings: University Hospitals (UHs), non-University Hospitals (N-UHs) and outpatient clinics. METHODS This survey was created by ILDs Study Group Società Italiana di Pneumologia/ Italian Respiratory Society (SIP-IRS) and diffused via email to all SIP-IRS members. The descriptive statistical analysis was conducted through GraphPad Prism software (version 8.0). Results: Twenty participants replied to the survey, of which 65% (13/20) worked at UH while the remaining 25% (6/20) and 5% (1/20) worked at N-UH and outpatient clinics, respectively. Centers with, at least, 150 ILD patients visits/year followed a higher number of FPF patients, regardless of University affiliation (p=0.0046). Despite significant discrepancies in genetic testing and availability of counselling were registered, no statistically significant differences in patients' anamnesis assessment were observed between UHs and N-UHs (p=0.4192 and p=0.6525). However, there were relevant differences in the number of FPF patients undergoing genetic assessment in the Centers with Genetics Lab or Unit inside the Hospital (p=0.0253). There was no consensus regarding the impact of FPF diagnosis on lung transplantation and screening of asymptomatic relatives. Similarly, no differences were reported in antifibrotic prescriptions between UHs and N-UHs. Although the typical UIP pattern was the most common radiological pattern observed in FPF patients, there were no differences in the prevalence of histopathological patterns between UH and N-UH. CONCLUSIONS Improving pulmonologists' knowledge of the approach, diagnosis and management of FPF is a global medical topic. Scientific societies can provide significant support in raising physicians' awareness of this issue.
Collapse
Affiliation(s)
| | | | - Laura Bergantini
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Miriana D’Alessandro
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Tommaso Pianigiani
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | | | - Bruno Iovene
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Varone
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giacomo Sgalla
- Catholic University of Sacred Heart, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Richeldi
- Catholic University of Sacred Heart, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elena Bargagli
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Paolo Cameli
- Respiratory Diseases Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| |
Collapse
|
13
|
Behr J, Bonella F, Frye BC, Günther A, Hagmeyer L, Henes J, Klemm P, Koschel D, Kreuter M, Leuschner G, Nowak D, Prasse A, Quadder B, Sitter H, Costabel U. Pharmacological Treatment of Idiopathic Pulmonary Fibrosis (Update) and Progressive Pulmonary Fibroses: S2k Guideline of the German Respiratory Society. Respiration 2024; 103:782-810. [PMID: 39250885 DOI: 10.1159/000540856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 07/24/2024] [Indexed: 09/11/2024] Open
Affiliation(s)
- Jürgen Behr
- Department of Medicine V, Comprehensice Pneumology Center Munich, German Center for Lung Research Munich, LMU University Hospital, LMU Munich, Munich, Germany
| | - Francesco Bonella
- Pneumology Department, Center for Interstitial and Rare Lung Diseases, Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Björn Christian Frye
- Department for Pneumology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Andreas Günther
- Center for Interstitial and Rare Lung Diseases, Agaplesion Evangelisches Krankenhaus Mittelhessen, University Hospital Giessen Marburg, Giessen, Germany
| | - Lars Hagmeyer
- Clinic for Pulmonology and Allergology, Center for Sleep Medicine and Respiratory Care, Bethanien Hospital Solingen, Institute for Pulmonology with the University of Cologne, Cologne, Germany
| | - Jörg Henes
- Department for Internal Medicine II (Hematology, Oncology, Rheumatology and Clinical Immunology), University Hospital Tuebingen, Tuebingen, Germany
| | - Philipp Klemm
- Deptartment of Rheumatology and Clinical Immunology, Campus Kerckhoff, Kerckhoff Clinic, Justus-Liebig-University Giessen, Bad Nauheim, Germany
| | - Dirk Koschel
- Fachkrankenhaus Coswig, Lung Center Coswig, and Medical Department I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Michael Kreuter
- Department of Pneumology, Mainz University Medical Center, Mainz, Germany
- Department of Pulmonary, Critical Care and Sleep Medicine, Marienhaus Clinic Mainz, Mainz, Germany
| | - Gabriela Leuschner
- Department of Medicine V, Comprehensice Pneumology Center Munich, German Center for Lung Research Munich, LMU University Hospital, LMU Munich, Munich, Germany
| | - Dennis Nowak
- Institute and Policlinic for Occupational, Social and Environmental Medicine, Omprehensive Pulmonology Center (CPC) Munich, Member of the German Lung Research Center, Munich, Germany
| | - Antje Prasse
- Department of Pulmonology and Infectiology, German DZL BREATH and Fibrosis Research Department, Hannover Medical School, Fraunhofer ITEM, Hannover, Germany
| | | | - Helmut Sitter
- Institute for Surgical Research, Philipps University Marburg, Marburg, Germany
| | - Ulrich Costabel
- Pneumology Department, Center for Interstitial and Rare Lung Diseases, Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
14
|
Gorlanova O, Rüttimann C, Soti A, de Hoogh K, Vienneau D, Künstle N, Da Silva Sena CR, Steinberg R, Bovermann X, Schulzke S, Latzin P, Röösli M, Frey U, Müller L. TOLLIP and MUC5B modulate the effect of ambient NO 2 on respiratory symptoms in infancy. CHEMOSPHERE 2024; 363:142837. [PMID: 39009092 DOI: 10.1016/j.chemosphere.2024.142837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/25/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Current knowledge suggests that the gene region containing MUC5B and TOLLIP plays a role in airway defence and airway inflammation, and hence respiratory disease. It is also known that exposure to air pollution increases susceptibility to respiratory disease. We aimed to study whether the effect of air pollutants on the immune response and respiratory symptoms in infants may be modified by polymorphisms in MUC5B and TOLLIP genes. METHODS 359 healthy term infants from the prospective Basel-Bern Infant Lung Development (BILD) birth cohort were included in the study. The main outcome was the score of weekly assessed respiratory symptoms in the first year of life. Using the candidate gene approach, we selected 10 single nucleotide polymorphisms (SNPs) from the MUC5B and TOLLIP regions. Nitrogen dioxide (NO2) and particulate matter ≤10 μm in aerodynamic diameter (PM10) exposure was estimated on a weekly basis. We used generalised additive mixed models adjusted for known covariates. To validate our results in vitro, cells from a lung epithelial cell line were downregulated in TOLLIP expression and exposed to diesel particulate matter (DPM) and polyinosinic-polycytidylic acid. RESULTS Significant interaction was observed between modelled air pollution (weekly NO2 exposure) and 5 SNPs within MUC5B and TOLLIP genes regarding respiratory symptoms as outcome: E.g., infants carrying minor alleles of rs5744034, rs3793965 and rs3750920 (all TOLLIP) had an increased risk of respiratory symptoms with increasing NO2 exposure. In vitro experiments showed that cells downregulated for TOLLIP react differently to environmental pollutant exposure with DPM and viral stimulation. CONCLUSION Our findings suggest that the effect of air pollution on respiratory symptoms in infancy may be influenced by the genotype of specific SNPs from the MUC5B and TOLLIP regions. For validation of the findings, we provided in vitro evidence for the interaction of TOLLIP with air pollution.
Collapse
Affiliation(s)
- Olga Gorlanova
- University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland; Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Céline Rüttimann
- University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland; Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andras Soti
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Paediatrics and Youth Medicine, Clinic Donaustadt, Vienna, Austria
| | - Kees de Hoogh
- Swiss Tropical and Public Health Institute Basel, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Danielle Vienneau
- Swiss Tropical and Public Health Institute Basel, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Noëmi Künstle
- University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland; Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Carla Rebeca Da Silva Sena
- University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland; Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Priority Research Centre GrowUpWell® and Hunter Medical Research Institute, University of Newcastle, NSW, Australia
| | - Ruth Steinberg
- University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland; Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Xenia Bovermann
- University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland; Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sven Schulzke
- University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Philipp Latzin
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Martin Röösli
- Swiss Tropical and Public Health Institute Basel, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Urs Frey
- University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.
| | - Loretta Müller
- Division of Paediatric Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Lung Precision Medicine, Department for BioMedical Research (DBMR), University of Bern, Switzerland
| |
Collapse
|
15
|
Maddali MV, Moore AR, Sinha P, Newton CA, Kim JS, Adegunsoye A, Ma SF, Strek ME, Chen CH, Linderholm AL, Zemans RL, Moore BB, Wolters PJ, Martinez FJ, Rogers AJ, Raj R, Noth I, Oldham JM. Molecular Endotypes of Idiopathic Pulmonary Fibrosis: A Latent Class Analysis of Two Multicenter Observational Cohorts. Am J Respir Crit Care Med 2024; 210:455-464. [PMID: 38913573 PMCID: PMC11351813 DOI: 10.1164/rccm.202402-0339oc] [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: 02/12/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024] Open
Abstract
Rationale: Idiopathic pulmonary fibrosis (IPF) causes irreversible fibrosis of the lung parenchyma. Although antifibrotic therapy can slow IPF progression, treatment response is variable. There exists a critical need to develop a precision medicine approach to IPF. Objectives: To identify and validate biologically driven molecular endotypes of IPF. Methods: Latent class analysis (LCA) was independently performed in prospectively recruited discovery (n = 875) and validation (n = 347) cohorts. Twenty-five plasma biomarkers associated with fibrogenesis served as class-defining variables. The association between molecular endotype and 4-year transplant-free survival was tested using multivariable Cox regression adjusted for baseline confounders. Endotype-dependent differential treatment response to future antifibrotic exposure was then assessed in a pooled cohort of patients naive to antifibrotic therapy at the time of biomarker measurement (n = 555). Measurements and Main Results: LCA independently identified two latent classes in both cohorts (P < 0.0001). WFDC2 (WAP four-disulfide core domain protein 2) was the most important determinant of class membership across cohorts. Membership in class 2 was characterized by higher biomarker concentrations and a higher risk of death or transplant (discovery, hazard ratio [HR], 2.02; 95% confidence interval [CI], 1.64-2.48; P < 0.001; validation, HR, 1.95; 95% CI, 1.34-2.82; P < 0.001). In pooled analysis, significant heterogeneity in treatment effect was observed between endotypes (P = 0.030 for interaction), with a favorable antifibrotic response in class 2 (HR, 0.64; 95% CI, 0.45-0.93; P = 0.018) but not in class 1 (HR, 1.19; 95% CI, 0.77-1.84; P = 0.422). Conclusions: In this multicohort study, we identified two novel molecular endotypes of IPF with divergent clinical outcomes and responses to antifibrotic therapy. Pending further validation, these endotypes could enable a precision medicine approach for future IPF clinical trials.
