1
|
Zhang D, Eckhardt CM, McGroder C, Benesh S, Porcelli J, Depender C, Bogyo K, Westrich J, Thomas-Wilson A, Jobanputra V, Garcia CK. Clinical Impact of Telomere Length Testing for Interstitial Lung Disease. Chest 2024; 166:1071-1081. [PMID: 38950694 DOI: 10.1016/j.chest.2024.06.006] [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: 03/19/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Shortened telomere length (TL) is a genomic risk factor for fibrotic interstitial lung disease (ILD), but its role in clinical management is unknown. RESEARCH QUESTION What is the clinical impact of TL testing on the management of ILD? STUDY DESIGN AND METHODS Patients were evaluated in the Columbia University ILD clinic and underwent Clinical Laboratory Improvement Amendments-certified TL testing by flow cytometry and fluorescence in situ hybridization (FlowFISH) as part of clinical treatment. Short TL was defined as below the 10th age-adjusted percentile for either granulocytes or lymphocytes by FlowFISH. Patients were offered genetic counseling and testing if they had short TL or a family history of ILD. FlowFISH TL was compared with research quantitative polymerase chain reaction (qPCR) TL measurement. RESULTS A total of 108 patients underwent TL testing, including those with clinical features of short telomere syndrome such as familial pulmonary fibrosis (50%) or extrapulmonary manifestations in the patient (25%) or a relative (41%). The overall prevalence of short TL was 46% and was similar across clinical ILD diagnoses. The number of short telomere clinical features was independently associated with detecting short TL (OR, 2.00; 95% CI, 1.27-3.32). TL testing led to clinical treatment changes for 35 patients (32%), most commonly resulting in reduction or avoidance of immunosuppression. Of the patients who underwent genetic testing (n = 34), a positive or candidate diagnostic finding in telomere-related genes was identified in 10 patients (29%). Inclusion of TL testing below the 1st percentile helped reclassify eight of nine variants of uncertain significance into actionable findings. The quantitative polymerase chain reaction test correlated with FlowFISH, but age-adjusted percentile cutoffs may not be equivalent between the two assays. INTERPRETATION Incorporating TL testing in ILD impacted clinical management and led to the discovery of new actionable genetic variants.
Collapse
Affiliation(s)
- David Zhang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY.
| | | | - Claire McGroder
- Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Shannon Benesh
- Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | | | | | - Kelsie Bogyo
- Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Joseph Westrich
- Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | | | - Vaidehi Jobanputra
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY; New York Genome Center, New York, NY
| | - Christine K Garcia
- Department of Medicine, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
2
|
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 2024:1-29. [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
|
3
|
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
|
4
|
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: 0] [Impact Index Per Article: 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
|
5
|
Behr J, Salisbury ML, Walsh SLF, Podolanczuk AJ, Hariri LP, Hunninghake GM, Kolb M, Ryerson CJ, Cottin V, Beasley MB, Corte T, Glanville AR, Adegunsoye A, Hogaboam C, Wuyts WA, Noth I, Oldham JM, Richeldi L, Raghu G, Wells AU. The Role of Inflammation and Fibrosis in Interstitial Lung Disease Treatment Decisions. Am J Respir Crit Care Med 2024; 210:392-400. [PMID: 38484133 DOI: 10.1164/rccm.202401-0048pp] [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: 01/06/2023] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Affiliation(s)
- Juergen Behr
- Department of Medicine V, LMU University Hospital, LMU Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - Margaret L Salisbury
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Simon L F Walsh
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Anna J Podolanczuk
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Lida P Hariri
- Department of Pathology and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, and
| | - Gary M Hunninghake
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Martin Kolb
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare and McMaster University, Hamilton, Ontario, Canada
| | - Christopher J Ryerson
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vincent Cottin
- Department of Respiratory Medicine, National Reference Centre for Rare Pulmonary Diseases, ERN-LUNG, Louis Pradel Hospital, Hospices Civils de Lyon, UMR 754, INRAE, Claude Bernard University Lyon 1, Lyon, France
| | - Mary B Beasley
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Tamera Corte
- Royal Prince Alfred Hospital and
- University of Sydney, Sydney, New South Wales, Australia
| | | | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care, Department of Medicine, and
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Cory Hogaboam
- Women's Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Wim A Wuyts
- Unit for Interstitial Lung Diseases, Department of Respiratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Justin M Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Luca Richeldi
- Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ganesh Raghu
- Center for Interstitial Lung Diseases, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, and
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington; and
| | - Athol U Wells
- Royal Brompton Hospital and Imperial College, London, United Kingdom
| |
Collapse
|
6
|
Ng GYQ, Hande MP. Use of peptide nucleic acid probe to determine telomere dynamics in improving chromosome analysis in genetic toxicology studies. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2024; 897:503773. [PMID: 39054004 DOI: 10.1016/j.mrgentox.2024.503773] [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: 02/10/2024] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 07/27/2024]
Abstract
Genetic toxicology, strategically located at the intersection of genetics and toxicology, aims to demystify the complex interplay between exogenous agents and our genetic blueprint. Telomeres, the protective termini of chromosomes, play instrumental roles in cellular longevity and genetic stability. Traditionally karyotyping and fluorescence in situ hybridisation (FISH), have been indispensable tools for chromosomal analysis following exposure to genotoxic agents. However, their scope in discerning nuanced molecular dynamics is limited. Peptide Nucleic Acids (PNAs) are synthetic entities that embody characteristics of both proteins and nucleic acids and have emerged as potential game-changers. This perspective report comprehensively examines the vast potential of PNAs in genetic toxicology, with a specific emphasis on telomere research. PNAs' superior resolution and precision make them a favourable choice for genetic toxicological assessments. The integration of PNAs in contemporary analytical workflows heralds a promising evolution in genetic toxicology, potentially revolutionizing diagnostics, prognostics, and therapeutic avenues. In this timely review, we attempted to assess the limitations of current PNA-FISH methodology and recommend refinements.
Collapse
Affiliation(s)
- Gavin Yong Quan Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Manoor Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
7
|
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
|
8
|
Mullin ML, Fernandez G, Marinescu DC, Zheng B, Wong AW, Assayag D, Fisher JH, Johannson KA, Khalil N, Kolb M, Manganas H, Marcoux V, Morisset J, Min B, Farrand E, Ryerson CJ. Impact of Antigen Exposure on Outcomes and Treatment Response in Fibrotic Hypersensitivity Pneumonitis. Chest 2024; 165:1435-1443. [PMID: 38128609 DOI: 10.1016/j.chest.2023.12.019] [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: 10/06/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Patients with fibrotic hypersensitivity pneumonitis (fHP) are frequently treated with immunosuppression to slow lung function decline; however, the impact of this treatment has not been studied across different types of antigen exposure. RESEARCH QUESTION In patients with fHP, do disease outcomes and response to treatment vary by antigen type? STUDY DESIGN AND METHODS A multicenter interstitial lung disease database (Canadian Registry for Pulmonary Fibrosis) was used to identify patients with fHP. The causative antigen was categorized as avian, mold, unknown, or other. Treatment was defined as mycophenolate ≥ 1,000 mg/d or azathioprine ≥ 75 mg/d for ≥ 30 days. Statistical analysis included t tests, χ2 tests, and one-way analysis of variance. Unadjusted and adjusted competing risks and Cox proportional hazards models were used to assess survival. RESULTS A total of 344 patients were identified with the following causative antigens: avian (n = 93; 27%), mold (n = 88; 26%), other (n = 15; 4%), and unknown (n = 148; 43%). Patient characteristics and lung function were similar among antigen groups with a mean FVC % predicted of 75 ± 20. The percent of patients treated with immunosuppression was similar between antigens with 58% of patients treated. There was no change in lung function or symptom scores with the initiation of immunosuppression in the full cohort. Immunosuppression was not associated with a change in survival for patients with avian or mold antigen (avian: hazard ratio, 0.41; 95% CI, 0.11-1.59; P = .20; mold: hazard ratio, 1.13; 95% CI, 0.26-4.97; P = .88). For patients with unknown causative antigen, survival was worse when treated with immunosuppression (hazard ratio, 2.65; 95% CI, 1.01-6.92; P = .047). INTERPRETATION Response to immunosuppression varies by antigen type in patients with fHP. Additional studies are needed to test the role of immunosuppression in fHP, and particularly in those with an unknown antigen.