Collapse
Affiliation(s)
- Manoj V. Maddali
- Division of Pulmonary, Allergy, and Critical Care Medicine and
- Department of Biomedical Data Science, Stanford University, Stanford, California
| | - Andrew R. Moore
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Pratik Sinha
- Division of Clinical and Translational Research, Washington University School of Medicine, St. Louis, Missouri
- Division of Critical Care, Department of Anesthesia, Washington University, St. Louis, Missouri
| | - Chad A. Newton
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - John S. Kim
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Shwu-Fan Ma
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Mary E. Strek
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Ching-Hsien Chen
- Division of Pulmonary and Critical Care Medicine, University of California, Davis, Davis, California
| | - Angela L. Linderholm
- Division of Pulmonary and Critical Care Medicine, University of California, Davis, Davis, California
| | | | - Bethany B. Moore
- Division of Pulmonary and Critical Care Medicine
- Department of Microbiology and Immunology, and
| | - Paul J. Wolters
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, University of California, San Francisco, San Francisco, California; and
| | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Cornell University, New York, New York
| | | | - Rishi Raj
- Division of Pulmonary, Allergy, and Critical Care Medicine and
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
16
|
Adegunsoye A, Kropski JA, Behr J, Blackwell TS, Corte TJ, Cottin V, Glanville AR, Glassberg MK, Griese M, Hunninghake GM, Johannson KA, Keane MP, Kim JS, Kolb M, Maher TM, Oldham JM, Podolanczuk AJ, Rosas IO, Martinez FJ, Noth I, Schwartz DA. Genetics and Genomics of Pulmonary Fibrosis: Charting the Molecular Landscape and Shaping Precision Medicine. Am J Respir Crit Care Med 2024; 210:401-423. [PMID: 38573068 PMCID: PMC11351799 DOI: 10.1164/rccm.202401-0238so] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/04/2024] [Indexed: 04/05/2024] Open
Abstract
Recent genetic and genomic advancements have elucidated the complex etiology of idiopathic pulmonary fibrosis (IPF) and other progressive fibrotic interstitial lung diseases (ILDs), emphasizing the contribution of heritable factors. This state-of-the-art review synthesizes evidence on significant genetic contributors to pulmonary fibrosis (PF), including rare genetic variants and common SNPs. The MUC5B promoter variant is unusual, a common SNP that markedly elevates the risk of early and established PF. We address the utility of genetic variation in enhancing understanding of disease pathogenesis and clinical phenotypes, improving disease definitions, and informing prognosis and treatment response. Critical research gaps are highlighted, particularly the underrepresentation of non-European ancestries in PF genetic studies and the exploration of PF phenotypes beyond usual interstitial pneumonia/IPF. We discuss the role of telomere length, often critically short in PF, and its link to progression and mortality, underscoring the genetic complexity involving telomere biology genes (TERT, TERC) and others like SFTPC and MUC5B. In addition, we address the potential of gene-by-environment interactions to modulate disease manifestation, advocating for precision medicine in PF. Insights from gene expression profiling studies and multiomic analyses highlight the promise for understanding disease pathogenesis and offer new approaches to clinical care, therapeutic drug development, and biomarker discovery. Finally, we discuss the ethical, legal, and social implications of genomic research and therapies in PF, stressing the need for sound practices and informed clinical genetic discussions. Looking forward, we advocate for comprehensive genetic testing panels and polygenic risk scores to improve the management of PF and related ILDs across diverse populations.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Pulmonary/Critical Care, and
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Juergen Behr
- Department of Medicine V, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, member of the German Center for Lung Research (DZL), Munich, Germany
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Tamera J. Corte
- Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases (OrphaLung), Louis Pradel Hospital, Hospices Civils de Lyon, ERN-LUNG (European Reference Network on Rare Respiratory Diseases), Lyon, France
- Claude Bernard University Lyon, Lyon, France
| | - Allan R. Glanville
- Lung Transplant Unit, St. Vincent’s Hospital Sydney, Sydney, New South Wales, Australia
| | - Marilyn K. Glassberg
- Department of Medicine, Loyola Chicago Stritch School of Medicine, Chicago, Illinois
| | - Matthias Griese
- Department of Pediatric Pneumology, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Munich, Germany
| | - Gary M. Hunninghake
- Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Michael P. Keane
- Department of Respiratory Medicine, St. Vincent’s University Hospital and School of Medicine, University College Dublin, Dublin, Ireland
| | - John S. Kim
- Department of Medicine, School of Medicine, and
| | - Martin Kolb
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Toby M. Maher
- Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York; and
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - David A. Schwartz
- Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
17
|
Whalen W, Berger K, Kim JS, Simmons W, Ma SF, Kaner RJ, Martinez FJ, Anstrom KJ, Parfrey H, Maher TM, Hammond M, Clark AB, Thickett D, Jenkins RG, Wilson AM, Noth I. TOLLIP SNP and Antimicrobial Treatment Effect in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2024; 210:508-511. [PMID: 38762791 PMCID: PMC11351790 DOI: 10.1164/rccm.202312-2224le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/12/2024] [Indexed: 05/20/2024] Open
Affiliation(s)
- William Whalen
- Division of Pulmonary and Critical Care, Department of Medicine
| | - Kristin Berger
- Division of Pulmonary and Critical Care, Department of Medicine
| | - John S. Kim
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Will Simmons
- Division of Biostatistics, Department of Population Health Sciences, and
| | - Shwu-Fan Ma
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Robert J. Kaner
- Division of Pulmonary and Critical Care, Department of Medicine
- Department of Genetic Medicine, New York Presbyterian–Weill Cornell Medicine, New York, New York
| | | | - Kevin J. Anstrom
- University of North Carolina at Chapel Hill Gillings School of Global Public Health, Chapel Hill, North Carolina
| | - Helen Parfrey
- University of Cambridge and Royal Papworth Hospital National Health Service Foundation Trust, Cambridge, United Kingdom
| | - Toby M. Maher
- University of Southern California, Los Angeles, California
- National Heart and Lung Institute, Imperial Biomedical Research Centre, and National Institute for Health and Care Research Margaret Turner Warwick Centre for Fibrosing Lung Disease, Imperial College London, London, United Kingdom
| | | | | | - David Thickett
- Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
| | - R. Gisli Jenkins
- National Heart and Lung Institute, Imperial Biomedical Research Centre, and National Institute for Health and Care Research Margaret Turner Warwick Centre for Fibrosing Lung Disease, Imperial College London, London, United Kingdom
| | - Andrew M. Wilson
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom; and
| | - Imre Noth
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
18
|
Sampsonas F, Bosgana P, Bravou V, Tzouvelekis A, Dimitrakopoulos FI, Kokkotou E. Interstitial Lung Diseases and Non-Small Cell Lung Cancer: Particularities in Pathogenesis and Expression of Driver Mutations. Genes (Basel) 2024; 15:934. [PMID: 39062713 PMCID: PMC11276289 DOI: 10.3390/genes15070934] [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: 06/15/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION Interstitial lung diseases are a varied group of diseases associated with chronic inflammation and fibrosis. With the emerging and current treatment options, survival rates have vastly improved. Having in mind that the most common type is idiopathic pulmonary fibrosis and that a significant proportion of these patients will develop lung cancer as the disease progresses, prompt diagnosis and personalized treatment of these patients are fundamental. SCOPE AND METHODS The scope of this review is to identify and characterize molecular and pathogenetic pathways that can interconnect Interstitial Lung Diseases and lung cancer, especially driver mutations in patients with NSCLC, and to highlight new and emerging treatment options in that view. RESULTS Common pathogenetic pathways have been identified in sites of chronic inflammation in patients with interstitial lung diseases and lung cancer. Of note, the expression of driver mutations in EGFR, BRAF, and KRAS G12C in patients with NSCLC with concurrent interstitial lung disease is vastly different compared to those patients with NSCLC without Interstitial Lung Disease. CONCLUSIONS NSCLC in patients with Interstitial Lung Disease is a challenging diagnostic and clinical entity, and a personalized medicine approach is fundamental to improving survival and quality of life. Newer anti-fibrotic medications have improved survival in IPF/ILD patients; thus, the incidence of lung cancer is going to vastly increase in the next 5-10 years.
Collapse
Affiliation(s)
- Fotios Sampsonas
- Department of Respiratory Medicine, Medical School, University of Patras, 26504 Patras, Greece;
| | - Pinelopi Bosgana
- Department of Pathology, Medical School, University of Patras, 26504 Patras, Greece;
| | - Vasiliki Bravou
- Department of Anatomy, Embryology and Histology, Medical School, University of Patras, 26504 Patras, Greece;
| | - Argyrios Tzouvelekis
- Department of Respiratory Medicine, Medical School, University of Patras, 26504 Patras, Greece;
| | | | - Eleni Kokkotou
- Oncology Unit, The Third Department of Medicine, Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| |
Collapse
|
19
|
Santus P, Signorello JC, Danzo F, Lazzaroni G, Saad M, Radovanovic D. Anti-Inflammatory and Anti-Oxidant Properties of N-Acetylcysteine: A Fresh Perspective. J Clin Med 2024; 13:4127. [PMID: 39064168 PMCID: PMC11278452 DOI: 10.3390/jcm13144127] [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: 05/30/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
N-acetyl-L-cysteine (NAC) was initially introduced as a treatment for mucus reduction and widely used for chronic respiratory conditions associated with mucus overproduction. However, the mechanism of action for NAC extends beyond its mucolytic activity and is complex and multifaceted. Contrary to other mucoactive drugs, NAC has been found to exhibit antioxidant, anti-infective, and anti-inflammatory activity in pre-clinical and clinical reports. These properties have sparked interest in its potential for treating chronic lung diseases, including chronic obstructive pulmonary disease (COPD), bronchiectasis (BE), cystic fibrosis (CF), and idiopathic pulmonary fibrosis (IPF), which are associated with oxidative stress, increased levels of glutathione and inflammation. NAC's anti-inflammatory activity is noteworthy, and it is not solely secondary to its antioxidant capabilities. In ex vivo models of COPD exacerbation, the anti-inflammatory effects have been observed even at very low doses, especially with prolonged treatment. The mechanism involves the inhibition of the activation of NF-kB and neurokinin A production, resulting in a reduction in interleukin-6 production, a cytokine abundantly present in the sputum and breath condensate of patients with COPD and correlates with the number of exacerbations. The unique combination of mucolytic, antioxidant, anti-infective, and anti-inflammatory properties positions NAC as a safe, cost-effective, and efficacious therapy for a plethora of respiratory conditions.