Collapse
Affiliation(s)
- Monica L Mullin
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gustavo Fernandez
- Luis Razetti School of Medicine, Central University of Venezuela, Caracas, Venezuela
| | - Daniel-Costin Marinescu
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada; Centre for Lung Health, Vancouver General Hospital, Vancouver, BC, Canada
| | - Boyang Zheng
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Alyson W Wong
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Deborah Assayag
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Jolene H Fisher
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Nasreen Khalil
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Martin Kolb
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Helene Manganas
- Département de Médecine, Centre de recherche du Centre hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Veronica Marcoux
- Department of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Julie Morisset
- Département de Médecine, Centre de recherche du Centre hospitalier de l'Université de Montréal, Montreal, QC, Canada
| | - Bohyung Min
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Erica Farrand
- Department of Medicine, University California San Francisco, San Francisco, CA
| | - Christopher J Ryerson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada; Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada.
| |
Collapse
|
9
|
Hurley K, Ozaki M, Philippot Q, Galvin L, Crosby D, Kirwan M, Gill DR, Alysandratos KD, Jenkins G, Griese M, Nathan N, Borie R. A roadmap to precision treatments for familial pulmonary fibrosis. EBioMedicine 2024; 104:105135. [PMID: 38718684 PMCID: PMC11096859 DOI: 10.1016/j.ebiom.2024.105135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024] Open
Abstract
Interstitial lung diseases (ILDs) in adults and children (chILD) are a heterogeneous group of lung disorders leading to inflammation, abnormal tissue repair and scarring of the lung parenchyma often resulting in respiratory failure and death. Inherited factors directly cause, or contribute significantly to the risk of developing ILD, so called familial pulmonary fibrosis (FPF), and monogenic forms may have a poor prognosis and respond poorly to current treatments. Specific, variant-targeted or precision treatments are lacking. Clinical trials of repurposed drugs, anti-fibrotic medications and specific treatments are emerging but for many patients no interventions exist. We convened an expert working group to develop an overarching framework to address the existing research gaps in basic, translational, and clinical research and identified areas for future development of preclinical models, candidate medications and innovative clinical trials. In this Position Paper, we summarise working group discussions, recommendations, and unresolved questions concerning precision treatments for FPF.
Collapse
Affiliation(s)
- Killian Hurley
- Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland; Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| | - Mari Ozaki
- Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland; Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Quentin Philippot
- Université Paris Cité, Inserm, PHERE, Hôpital Bichat, AP-HP, Service de Pneumologie A, Centre Constitutif du Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France; Physiopathology and Epidemiology of Respiratory Diseases, Inserm U1152, UFR de Médecine, Université Paris Cité, 75018, Paris, France
| | - Liam Galvin
- European Pulmonary Fibrosis Federation, Overijse, Belgium
| | | | - Mary Kirwan
- Department of General Practice, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Deborah R Gill
- UK Respiratory Gene Therapy Consortium, London, United Kingdom; Gene Medicine Research Group, Radcliffe Department of Medicine (NDCLS), University of Oxford, Oxford, United Kingdom
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Gisli Jenkins
- Imperial College London, 4615, National Heart & Lung Institute, London, United Kingdom of Great Britain and Northern Ireland
| | - Matthias Griese
- Department of Pediatric Pneumology, German Center for Lung Research (DZL), Dr von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Nadia Nathan
- Sorbonne Université, Pediatric Pulmonology and Reference Center for Rare Lung Diseases RespiRare, Inserm U933 Laboratory of Childhood Genetic Diseases, Armand Trousseau Hospital, APHP, Paris, France
| | - Raphael Borie
- Université Paris Cité, Inserm, PHERE, Hôpital Bichat, AP-HP, Service de Pneumologie A, Centre Constitutif du Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| |
Collapse
|
10
|
Yao F, Xu M, Dong L, Shen X, Shen Y, Jiang Y, Zhu T, Zhang C, Yu G. Sinomenine attenuates pulmonary fibrosis by downregulating TGF-β1/Smad3, PI3K/Akt and NF-κB signaling pathways. BMC Pulm Med 2024; 24:229. [PMID: 38730387 PMCID: PMC11088103 DOI: 10.1186/s12890-024-03050-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/07/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Since COVID-19 became a global epidemic disease in 2019, pulmonary fibrosis (PF) has become more prevalent among persons with severe infections, with IPF being the most prevalent form. In traditional Chinese medicine, various disorders are treated using Sinomenine (SIN). The SIN's strategy for PF defense is unclear. METHODS Bleomycin (BLM) was used to induce PF, after which inflammatory factors, lung histological alterations, and the TGF-/Smad signaling pathway were assessed. By administering various dosages of SIN and the TGF- receptor inhibitor SB-431,542 to human embryonic lung fibroblasts (HFL-1) and A549 cells, we were able to examine proliferation and migration as well as the signaling molecules implicated in Epithelial-Mesenchymal Transition (EMT) and Extra-Cellular Matrix (ECM). RESULTS In vivo, SIN reduced the pathological changes in the lung tissue induced by BLM, reduced the abnormal expression of inflammatory cytokines, and improved the weight and survival rate of mice. In vitro, SIN inhibited the migration and proliferation by inhibiting TGF-β1/Smad3, PI3K/Akt, and NF-κB pathways, prevented the myofibroblasts (FMT) of HFL-1, reversed the EMT of A549 cells, restored the balance of matrix metalloenzymes, and reduced the expression of ECM proteins. CONCLUSION SIN attenuated PF by down-regulating TGF-β/Smad3, PI3K/Akt, and NF-κB signaling pathways, being a potential effective drug in the treatment of PF.
Collapse
Affiliation(s)
- Fuqiang Yao
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Minghao Xu
- School of Medicine, ShaoXing University, Shaoxing, Zhejiang, China
| | - Lingjun Dong
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Xiao Shen
- School of Medicine, ShaoXing University, Shaoxing, Zhejiang, China
| | - Yujie Shen
- School of Medicine, ShaoXing University, Shaoxing, Zhejiang, China
| | - Yisheng Jiang
- School of Medicine, ShaoXing University, Shaoxing, Zhejiang, China
| | - Ting Zhu
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Chu Zhang
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Guangmao Yu
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China.
| |
Collapse
|
11
|
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
|
12
|
Joerns EK, Sparks JA. Interstitial Pneumonia with Autoimmune Features: Aiming to Define, Refine, and Treat. REVISTA COLOMBIANA DE REUMATOLOGIA 2024; 31:S45-S53. [PMID: 39399289 PMCID: PMC11469586 DOI: 10.1016/j.rcreu.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Interstitial pneumonia with autoimmune features (IPAF) was defined for research purposes as interstitial lung disease (ILD) associated with features of autoimmunity without diagnosed rheumatic disease. Since publication of the IPAF criteria in 2015, there have been multiple studies of IPAF. However, much remains unknown regarding pathogenesis, prognosis, and treatment in IPAF. This narrative review details the history and classification of IPAF, lists challenges associated with classifying patients as IPAF, and explores the prevalence, epidemiology and presentation of IPAF. We also examine prognosis and important features determining IPAF clinical course, outline pathogenesis, and review treatment strategies.