Collapse
Affiliation(s)
- Pierachille Santus
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Juan Camilo Signorello
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Fiammetta Danzo
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Giada Lazzaroni
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
| | - Marina Saad
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20122 Milano, Italy;
| | - Dejan Radovanovic
- Division of Respiratory Diseases, “L. Sacco” University Hospital, Università degli Studi di Milano, 20122 Milano, Italy; (J.C.S.); (F.D.); (G.L.); (D.R.)
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20122 Milano, Italy;
| |
Collapse
|
20
|
Polat G, Özdemir Ö, Ermin S, Serçe Unat D, Demirci Üçsular F. Predictive factors of mortality in patients with idiopathic pulmonary fibrosis treated with antifibrotics: a novel prognostic scoring system. SARCOIDOSIS, VASCULITIS, AND DIFFUSE LUNG DISEASES : OFFICIAL JOURNAL OF WASOG 2024; 41:e2024021. [PMID: 38940720 PMCID: PMC11275550 DOI: 10.36141/svdld.v41i2.13779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 01/25/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND AND AIM Any test that provides sufficient prognostic information to guide treatment decisions in idiopathic pulmonary fibrosis (IPF) is not available. The aim of our study was to determine the predictive factors of mortality in patients with IPF treated with antifibrotics. METHODS Patients with diagnosis of IPF who were treated with antifibrotics between 2016 - 2021 were included in the study. Demographic, clinical and laboratory characteristics of the patients was derived from hospital records retrospectively. Kaplan Meier and multivariate cox regression analysis were achieved for detection of mortality predictors. RESULTS Study population was composed of 119 IPF patients with a male predominance of 80.7% (n=96). Mean age of the patients was 67.9 ± 7.07 years. On univariate analysis, sex was not a significant predictor of mortality (HR 1.79; 95% CI: 0.87 - 3.69, p =0.11). BMI ≤ 26,6 m2/kg, DLCO ≤ 3.11 ml/mmHg/min, age over 62 years, 6DWT ≤ 382 meters, NLR ≤ 2.67 and PDW ≤ 16.7% were found to be significant for predicting mortality. On multivariate cox regression analysis four parameters remained significant for prediction of mortality: RDW > 14%, NLR ≤ 2.67, BMI ≤ 26,6 m2/kg and DLCO ≤ 3.11 ml/mmHg/min (respectively, HR: 2.0. 95% CI: 1.02 - 3.91, p=0.44; HR: 2.68. 95% CI: 1.48 - 4.85, p=0.001, HR: 2.07. 95% CI: 1.14 - 3.76, p=0.02, HR: 3.46. 95% CI: 1.85 - 6.47, p<0.001). A scoring system with these parameters discriminated patients with worse prognosis with a sensitivity of 89.1 % and a specificity of 65.8 % when total point was over 2 (AUC0.83, p<0.001). Conclusions In this study, DLCO, BMI, RDW and NLR levels significantly predicted mortality in IPF patients. Along with GAP index, scoring system with these simple parameters may give information about the prognosis of an IPF patient treated with antifibrotics.
Collapse
Affiliation(s)
- Gülru Polat
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| | - Özer Özdemir
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| | - Sinem Ermin
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| | - Damla Serçe Unat
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| | - Fatma Demirci Üçsular
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| |
Collapse
|
21
|
Perrotta F, Sanduzzi Zamparelli S, D’Agnano V, Montella A, Fomez R, Pagliaro R, Schiattarella A, Cazzola M, Bianco A, Mariniello DF. Genomic Profiling for Predictive Treatment Strategies in Fibrotic Interstitial Lung Disease. Biomedicines 2024; 12:1384. [PMID: 39061958 PMCID: PMC11274143 DOI: 10.3390/biomedicines12071384] [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: 04/17/2024] [Revised: 06/01/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) has traditionally been considered the archetype of progressive fibrotic interstitial lung diseases (f-ILDs), but several other f-ILDs can also manifest a progressive phenotype. Integrating genomic signatures into clinical practice for f-ILD patients may help to identify patients predisposed to a progressive phenotype. In addition to the risk of progressive pulmonary fibrosis, there is a growing body of literature examining how pharmacogenomics influences treatment response, particularly regarding the efficacy and safety profiles of antifibrotic and immunomodulatory agents. In this narrative review, we discuss current studies in IPF and other forms of pulmonary fibrosis, including systemic autoimmune disorders associated ILDs, sarcoidosis and hypersensitivity pneumonitis. We also provide insights into the future direction of research in this complex field.
Collapse
Affiliation(s)
- Fabio Perrotta
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (A.M.); (R.F.); (R.P.); (A.S.); (A.B.)
- Unit of Respiratory Medicine “L. Vanvitelli”, A.O. dei Colli, Monaldi Hospital, 80131 Naples, Italy
| | | | - Vito D’Agnano
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (A.M.); (R.F.); (R.P.); (A.S.); (A.B.)
- Unit of Respiratory Medicine “L. Vanvitelli”, A.O. dei Colli, Monaldi Hospital, 80131 Naples, Italy
| | - Antonia Montella
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (A.M.); (R.F.); (R.P.); (A.S.); (A.B.)
- Unit of Respiratory Medicine “L. Vanvitelli”, A.O. dei Colli, Monaldi Hospital, 80131 Naples, Italy
| | - Ramona Fomez
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (A.M.); (R.F.); (R.P.); (A.S.); (A.B.)
- Unit of Respiratory Medicine “L. Vanvitelli”, A.O. dei Colli, Monaldi Hospital, 80131 Naples, Italy
| | - Raffaella Pagliaro
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (A.M.); (R.F.); (R.P.); (A.S.); (A.B.)
- Unit of Respiratory Medicine “L. Vanvitelli”, A.O. dei Colli, Monaldi Hospital, 80131 Naples, Italy
| | - Angela Schiattarella
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (A.M.); (R.F.); (R.P.); (A.S.); (A.B.)
- Unit of Respiratory Medicine “L. Vanvitelli”, A.O. dei Colli, Monaldi Hospital, 80131 Naples, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Andrea Bianco
- Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.D.); (A.M.); (R.F.); (R.P.); (A.S.); (A.B.)
- Unit of Respiratory Medicine “L. Vanvitelli”, A.O. dei Colli, Monaldi Hospital, 80131 Naples, Italy
| | | |
Collapse
|
22
|
Sciacca E, Muscato G, Spicuzza L, Fruciano M, Gili E, Sambataro G, Palmucci S, Vancheri C, Libra A. Pharmacological treatment in Idiopathic Pulmonary Fibrosis: current issues and future perspectives. Multidiscip Respir Med 2024; 19:982. [PMID: 38869027 PMCID: PMC11186439 DOI: 10.5826/mrm.2024.982] [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: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 06/14/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) represents a fibrotic interstitial lung disease characterized by uncertain etiology and poor prognosis. Over the years, the path to effective treatments has been marked by a series of advances and setbacks. The introduction of approved antifibrotic drugs, pirfenidone and nintedanib, marked a pivotal moment in the management of IPF. However, despite these advances, these drugs are not curative, although they can slow the natural progression of the disease. The history of drug therapy for IPF goes together with the increased understanding of the pathogenic mechanisms underlying the disease. Based on that, current research efforts continue to explore new therapies, possible personalized treatment strategies, drug combinations, and potential biomarkers for diagnosis and prognosis. In this review, we outline the route that led to the discover of the first effective therapies, ongoing clinical trials, and future directions in the search for more effective treatments.
Collapse
Affiliation(s)
- Enrico Sciacca
- Department of Clinical and Experimental Medicine, “Regional Referral Center for Rare Lung Diseases”, University - Hospital Policlinico “G. Rodolico- San Marco”, University of Catania, Catania, Italy
| | - Giuseppe Muscato
- Department of Clinical and Experimental Medicine, “Regional Referral Center for Rare Lung Diseases”, University - Hospital Policlinico “G. Rodolico- San Marco”, University of Catania, Catania, Italy
| | - Lucia Spicuzza
- Department of Clinical and Experimental Medicine, “Regional Referral Center for Rare Lung Diseases”, University - Hospital Policlinico “G. Rodolico- San Marco”, University of Catania, Catania, Italy
| | - Mary Fruciano
- Department of Clinical and Experimental Medicine, “Regional Referral Center for Rare Lung Diseases”, University - Hospital Policlinico “G. Rodolico- San Marco”, University of Catania, Catania, Italy
| | - Elisa Gili
- Department of Clinical and Experimental Medicine, “Regional Referral Center for Rare Lung Diseases”, University - Hospital Policlinico “G. Rodolico- San Marco”, University of Catania, Catania, Italy
| | - Gianluca Sambataro
- Artroreuma s.r.l., Rheumatology outpatient Clinic, Mascalucia (CT), Italy
- Internal Medicine Unit, Department of Clinical and Experimental Medicine, Division of Rheumatology, Cannizzaro Hospital, University of Catania, Catania, Italy
| | - Stefano Palmucci
- Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University -Hospital Policlinico “G. Rodolico-San Marco”, Unità Operativa Semplice Dipartimentale di Imaging Polmonare e Tecniche Radiologiche Avanzate (UOSD IPTRA), Catania, Italy
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, “Regional Referral Center for Rare Lung Diseases”, University - Hospital Policlinico “G. Rodolico- San Marco”, University of Catania, Catania, Italy
| | - Alessandro Libra
- Department of Clinical and Experimental Medicine, “Regional Referral Center for Rare Lung Diseases”, University - Hospital Policlinico “G. Rodolico- San Marco”, University of Catania, Catania, Italy
| |
Collapse
|
23
|
Laniak OT, Winans T, Patel A, Park J, Perl A. Redox Pathogenesis in Rheumatic Diseases. ACR Open Rheumatol 2024; 6:334-346. [PMID: 38664977 PMCID: PMC11168917 DOI: 10.1002/acr2.11668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 06/14/2024] Open
Abstract
Despite being some of the most anecdotally well-known roads to pathogenesis, the mechanisms governing autoimmune rheumatic diseases are not yet fully understood. The overactivation of the cellular immune system and the characteristic development of autoantibodies have been linked to oxidative stress. Typical clinical manifestations, such as joint swelling and deformities and inflammation of the skin and internal organs, have also been connected directly or indirectly to redox mechanisms. The differences in generation and restraint of oxidative stress provide compelling evidence for the broad variety in pathology among rheumatic diseases and explain some of the common triggers and discordant manifestations in these diseases. Growing evidence of redox mechanisms in pathogenesis has provided a broad array of new potential therapeutic targets. Here, we explore the mechanisms by which oxidative stress is generated, explore its roles in autoimmunity and end-organ damage, and discuss how individual rheumatic diseases exhibit unique features that offer targets for therapeutic interventions.
Collapse
Affiliation(s)
- Olivia T. Laniak
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Thomas Winans
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Akshay Patel
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Joy Park
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| | - Andras Perl
- Norton College of MedicineState University of New York Upstate Medical UniversitySyracuse
| |
Collapse
|
24
|
Spagnolo P, Maher TM. A Long and Winding Road: Drug Development in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2024; 209:1072-1073. [PMID: 38445949 PMCID: PMC11092949 DOI: 10.1164/rccm.202402-0290vp] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/05/2024] [Indexed: 03/07/2024] Open
Affiliation(s)
- Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Toby M. Maher
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Southern California Keck School of Medicine, Los Angeles, California; and
- Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
25
|
Raghu G, Torres JM, Bennett RL. Genetic factors for ILD-the path of precision medicine. THE LANCET. RESPIRATORY MEDICINE 2024; 12:350-352. [PMID: 38521082 DOI: 10.1016/s2213-2600(24)00071-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 02/23/2024] [Indexed: 03/25/2024]
Affiliation(s)
- Ganesh Raghu
- Center for Interstitial Lung Diseases, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Juliet M Torres
- Center for Interstitial Lung Diseases, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Robin L Bennett
- Center for Interstitial Lung Diseases, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
26
|
Bando M, Homma S, Date H, Kishi K, Yamauchi H, Sakamoto S, Miyamoto A, Goto Y, Nakayama T, Azuma A, Kondoh Y, Johkoh T, Nishioka Y, Fukuoka J, Miyazaki Y, Yoshino I, Suda T. Japanese guidelines for the treatment of idiopathic pulmonary fibrosis 2023:Revised edition. Respir Investig 2024; 62:402-418. [PMID: 38484504 DOI: 10.1016/j.resinv.2024.02.014] [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: 12/28/2023] [Revised: 02/08/2024] [Accepted: 02/22/2024] [Indexed: 04/20/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease with a poor prognosis and an unknown cause that generally progresses to pulmonary fibrosis and leads to irreversible tissue alteration. The "Guidelines for the treatment of idiopathic pulmonary fibrosis 2017," specializing in the treatment of IPF for the first time in Japan and presenting evidence-based standard treatment methods suited to the state of affairs in Japan, was published in 2017, in line with the 2014 version of "Formulation procedure for Minds Clinical Practice Guidelines." Because new evidence had accumulated, we formulated the "Guidelines for the treatment of Idiopathic Pulmonary Fibrosis 2023 (revised 2nd edition)." While keeping the revision consistent with the ATS/ERS/JRS/ALAT IPF treatment guidelines, new clinical questions (CQs) on pulmonary hypertension were added to the chronic stage, in addition to acute exacerbation and comorbid lung cancer, which greatly affect the prognosis but are not described in the ATS/ERS/JRS/ALAT IPF guidelines. Regarding the advanced stages, we additionally created expert consensus-based advice for palliative care and lung transplantation. The number of CQs increased from 17 in the first edition to 24. It is important that these guidelines be used not only by respiratory specialists but also by general practitioners, patients, and their families; therefore, we plan to revise them appropriately in line with ever-advancing medical progress.
Collapse
Affiliation(s)
- Masashi Bando
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Sakae Homma
- Department of Respiratory Medicine, Toho University Omori Medical Center, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kazuma Kishi
- Department of Respiratory Medicine, Toho University Omori Medical Center, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Hiroyoshi Yamauchi
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Susumu Sakamoto
- Department of Respiratory Medicine, Toho University Omori Medical Center, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541, Japan
| | - Atsushi Miyamoto
- Department of Respiratory Medicine, Respiratory Center, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Yoshihito Goto
- Clinical Research Center, National Hospital Organization Kyoto Medical Center, 1-1, Mukaihata-cho, Fukakusa, Fushimi-ku, Kyoto, Kyoto, 612-8555, Japan
| | - Takeo Nakayama
- Department of Health Informatics, Graduate School of Medicine and School of Public Health, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto, Kyoto, 606-8501, Japan
| | - Arata Azuma
- Pulmonary Medicine, Tokorozawa Mihara General Hospital, 2-2934-3 Mihara-cho, Tokorozawa-shi, Saitama, 359-0045, Japan; Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan
| | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, 160 Nishioiwake-cho, Seto, Aichi, 489-8642, Japan
| | - Takeshi Johkoh
- Department of Radiology, Kansai Rosai Hospital, 3-1-69 Inabaso, Amagasaki, Hyogo, 660-8511, Japan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Junya Fukuoka
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Ichiro Yoshino
- Department of Thoracic Surgery, International University of Health and Welfare Narita Hospital, 852 Hatakeda, Narita City, Chiba, 286-8520, Japan; Department of General Thoracic Surgery, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatus, 431-3192, Japan
| |
Collapse
|
27
|
Selman M, Pardo A. Idiopathic Pulmonary Fibrosis: From Common Microscopy to Single-Cell Biology and Precision Medicine. Am J Respir Crit Care Med 2024; 209:1074-1081. [PMID: 38289233 DOI: 10.1164/rccm.202309-1573pp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/29/2024] [Indexed: 05/02/2024] Open
Affiliation(s)
- Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico; and
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
28
|
Kam MLW, Chong ST, Chan SH, Swigris JJ, Chew EL, Tan YH, Ngeow JYY, Low SY. First ever characterisation of the effects of short telomeres in a Singapore interstitial lung disease cohort. Respir Investig 2024; 62:348-355. [PMID: 38422914 DOI: 10.1016/j.resinv.2024.02.004] [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: 10/24/2023] [Revised: 01/16/2024] [Accepted: 02/11/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Differences in disease behaviour and genotypes are described in Asian and Western interstitial lung disease (ILD) cohorts. Short leukocyte telomere length (LTL) correlates with poor outcomes in Western ILD cohorts but its significance in Asian populations is unknown. We aim to characterise the burden and clinical implications of short LTL in Singaporean ILD patients. METHODS Patients diagnosed with ILD at Singapore General Hospital were prospectively recruited and compared against 36 healthy controls. The primary outcome was transplant-free survival. Genomic DNA from peripheral blood was extracted and LTL measured using quantitative polymerase chain reaction assay (qPCR). RESULTS Amongst 165 patients, 37% had short LTL. There was a higher proportion of combined pulmonary fibrosis and emphysema (CPFE) patients with short LTL (n = 21, 34.4% vs n = 16, 15.4%; p < 0.001). Short LTL patients had reduced survival at 12-, 24- and 36-months and median survival of 24 months (p < 0.001) which remained significant following adjustment for smoking, GAP Stage and radiological UIP pattern (Hazard Ratio (HR), 2.74; 95%CI:1.46, 5.11; p = 0.002). They had increased respiratory-related mortality and acute exacerbation incidences. Despite similar baseline lung function, short LTL patients had a faster decline in absolute forced vital capacity (FVC) of -105.3 (95% CI: 151.4, -59.1) mL/year compared to -58.2 (95% CI: 82.9, -33.6) mL/year (p < 0.001) in normal LTL patients. CONCLUSION Short LTL correlated with increased mortality and faster lung function decline in our Singaporean ILD cohort with a magnitude similar to that in Western ILD cohorts. Further research is needed to integrate LTL assessment into clinical practice.