Collapse
Affiliation(s)
- Elena K Joerns
- Department of Internal Medicine, Division of Rheumatic Diseases, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Jeffrey A Sparks
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Alrehaili G, Kemppainen J, Kalra S, Pinto E Vairo F, Moua T, Yi ES, Ferrer A, Patnaik MM, Carmona EM. Genetic Testing Goes Beyond Imaging and Histological Evaluation in Pleuroparenchymal Fibroelastosis. Lung 2024; 202:151-156. [PMID: 38461429 PMCID: PMC11009725 DOI: 10.1007/s00408-024-00685-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/17/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Lung biopsy remains the gold standard in the diagnosis of fibrotic interstitial lung disease (F-ILD), but there is a growing appreciation of the role of pathogenic gene variants in telomere and surfactant protein genes, especially in familial pulmonary fibrosis (FPF). Pleuroparenchymal fibroelastosis (PPFE) is a rare disease that can coexist with different patterns of F-ILD, including FPF. It can be progressive and often leads to respiratory failure and death. This study tested the hypothesis that genetic testing goes beyond radiological and histological findings in PPFE and other F-ILD further informing clinical decision-making for patients and affected family members by identifying pathological gene variants in telomere and surfactant protein genes. METHODS This is a retrospective review of 70 patients with F-ILD in the setting of FPF or premature lung fibrosis. Six out of 70 patients were diagnosed with PPFE based on radiological or histological characteristics. All patients underwent telomere length evaluation in peripheral blood by Flow-FISH or genetic testing using a customized exome-based panel that included telomere and surfactant protein genes associated with lung fibrosis. RESULTS Herein, we identified six individuals where radiographic or histopathological analyses of PPFE were linked with telomere biology disorders (TBD) or variants in surfactant protein genes. Each case involved individuals with either personal early-onset lung fibrosis or a family history of the disease. Assessments of telomere length and genetic testing offered insights beyond traditional radiological and histopathological evaluations. CONCLUSION Detecting anomalies in TBD-related or surfactant protein genes can significantly refine the diagnosis and treatment strategies for individuals with PPFE and other F-ILD.
Collapse
Affiliation(s)
- Ghadah Alrehaili
- Department of Pulmonary and Critical Care, Mayo Clinic, Rochester, MN, USA
| | - Jennifer Kemppainen
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Sanjay Kalra
- Department of Pulmonary and Critical Care, Mayo Clinic, Rochester, MN, USA
| | - Filippo Pinto E Vairo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
- Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | - Teng Moua
- Department of Pulmonary and Critical Care, Mayo Clinic, Rochester, MN, USA
| | - Eunhee S Yi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Alejandro Ferrer
- Hematology/Oncology Departments, Mayo Clinic, Rochester, MN, USA
| | - Mrinal M Patnaik
- Hematology/Oncology Departments, Mayo Clinic, Rochester, MN, USA
| | - Eva M Carmona
- Department of Pulmonary and Critical Care, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
14
|
Hu A, Patnaik MM, Kelm DJ. A Woman with Progressive Cough, Dyspnea, and Early Graying of Her Hair. Ann Am Thorac Soc 2024; 21:499-503. [PMID: 38426822 DOI: 10.1513/annalsats.202304-336cc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Affiliation(s)
- Alan Hu
- Department of Internal Medicine
| | | | - Diana J Kelm
- Department of Internal Medicine
- Division of Pulmonary and Critical Care, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW There has been a rapid increase in silicosis cases, particularly related to artificial stone. The key to management is avoidance of silica exposure. Despite this, many develop progressive disease and there are no routinely recommended treatments. This review provides a summary of the literature pertaining to pharmacological therapies for silicosis and examines the plausibility of success of such treatments given the disease pathogenesis. RECENT FINDINGS In-vitro and in-vivo models demonstrate potential efficacy for drugs, which target inflammasomes, cytokines, effector cells, fibrosis, autophagy, and oxidation. SUMMARY There is some evidence for potential therapeutic targets in silicosis but limited translation into human studies. Treatment of silicosis likely requires a multimodal approach, and there is considerable cross-talk between pathways; agents that modulate both inflammation, fibrosis, autophagy, and ROS production are likely to be most efficacious.