Collapse
Affiliation(s)
- Michelle Li Wei Kam
- Department of Respiratory and Critical Care Medicine, Singapore General Hospital, Outram Road, 169608, Singapore; Center for Interstitial Lung Disease, National Jewish Health, 1400 Jackson Street, Denver, CO, USA.
| | - Siao Ting Chong
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, 308232, Singapore
| | - Sock Hoai Chan
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, 168583, Singapore
| | - Jeffrey J Swigris
- Center for Interstitial Lung Disease, National Jewish Health, 1400 Jackson Street, Denver, CO, USA
| | - Ee Ling Chew
- Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, 168583, Singapore
| | - Yi Hern Tan
- Department of Respiratory and Critical Care Medicine, Singapore General Hospital, Outram Road, 169608, Singapore
| | - Joanne Yuen Yie Ngeow
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, 308232, Singapore; Cancer Genetics Service, Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, 168583, Singapore
| | - Su Ying Low
- Department of Respiratory and Critical Care Medicine, Singapore General Hospital, Outram Road, 169608, Singapore
| |
Collapse
|
29
|
Enomoto N. Relationship between idiopathic interstitial pneumonias (IIPs) and connective tissue disease-related interstitial lung disease (CTD-ILD): A narrative review. Respir Investig 2024; 62:465-480. [PMID: 38564878 DOI: 10.1016/j.resinv.2024.03.006] [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: 11/03/2023] [Revised: 01/17/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Abstract
While idiopathic interstitial pneumonia (IIP) centering on idiopathic pulmonary fibrosis (IPF) is the most prevalent interstitial lung disease (ILD), especially in the older adult population, connective tissue disease (CTD)-related ILD is the second most prevalent ILD. The pathogenesis of IPF is primarily fibrosis, whereas that of other ILDs, particularly CTD-ILD, is mainly inflammation. Therefore, a precise diagnosis is crucial for selecting appropriate treatments, such as antifibrotic or immunosuppressive agents. In addition, some patients with IIP have CTD-related features, such as arthritis and skin eruption, but do not meet the criteria for any CTD, this is referred to as interstitial pneumonia with autoimmune features (IPAF). IPAF is closely associated with idiopathic nonspecific interstitial pneumonia (iNSIP) and cryptogenic organizing pneumonia (COP). Furthermore, patients with iNSIP or those with NSIP with OP overlap frequently develop polymyositis/dermatomyositis after the diagnosis of IIP. Acute exacerbation of ILD, the most common cause of death, occurs more frequently in patients with IPF than in those with other ILDs. Although acute exacerbation of CTD-ILD occurs at a low rate of incidence, patients with rheumatoid arthritis, microscopic polyangiitis, or systemic sclerosis experience more acute exacerbation of CTD-ILD than those with other CTD. In this review, the features of each IIP, focusing on CTD-related signatures, are summarized, and the pathogenesis and appropriate treatments to improve the prognoses of patients with various ILDs are discussed.
Collapse
Affiliation(s)
- Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan; Health Administration Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, 431-3192, Japan.
| |
Collapse
|
30
|
Althobiani MA, Russell AM, Jacob J, Ranjan Y, Folarin AA, Hurst JR, Porter JC. Interstitial lung disease: a review of classification, etiology, epidemiology, clinical diagnosis, pharmacological and non-pharmacological treatment. Front Med (Lausanne) 2024; 11:1296890. [PMID: 38698783 PMCID: PMC11063378 DOI: 10.3389/fmed.2024.1296890] [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: 09/19/2023] [Accepted: 03/26/2024] [Indexed: 05/05/2024] Open
Abstract
Interstitial lung diseases (ILDs) refer to a heterogeneous and complex group of conditions characterized by inflammation, fibrosis, or both, in the interstitium of the lungs. This results in impaired gas exchange, leading to a worsening of respiratory symptoms and a decline in lung function. While the etiology of some ILDs is unclear, most cases can be traced back to factors such as genetic predispositions, environmental exposures (including allergens, toxins, and air pollution), underlying autoimmune diseases, or the use of certain medications. There has been an increase in research and evidence aimed at identifying etiology, understanding epidemiology, improving clinical diagnosis, and developing both pharmacological and non-pharmacological treatments. This review provides a comprehensive overview of the current state of knowledge in the field of interstitial lung diseases.
Collapse
Affiliation(s)
- Malik A. Althobiani
- Royal Free Campus, UCL Respiratory, University College London, London, United Kingdom
- Department of Respiratory Therapy, Faculty of Medical Rehabilitation Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anne-Marie Russell
- School of Health and Care Professions, University of Exeter, Exeter, United Kingdom
- School of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Joseph Jacob
- UCL Respiratory, University College London, London, United Kingdom
- Satsuma Lab, Centre for Medical Image Computing, University College London Respiratory, University College London, London, United Kingdom
| | - Yatharth Ranjan
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Amos A. Folarin
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, King's College London, London, United Kingdom
- Institute of Health Informatics, University College London, London, United Kingdom
- NIHR Biomedical Research Centre at University College London Hospitals, NHS Foundation Trust, London, United Kingdom
| | - John R. Hurst
- Royal Free Campus, UCL Respiratory, University College London, London, United Kingdom
| | - Joanna C. Porter
- UCL Respiratory, University College London, London, United Kingdom
| |
Collapse
|
31
|
Ma J, Li G, Wang H, Mo C. Comprehensive review of potential drugs with anti-pulmonary fibrosis properties. Biomed Pharmacother 2024; 173:116282. [PMID: 38401514 DOI: 10.1016/j.biopha.2024.116282] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/02/2024] [Accepted: 02/17/2024] [Indexed: 02/26/2024] Open
Abstract
Pulmonary fibrosis is a chronic and progressive lung disease characterized by the accumulation of scar tissue in the lungs, which leads to impaired lung function and reduced quality of life. The prognosis for idiopathic pulmonary fibrosis (IPF), which is the most common form of pulmonary fibrosis, is generally poor. The median survival for patients with IPF is estimated to be around 3-5 years from the time of diagnosis. Currently, there are two approved drugs (Pirfenidone and Nintedanib) for the treatment of IPF. However, Pirfenidone and Nintedanib are not able to reverse or cure pulmonary fibrosis. There is a need for new pharmacological interventions that can slow or halt disease progression and cure pulmonary fibrosis. This review aims to provide an updated overview of current and future drug interventions for idiopathic pulmonary fibrosis, and to summarize possible targets of potential anti-pulmonary fibrosis drugs, providing theoretical support for further clinical combination therapy or the development of new drugs.
Collapse
Affiliation(s)
- Jie Ma
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; The Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Gang Li
- Department of Thoracic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Han Wang
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Center for RNA Science and Therapeutics, School of Medicine, Cleveland, OH, USA
| | - Chunheng Mo
- The Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
32
|
Amati F, Stainer A, Aliberti S. Response to: are there over 200 distinct types of interstitial lung diseases? Respir Res 2024; 25:114. [PMID: 38448986 PMCID: PMC10918899 DOI: 10.1186/s12931-024-02751-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024] Open
Affiliation(s)
- Francesco Amati
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20072, Italy.
- Respiratory Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan, 20089, Italy.
| | - Anna Stainer
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20072, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan, 20089, Italy
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, 20072, Italy
- Respiratory Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan, 20089, Italy
| |
Collapse
|
33
|
Luo Y, Wang H, Wang L, Wu W, Zhao J, Li X, Xiong R, Ding X, Yuan D, Yuan C. LncRNA MEG3: Targeting the Molecular Mechanisms and Pathogenic causes of Metabolic Diseases. Curr Med Chem 2024; 31:6140-6153. [PMID: 37855346 DOI: 10.2174/0109298673268051231009075027] [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: 07/03/2023] [Revised: 08/27/2023] [Accepted: 09/08/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Non-coding RNA is a type of RNA that does not encode proteins, distributed among rRNA, tRNA, snRNA, snoRNA, microRNA and other RNAs with identified functions, where the Long non-coding RNA (lncRNA) displays a nucleotide length over 200. LncRNAs enable multiple biological processes in the human body, including cancer cell invasion and metastasis, apoptosis, cell autophagy, inflammation, etc. Recently, a growing body of studies has demonstrated the association of lncRNAs with obesity and obesity-induced insulin resistance and NAFLD, where MEG3 is related to glucose metabolism, such as insulin resistance. In addition, MEG3 has been demonstrated in the pathological processes of various cancers, such as mediating inflammation, cardiovascular disease, liver disease and other metabolic diseases. OBJECTIVE To explore the regulatory role of lncRNA MEG3 in metabolic diseases. It provides new ideas for clinical treatment or experimental research. METHODS In this paper, in order to obtain enough data, we integrate and analyze the data in the PubMed database. RESULTS LncRNA MEG3 can regulate many metabolic diseases, such as insulin resistance, NAFLD, inflammation and so on. CONCLUSION LncRNA MEG3 has a regulatory role in a variety of metabolic diseases, which are currently difficult to be completely cured, and MEG3 is a potential target for the treatment of these diseases. Here, we review the role of lncRNA MEG3 in mechanisms of action and biological functions in human metabolic diseases.
Collapse
Affiliation(s)
- Yiyang Luo
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Hailin Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Lijun Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- Department of Biochemistry, College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Wei Wu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Jiale Zhao
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Xueqing Li
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Ruisi Xiong
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- Department of Biochemistry, College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Xueliang Ding
- Department of Clinical Laboratory, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443002, China
| | - Ding Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Chengfu Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- Department of Biochemistry, College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| |
Collapse
|
34
|
Chow Y, López‐Martínez C, Liles WC, Altemeier WA, Gharib SA, Hung CF. Toll-interacting protein inhibits transforming growth factor beta signaling in mouse lung fibroblasts. FASEB Bioadv 2024; 6:12-25. [PMID: 38223200 PMCID: PMC10782472 DOI: 10.1096/fba.2023-00054] [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: 05/22/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 01/16/2024] Open
Abstract
Variations in the Toll-interacting protein (TOLLIP) gene have been identified in genome-wide association studies to correlate with risk of disease, mortality, and response to N-acetylcysteine therapy in idiopathic pulmonary fibrosis. Although TOLLIP is known to modulate innate immune responses, its relevance in organ fibrogenesis remains unknown. Prior work in the literature suggests TOLLIP dampens transforming growth factor beta (TGFβ) signaling in human cell lines. In this study, we examined the role of TOLLIP in mouse lung fibroblast (MLF) responses to TGFβ and in the bleomycin model of experimental lung fibrosis using Tollip-/- mice. We hypothesize that if TOLLIP negatively regulates TGFβ signaling, then Tollip-/- mouse lung fibroblasts (MLFs) would have enhanced response to TGFβ treatment, and Tollip-/- mice would develop increased fibrosis following bleomycin challenge. Primary MLFs were stimulated with TGFβ (1 ng/mL) for 24 h. RNA was obtained to assess global transcriptional responses by RNA-seq and markers of myofibroblast transition by qPCR. Functional assessment of TGFβ-stimulated MLFs included cell migration by scratch assay, cell proliferation, and matrix invasion through Matrigel. In the in vivo model of lung fibrosis, Tollip-/- mice and wild-type (WT) littermates were administered bleomycin intratracheally and assessed for fibrosis. We further examined TGFβ signaling in vivo after bleomycin injury by SMAD2, ERK1/2, and TGFβR1 Western blot. In response to TGFβ treatment, both WT and Tollip-/- MLFs exhibited global transcriptional changes consistent with myofibroblast differentiation. However, Tollip-/- MLFs showed greater number of differentially expressed genes compared to WT MLFs and greater upregulation of Acta2 by qPCR. Functionally, Tollip-/- MLFs also exhibited increased migration and Matrigel invasiveness compared to WT. We found evidence of enhanced TGFβ signaling in Tollip-/- through SMAD2 in vitro and in vivo. Tollip-/- mice experienced lower survival using a standard weight-adjusted dosing without evidence of differences in fibrosis at Day 21. With adjustment of dosing for sex, no differences were observed in fibrosis at Day 21. However, Tollip-/- mice had greater weight loss and increased bronchoalveolar lavage fluid total protein during early resolution at Day 14 compared to WT without evidence of differences in acute lung injury at Day 7, suggesting impaired resolution of lung injury.
Collapse
Affiliation(s)
- Yu‐Hua Chow
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
- Center for Lung BiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Cecilia López‐Martínez
- Instituto de Investigación Sanitaria del Principado de AsturiasOviedoSpain
- Centro de Investigación Biomédica en Red (CIBER)‐Enfermedades respiratoriasMadridSpain
- Instituto Universitario de Oncología del Principado de AsturiasOviedoSpain
| | - W. Conrad Liles
- Center for Lung BiologyUniversity of WashingtonSeattleWashingtonUSA
- Division of Allergy and Infectious Diseases, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - William A. Altemeier
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
- Center for Lung BiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Sina A. Gharib
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
- Center for Lung BiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Chi F. Hung
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
- Center for Lung BiologyUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
35
|
Guo S, Dong Y, Wang C, Jiang Y, Xiang R, Fan LL, Luo H, Liu L. Integrative analysis reveals the recurrent genetic etiologies in idiopathic pulmonary fibrosis. QJM 2023; 116:983-992. [PMID: 37688571 DOI: 10.1093/qjmed/hcad206] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/04/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is increasingly recognized as a chronic, progressive and fatal lung disease with an unknown etiology. Current studies focus on revealing the genetic factors in the risk of IPF, making the integrative analysis of genetic variations and transcriptomic alterations of substantial value. AIM This study aimed to improve the understanding of the molecular basis of IPF through an integrative analysis of whole-exome sequencing (WES), bulk RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq) data. METHODS WES is a powerful tool for studying the genetic basis of IPF, allowing for the identification of genetic variants that may be associated with the development of the disease. RNA-seq data provide a comprehensive view of the transcriptional changes in IPF patients, while scRNA-seq data offer a more granule view of cell-type-specific alterations. RESULTS In this study, we identified a comprehensive mutational landscape of recurrent genomic and transcriptomic variations, including single-nucleotide polymorphisms, CNVs and differentially expressed genes, in IPF populations, which may play a significant role in the development and progression of IPF. CONCLUSIONS Our study provided valuable insights into the genetic and transcriptomic variations associated with IPF, revealing changes in gene expression that may contribute to disease development and progression. These findings highlight the importance of an integrative approach to understanding the molecular mechanisms underlying IPF and may pave the way for identifying potential therapeutic targets.
Collapse
Affiliation(s)
- S Guo
- From the Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, the Second Xiangya Hospital, Central South University, Changsha, China
- Department of Cell Biology, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Y Dong
- Department of Cell Biology, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - C Wang
- Department of Cell Biology, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Y Jiang
- Department of Cell Biology, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Department of Computer Science, Wake Forest University, Winston-Salem, NC, USA
| | - R Xiang
- Department of Cell Biology, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - L-L Fan
- From the Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, the Second Xiangya Hospital, Central South University, Changsha, China
- Department of Cell Biology, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - H Luo
- From the Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, the Second Xiangya Hospital, Central South University, Changsha, China
| | - L Liu
- From the Department of Pulmonary and Critical Care Medicine, Research Unit of Respiratory Disease, Hunan Diagnosis and Treatment Center of Respiratory Disease, the Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
36
|
Sofia C, Comes A, Sgalla G, Richeldi L. An update on emerging drugs for the treatment of idiopathic pulmonary fibrosis: a look towards 2023 and beyond. Expert Opin Emerg Drugs 2023; 28:283-296. [PMID: 37953604 DOI: 10.1080/14728214.2023.2281416] [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: 07/13/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
INTRODUCTION Currently approved drug treatments for idiopathic pulmonary fibrosis (IPF), pirfenidone and nintedanib, have been shown to slow lung function decline and improve clinical outcomes. Since significant advances in the understanding of pathogenetic mechanisms in IPF, novel potential agents are being tested to identify new targeted and better tolerated therapeutic strategies. AREAS COVERED This review describes the evidence from IPF phase II and III clinical trials that have been completed or are ongoing in recent years. The literature search was performed using Medline and Clinicaltrials.org databases. Particular attention is paid to the new inhibitor of phosphodiesterase 4B (BI 1015550), being studied in a more advanced research phase. Some emerging critical issues of the pharmacological research are highlighted considering the recent outstanding failures of several phase III trials. EXPERT OPINION An exponential number of randomized clinical trials are underway testing promising new molecules to increase treatment choices for patients with IPF and improve patients' quality of life. The next goals should aim at a deeper understanding of the pathogenic pathways of the disease with the challenging goal of being able not only to stabilize but also to reverse the ongoing fibrotic process in patients with IPF.
Collapse
Affiliation(s)
- Carmelo Sofia
- Dipartimento di scienze mediche e chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessia Comes
- Dipartimento di scienze mediche e chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giacomo Sgalla
- Dipartimento di scienze mediche e chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luca Richeldi
- Dipartimento di scienze mediche e chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
37
|
Li Q, Wang Y, Ji L, He J, Liu H, Xue W, Yue H, Dong R, Liu X, Wang D, Zhang H. Cellular and molecular mechanisms of fibrosis and resolution in bleomycin-induced pulmonary fibrosis mouse model revealed by spatial transcriptome analysis. Heliyon 2023; 9:e22461. [PMID: 38125541 PMCID: PMC10730595 DOI: 10.1016/j.heliyon.2023.e22461] [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: 05/27/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
The bleomycin-induced pulmonary fibrosis mouse model is commonly used in idiopathic pulmonary fibrosis research, but its cellular and molecular changes and efficiency as a model at the molecular level are not fully understood. In this study, we used spatial transcriptome technology to investigate the cellular and molecular changes in the lungs of bleomycin-induced pulmonary fibrosis mouse models. Our analyses revealed cell dynamics during fibrosis in epithelial cells, mesenchymal cells, immunocytes, and erythrocytes with their spatial distribution available. We confirmed the differentiation of the alveolar type II (AT2) cell type expressing Krt8, and we inferred their trajectories from both the AT2 cells and club cells. In addition to the fibrosis process, we also noticed evidence of self-resolving, especially to identify possible self-resolving related genes, including Prkca. Our findings provide insights into the cellular and molecular mechanisms underlying fibrosis resolution and represent the first spatiotemporal transcriptome dataset of the bleomycin-induced fibrosis mouse model.
Collapse
Affiliation(s)
| | - Yue Wang
- BGI-Beijing, Beijing 102601, China
| | - Liu Ji
- Dalian Maternal and Child Health Hospital of Liaoning Province, Dalian 116033, China
| | - Jianhan He
- Department of Clinical Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | | | | | - Huihui Yue
- Department of Clinical Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Ruihan Dong
- Department of Clinical Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| | - Xin Liu
- BGI-Beijing, Beijing 102601, China
| | - Daqing Wang
- Dalian Maternal and Child Health Hospital of Liaoning Province, Dalian 116033, China
| | - Huilan Zhang
- Department of Clinical Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, Hubei, China
| |
Collapse
|
38
|
Zhang XL, Cao Y, Zheng B. Efficacy of N-acetylcysteine plus pirfenidone in the treatment of idiopathic pulmonary fibrosis: a systematic review and meta-analysis. BMC Pulm Med 2023; 23:479. [PMID: 38031002 PMCID: PMC10685588 DOI: 10.1186/s12890-023-02778-w] [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: 07/10/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Numerous studies have demonstrated the potential of pirfenidone to enhance the prognosis of patients afflicted with idiopathic pulmonary fibrosis (IPF). Although N-acetylcysteine (NAC) is utilized as an antioxidant in IPF treatment, the combination of NAC and pirfenidone has produced inconsistent outcomes in certain studies. To assess the clinical effectiveness and safety of NAC plus pirfenidone (designated as the treatment group) versus pirfenidone monotherapy (designated as the control group), we conducted a systematic review and meta-analysis of randomized controlled trials (RCTs). METHODS RCTs of NAC plus pirfenidone were reviewed searching from databases and networks of unpublished and published studies in any language. Using pair-wise meta-analysis, changes in pulmonary function test (PFT) parameters and safety were evaluated. RESULTS Two independent reviewers selected and obtained data from 5 RCTs (n = 398), comprising 1 study from Japan, 1 from Europe, and 3 from China. NAS plus pirfenidone as compared to pirfenidone monotherapy for IPF may not reduce the incidence of skin effects(RR 1.26 [95%CI 0.64 to 2.45]) and mortality(RR 0.35 [95%CI 0.07 to 1.68])(both moderate certainty). NAS plus pirfenidone as compared to pirfenidone monotherapy for IPF may not reduce the incidence of at least one side effects(RR 1.00 [95%CI 0.84 to 1.19]; low certainty),severe side effects(RR 0.67 [95%CI 0.30 to 1.47]; low certainty) and gastrointestinal effects(RR 0.67 [95%CI 0.41 to 1.09]; low certainty) with possibly no effect in Δ%DLco(SMD -0.17 [95%CI -0.15 to 0.48]; low certainty). Meanwhile, the effect of NAS plus pirfenidone as compared to pirfenidone monotherapy on ΔFVC(SMD 0.18 [95%CI -0.68 to 1.05]), Δ%FVC(SMD -2.62 [95%CI -5.82 to 0.59]) and Δ6MWT(SMD -0.35 [95%CI -0.98 to 0.28]) is uncertain(extremely low certainty). CONCLUSION Moderate certainty evidence suggests that NAS plus pirfenidone, compared to pirfenidone monotherapy for IPF, does not reduce the incidence of skin effects and mortality.
Collapse
Affiliation(s)
- Xiu-Li Zhang
- Medical Department, Chengdu Qingbaijiang District People's Hospital, No 9, Fenghuang East Fourth Road, Qingbaijiang District, Chengdu, 610300, China
| | - Ying Cao
- Department of Infectious Diseases, Chengdu Xinjin District People's Hospital, No 149, Wujin West Road, Xinjin District, Chengdu, 611430, China
| | - Bo Zheng
- Medical Department, Chengdu Qingbaijiang District People's Hospital, No 9, Fenghuang East Fourth Road, Qingbaijiang District, Chengdu, 610300, China.
| |
Collapse
|
39
|
Cottin V, Kolb M. Leukocyte telomere length: the dawn of a new era of personalised medicine in fibrotic interstitial lung diseases? Eur Respir J 2023; 62:2301852. [PMID: 38035695 DOI: 10.1183/13993003.01852-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023]
Affiliation(s)
- Vincent Cottin
- Department of Respiratory Medicine, National Reference Centre for Rare Pulmonary Diseases, member of ERN-LUNG, Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France
- UMR 754, INRAE, Claude Bernard University Lyon 1, Lyon, France
| | - Martin Kolb
- Department of Medicine, McMaster University and St. Joseph's Healthcare, Hamilton, ON, Canada
| |
Collapse
|
40
|
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: 5] [Impact Index Per Article: 2.5] [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
|
41
|
Caminati A, Awad I, Elia D, Cassandro R, Mozzanica F, Pelosi G, Zompatori M, Harari S. Significant functional improvement in IPF patient treated with antifibrotic drugs: preliminary results and clinical outcome. Minerva Med 2023; 114:736-738. [PMID: 37310710 DOI: 10.23736/s0026-4806.23.08720-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Affiliation(s)
- Antonella Caminati
- Unit of Pulmonology and Semi-Intensive Respiratory Therapy, Section of Respiratory Pathophysiology and Pulmonary Hemodynamics, MultiMedica IRCCS, Milan, Italy -
| | - Inas Awad
- Unit of Pulmonology and Semi-Intensive Respiratory Therapy, Section of Respiratory Pathophysiology and Pulmonary Hemodynamics, MultiMedica IRCCS, Milan, Italy
| | - Davide Elia
- Unit of Pulmonology and Semi-Intensive Respiratory Therapy, Section of Respiratory Pathophysiology and Pulmonary Hemodynamics, MultiMedica IRCCS, Milan, Italy
| | - Roberto Cassandro
- Unit of Pulmonology and Semi-Intensive Respiratory Therapy, Section of Respiratory Pathophysiology and Pulmonary Hemodynamics, MultiMedica IRCCS, Milan, Italy
| | - Francesco Mozzanica
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Department of Otorhinolaryngology, IRCCS Multimedica, Milan, Italy
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Pathology, IRCCS MultiMedica, Milan, Italy
| | | | - Sergio Harari
- Unit of Pulmonology and Semi-Intensive Respiratory Therapy, Section of Respiratory Pathophysiology and Pulmonary Hemodynamics, MultiMedica IRCCS, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
42
|
Mokra D, Mokry J, Barosova R, Hanusrichterova J. Advances in the Use of N-Acetylcysteine in Chronic Respiratory Diseases. Antioxidants (Basel) 2023; 12:1713. [PMID: 37760016 PMCID: PMC10526097 DOI: 10.3390/antiox12091713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
N-acetylcysteine (NAC) is widely used because of its mucolytic effects, taking part in the therapeutic protocols of cystic fibrosis. NAC is also administered as an antidote in acetaminophen (paracetamol) overdosing. Thanks to its wide antioxidative and anti-inflammatory effects, NAC may also be of benefit in other chronic inflammatory and fibrotizing respiratory diseases, such as chronic obstructive pulmonary disease, bronchial asthma, idiopathic lung fibrosis, or lung silicosis. In addition, NAC exerts low toxicity and rare adverse effects even in combination with other treatments, and it is cheap and easily accessible. This article brings a review of information on the mechanisms of inflammation and oxidative stress in selected chronic respiratory diseases and discusses the use of NAC in these disorders.
Collapse
Affiliation(s)
- Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Juraj Mokry
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia;
| | - Romana Barosova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Juliana Hanusrichterova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| |
Collapse
|
43
|
Karampitsakos T, Juan-Guardela BM, Tzouvelekis A, Herazo-Maya JD. Precision medicine advances in idiopathic pulmonary fibrosis. EBioMedicine 2023; 95:104766. [PMID: 37625268 PMCID: PMC10469771 DOI: 10.1016/j.ebiom.2023.104766] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/07/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a highly heterogeneous, unpredictable and ultimately lethal chronic lung disease. Over the last decade, two anti-fibrotic agents have been shown to slow disease progression, however, both drugs are administered uniformly with minimal consideration of disease severity and inter-individual molecular, genetic, and genomic differences. Advances in biological understanding of disease endotyping and the emergence of precision medicine have shown that "a one-size-fits-all approach" to the management of chronic lung diseases is no longer appropriate. While precision medicine approaches have revolutionized the management of other diseases such as lung cancer and asthma, the implementation of precision medicine in IPF clinical practice remains an unmet need despite several reports demonstrating a large number of diagnostic, prognostic and theragnostic biomarker candidates in IPF. This review article aims to summarize our current knowledge of precision medicine in IPF and highlight barriers to translate these research findings into clinical practice.
Collapse
Affiliation(s)
- Theodoros Karampitsakos
- Division of Pulmonary, Critical Care and Sleep Medicine, Ubben Center for Pulmonary Fibrosis Research, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Brenda M Juan-Guardela
- Division of Pulmonary, Critical Care and Sleep Medicine, Ubben Center for Pulmonary Fibrosis Research, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | - Jose D Herazo-Maya
- Division of Pulmonary, Critical Care and Sleep Medicine, Ubben Center for Pulmonary Fibrosis Research, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
44
|
Kubbara A, Amundson WH, Herman A, Lee AM, Bishop JR, Kim HJ. Genetic variations in idiopathic pulmonary fibrosis and patient response to pirfenidone. Heliyon 2023; 9:e18573. [PMID: 37560683 PMCID: PMC10407116 DOI: 10.1016/j.heliyon.2023.e18573] [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: 03/02/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Genetic variations in Idiopathic Pulmonary Fibrosis (IPF) affect survival and outcomes. Current antifibrotic agents are managed based on the patient's reported side effects, although certain single nucleotide polymorphisms (SNPs) might alter treatment response and survival depending on the antifibrotic administered. This study investigated variations in response and outcomes to pirfenidone based on patients-specific genetic profiles. METHODS Retrospective clinical data were collected from 56 IPF patients and had blood drawn for DNA extraction between 7/2013 and 3/2016, with the last patient followed until 10/2018. Nine SNPs were selected for pharmacogenetic investigation based on prior associations with IPF treatment outcomes or implications for pirfenidone metabolism. Genetic variants were examined in relation to clinical data and treatment outcomes. RESULTS Of the 56 patients, 38 were males (67.85%). The average age of IPF at diagnosis was 66.88 years. At the initiation of pirfenidone, the average percent predicted FVC was 70.7%, and the average DLCO percent predicted was 50.02% (IQR 40-61%). Among the genetic variants tested, the TOLLIP rs5743890 risk allele was significantly associated with improved survival, with increasing pirfenidone duration. This finding was observed with CC or CT genotype carriers but not for those with the TT genotype (p = 0.0457). Similarly, the TGF-B1 rs1800470 risk allele was also significantly associated with improved survival with longer pirfenidone therapy (p = 0.0395), even though it was associated with disease progression. CONCLUSION This pilot study suggests that in IPF patients, the TOLLIP rs5743890 genotypes CC and CT, as well as TGF-B1 rs 1800470 may be associated with increased survival when treated with pirfenidone.
Collapse
Affiliation(s)
- Aahd Kubbara
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep, University of Minnesota, Minneapolis, United States
| | - William H. Amundson
- Pulmonary and Critical Care Medicine, Regions Hospital, University of Minnesota, St. Paul, Minneapolis, MN, United States
| | - Adam Herman
- University of Minnesota, Supercomputing Institute, Minneapolis, United States
| | - Adam M. Lee
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, United States
| | - Jeffrey R. Bishop
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, United States
| | - Hyun Joo Kim
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep, University of Minnesota, Minneapolis, United States
| |
Collapse
|
45
|
Drakopanagiotakis F, Markart P, Steiropoulos P. Acute Exacerbations of Interstitial Lung Diseases: Focus on Biomarkers. Int J Mol Sci 2023; 24:10196. [PMID: 37373339 DOI: 10.3390/ijms241210196] [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: 03/16/2023] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Interstitial lung diseases (ILDs) are a large group of pulmonary disorders characterized histologically by the cardinal involvement of the pulmonary interstitium. The prototype of ILDs is idiopathic pulmonary fibrosis (IPF), an incurable disease characterized by progressive distortion and loss of normal lung architecture through unchecked collagen deposition. Acute exacerbations are dramatic events during the clinical course of ILDs, associated with high morbidity and mortality. Infections, microaspiration, and advanced lung disease might be involved in the pathogenesis of acute exacerbations. Despite clinical scores, the prediction of the onset and outcome of acute exacerbations is still inaccurate. Biomarkers are necessary to characterize acute exacerbations better. We review the evidence for alveolar epithelial cell, fibropoliferation, and immunity molecules as potential biomarkers for acute exacerbations of interstitial lung disease.
Collapse
Affiliation(s)
- Fotios Drakopanagiotakis
- Department of Respiratory Medicine, Medical School, Democritus University, 68100 Alexandroupolis, Greece
| | - Philipp Markart
- Department of Respiratory Medicine, Klinikum Fulda and University Medicine Campus Fulda, Pacelliallee 4, 36043 Fulda, Germany
| | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University, 68100 Alexandroupolis, Greece
| |
Collapse
|
46
|
Trachalaki A, Sultana N, Wells AU. An update on current and emerging drug treatments for idiopathic pulmonary fibrosis. Expert Opin Pharmacother 2023:1-18. [PMID: 37183672 DOI: 10.1080/14656566.2023.2213436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
INTRODUCTION Idiopathic Pulmonary Fibrosis (IPF) is a progressive and devastating lung disease, characterized by progressive lung scarring. AREAS COVERED Prior to antifibrotic therapy (pirfenidone and nintedanib), there was no validated pharmaceutical therapy for IPF. Both antifibrotics can slow disease progression, however, IPF remains a detrimental disease with poor prognosis and treated survival rates of less than 7 years from diagnosis. Despite their effect the disease remains non-reversible and progressing whilst their side effect profile is often challenging. Treatment of comorbidities is also crucial. In this review, we discuss the current pharmacological management as well as management of comorbidities and symptoms. We also reviewed clinicaltrials.gov and summarised all the mid to late stage clinical trials (phase II and III) registered in IPF over the last 7 years and discuss the most promising drugs in clinical development. EXPERT OPINION Future for IPF management will need to focus on current unresolved issues. First a primary pathogenetic pathway has not been clearly identified. Future management may involve a combination of brushstroke approach with antifibrotics with targeted treatments for specific pathways in patient subsets following an 'oncological' approach. Another unmet need is management of exacerbations, which are deathly in most cases as well as either treating or preventing lung cancer.
Collapse
Affiliation(s)
- Athina Trachalaki
- The Margaret Turner Warwick Centre for Fibrosing Lung Diseases, Imperial College London National Heart and Lung Institute, Imperial College, London, UK
- Imperial College NHS Hospitals, London UK
| | | | - Athol Umfrey Wells
- Interstitial Lung Disease Unit, Royal Brompton & Harefield Hospitals, London, UK
- The Margaret Turner Warwick Centre for Fibrosing Lung Diseases, Imperial College London National Heart and Lung Institute, Imperial College, London, UK
- Imperial College NHS Hospitals, London UK
| |
Collapse
|
47
|
Han D, Gong H, Wei Y, Xu Y, Zhou X, Wang Z, Feng F. Hesperidin inhibits lung fibroblast senescence via IL-6/STAT3 signaling pathway to suppress pulmonary fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154680. [PMID: 36736168 DOI: 10.1016/j.phymed.2023.154680] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/19/2022] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and fatal lung disease with obscure pathogenesis. Increasing evidence suggests that cellular senescence is an important mechanism underlying in IPF. Clinical treatment with drugs, such as pirfenidone and nintedanib, reduces the risk of acute exacerbation and delays the decline of pulmonary function in patients with mild to moderate pulmonary fibrosis, and with adverse reactions. Hesperidin was previously shown to alleviate pulmonary fibrosis in rats by attenuating the inflammation response. Our previous research indicated that the Citrus alkaline extracts, hesperidin as the main active ingredient, could exert anti-pulmonary fibrosis effects by inhibiting the senescence of lung fibroblasts. However, whether hesperidin could ameliorate pulmonary fibrosis by inhibiting fibroblast senescence needed further study. PURPOSE This work aimed to investigate whether and how hesperidin can inhibit lung fibroblast senescence and thereby alleviate pulmonary fibrosis METHODS: Bleomycin was used to establish a mouse model of pulmonary fibrosis and doxorubicin was used to establish a model of cellular senescence in MRC-5 cells in vitro. The therapeutic effects of hesperidin on pulmonary fibrosis using haematoxylin-eosin staining, Masson staining, enzyme-linked immunosorbent assay, immunohistochemistry, western blotting and quantitative Real-Time PCR. The anti-senescent effect of hesperidin in vivo and in vitro was assessed by western blotting, quantitative Real-Time PCR and senescence-associated β-galactosidase RESULTS: We demonstrated that hesperidin could alleviate bleomycin-induced pulmonary fibrosis in mice. The expression level of senescence marker proteins p53, p21, and p16 was were downregulated, along with the myofibroblast marker α-SMA. The number of senescence-associated β-galactosidase-positive cells was significantly reduced by hesperidin intervention in vivo and in vitro. In addition, hesperidin could inhibit the IL6/STAT3 signaling pathway. Furthermore, suppression of the IL-6/STAT3 signaling pathway by pretreatment with the IL-6 inhibitor LMT-28 attenuating effect of hesperidin on fibroblast senescence in vitro. CONCLUSIONS These data illustrated that hesperidin may be potentially used in the treatment of IPF based on its ability to inhibit lung fibroblast senescence.
Collapse
Affiliation(s)
- Di Han
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Haiying Gong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China
| | - Yun Wei
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yong Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China; School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xianmei Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Zhichao Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Fanchao Feng
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital Of Chinese Medicine, Nanjing, China; Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| |
Collapse
|
48
|
Thong L, McElduff EJ, Henry MT. Trials and Treatments: An Update on Pharmacotherapy for Idiopathic Pulmonary Fibrosis. Life (Basel) 2023; 13:486. [PMID: 36836843 PMCID: PMC9963632 DOI: 10.3390/life13020486] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive fibrosing interstitial lung disease that occurs predominantly in the older population. There is increasing incidence and prevalence in IPF globally. The emergence of anti-fibrotic therapies in the last decade have improved patient survival though a cure is yet to be developed. In this review article, we aim to summarize the existing and novel pharmacotherapies for the treatment of IPF (excluding treatments for acute exacerbations), focusing on the current knowledge on the pathophysiology of the disease, mechanism of action of the drugs, and clinical trials.
Collapse
Affiliation(s)
- Lorraine Thong
- Department of Clinical Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Enda James McElduff
- Department of Clinical Medicine, Royal College of Surgeons Ireland, D02 YN77 Dublin, Ireland
| | - Michael Thomas Henry
- Department of Respiratory Medicine, Cork University Hospital, T12 YE02 Cork, Ireland
| |
Collapse
|
49
|
Behr J, Bonella F, Frye BC, Günther A, Hagmeyer L, Henes J, Klemm P, Koschel D, Kreuter M, Leuschner G, Nowak D, Prasse A, Quadder B, Sitter H, Costabel U. [Pharmacological treatment of idiopathic pulmonary fibrosis (update) and progressive pulmonary fibrosis - S2k Guideline of the German Respiratory Society]. Pneumologie 2023; 77:94-119. [PMID: 36791790 DOI: 10.1055/a-1983-6796] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Affiliation(s)
- Jürgen Behr
- Medizinische Klinik und Polklinik V, LMU Klinikum der Universität München, Mitglied des Deutschen Zentrums für Lungenforschung; Delegierte/r der DGP
| | - Francesco Bonella
- Zentrum für interstitielle und seltene Lungenerkrankungen, Klinik für Pneumologie, Ruhrlandklinik, Universitätsmedizin Essen; Delegierter der DGP
| | - Björn C Frye
- Klinik für Pneumologie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Deutschland; Delegierter der DGP
| | - Andreas Günther
- Center for Interstitial and Rare Lung Diseases, University Hospital Giessen Marburg, Giessen, Agaplesion Evangelisches Krankenhaus Mittelhessen, Giessen, Germany; Delegierter der DGP
| | - Lars Hagmeyer
- Krankenhaus Bethanien Solingen, Klinik für Pneumologie und Allergologie, Zentrum für Schlaf- und Beatmungsmedizin, Institut für Pneumologie an der Universität zu Köln; Delegierter der DGP
| | - Jörg Henes
- Zentrum für interdisziplinäre Rheumatologie, Immunologie und Autoimmunerkrankungen (INDIRA) und Innere Medizin II; Delegierter DGRh
| | - Philipp Klemm
- Abt. Rheumatologie und klinische Immunologie, Kerckhoff Klinik und Campus Kerckhoff der Justus-Liebig-Universität Gießen, Bad Nauheim; Delegierter der DGRh
| | - Dirk Koschel
- Fachkrankenhaus Coswig, Lungenzentrum und Medizinische Klinik 1, Universitätsklinik Carl Gustav Carus der TU Dresden; Delegierter der DGP
| | - Michael Kreuter
- Zentrum für interstitielle und seltene Lungenerkrankungen & interdisziplinäres Sarkoidosezentrum, Thoraxklinik, Universitätsklinikum Heidelberg, Deutsches Zentrum für Lungenforschung Heidelberg und Klinik für Pneumologie, Interdisziplinäres Lungenzentrum Ludwigsburg, RKH Klinik Ludwigsburg; Delegierter der DGIM
| | - Gabriela Leuschner
- Medizinische Klinik und Polklinik V, LMU Klinikum der Universität München, Mitglied des Deutschen Zentrums für Lungenforschung; Delegierte/r der DGP
| | - Dennis Nowak
- Institut und Poliklinik für Arbeits-, Sozial- und Umweltmedizin, LMU Klinikum der Universität München, Comprehensive Pneumology Center (CPC) München, Mitglied des Deutsches Zentrums für Lungenforschung; Delegierter der DGAUM
| | - Antje Prasse
- Klinik für Pneumologie und Infektiologie, Medizinische Hochschule Hannover, DZL BREATH und Abteilung für Fibroseforschung, Fraunhofer ITEM, Hannover, Delegierte der DGP
| | | | - Helmut Sitter
- Institut für Theoretische Chirurgie, Philipps-Universität Marburg, Moderator
| | - Ulrich Costabel
- Zentrum für interstitielle und seltene Lungenerkrankungen, Klinik für Pneumologie, Ruhrlandklinik, Universitätsmedizin Essen; Delegierter der DGP
| |
Collapse
|
50
|
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: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|