Collapse
Affiliation(s)
- Hayley Barnes
- Monash Centre for Occupational and Environmental Health, Monash University
- Department of Respiratory Medicine, Alfred Health
- Central Clinical School, Monash University, Melbourne
| | - Maggie Lam
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Ryan Hoy
- Monash Centre for Occupational and Environmental Health, Monash University
- Department of Respiratory Medicine, Alfred Health
| |
Collapse
|
16
|
Mackintosh JA, Keir G, Troy LK, Holland AE, Grainge C, Chambers DC, Sandford D, Jo HE, Glaspole I, Wilsher M, Goh NSL, Reynolds PN, Chapman S, Mutsaers SE, de Boer S, Webster S, Moodley Y, Corte TJ. Treatment of idiopathic pulmonary fibrosis and progressive pulmonary fibrosis: A position statement from the Thoracic Society of Australia and New Zealand 2023 revision. Respirology 2024; 29:105-135. [PMID: 38211978 PMCID: PMC10952210 DOI: 10.1111/resp.14656] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 12/11/2023] [Indexed: 01/13/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease leading to significant morbidity and mortality. In 2017 the Thoracic Society of Australia and New Zealand (TSANZ) and Lung Foundation Australia (LFA) published a position statement on the treatment of IPF. Since that time, subsidized anti-fibrotic therapy in the form of pirfenidone and nintedanib is now available in both Australia and New Zealand. More recently, evidence has been published in support of nintedanib for non-IPF progressive pulmonary fibrosis (PPF). Additionally, there have been numerous publications relating to the non-pharmacologic management of IPF and PPF. This 2023 update to the position statement for treatment of IPF summarizes developments since 2017 and reaffirms the importance of a multi-faceted approach to the management of IPF and progressive pulmonary fibrosis.
Collapse
Affiliation(s)
- John A. Mackintosh
- Department of Respiratory MedicineThe Prince Charles HospitalBrisbaneQueenslandAustralia
- Centre of Research Excellence in Pulmonary FibrosisCamperdownNew South WalesAustralia
| | - Gregory Keir
- Department of Respiratory MedicinePrincess Alexandra HospitalBrisbaneQueenslandAustralia
| | - Lauren K. Troy
- Department of Respiratory and Sleep MedicineRoyal Prince Alfred HospitalCamperdownNew South WalesAustralia
- University of SydneySydneyNew South WalesAustralia
| | - Anne E. Holland
- Centre of Research Excellence in Pulmonary FibrosisCamperdownNew South WalesAustralia
- Department of PhysiotherapyThe Alfred HospitalMelbourneVictoriaAustralia
- Department of Respiratory Research@AlfredCentral Clinical School, Monash UniversityMelbourneVictoriaAustralia
| | - Christopher Grainge
- Department of Respiratory MedicineJohn Hunter HospitalNewcastleNew South WalesAustralia
| | - Daniel C. Chambers
- Department of Respiratory MedicineThe Prince Charles HospitalBrisbaneQueenslandAustralia
- Centre of Research Excellence in Pulmonary FibrosisCamperdownNew South WalesAustralia
| | - Debra Sandford
- Centre of Research Excellence in Pulmonary FibrosisCamperdownNew South WalesAustralia
- Department of Thoracic MedicineCentral Adelaide Local Health NetworkAdelaideSouth AustraliaAustralia
- University of AdelaideAdelaideSouth AustraliaAustralia
| | - Helen E. Jo
- Department of Respiratory and Sleep MedicineRoyal Prince Alfred HospitalCamperdownNew South WalesAustralia
- University of SydneySydneyNew South WalesAustralia
| | - Ian Glaspole
- Centre of Research Excellence in Pulmonary FibrosisCamperdownNew South WalesAustralia
- Department of Respiratory MedicineThe Alfred HospitalMelbourneVictoriaAustralia
| | - Margaret Wilsher
- Department of Respiratory MedicineTe Toka Tumai AucklandAucklandNew Zealand
| | - Nicole S. L. Goh
- Department of Respiratory MedicineAustin HospitalMelbourneVictoriaAustralia
- Institute for Breathing and SleepMelbourneVictoriaAustralia
- University of MelbourneMelbourneVictoriaAustralia
| | - Paul N. Reynolds
- Centre of Research Excellence in Pulmonary FibrosisCamperdownNew South WalesAustralia
- Department of Thoracic MedicineCentral Adelaide Local Health NetworkAdelaideSouth AustraliaAustralia
- University of AdelaideAdelaideSouth AustraliaAustralia
| | - Sally Chapman
- Institute for Respiratory Health, University of Western AustraliaNedlandsWestern AustraliaAustralia
| | - Steven E. Mutsaers
- Department of Respiratory MedicineFiona Stanley HospitalMurdochWestern AustraliaAustralia
| | - Sally de Boer
- Department of Respiratory MedicineTe Toka Tumai AucklandAucklandNew Zealand
| | - Susanne Webster
- Department of Respiratory and Sleep MedicineRoyal Prince Alfred HospitalCamperdownNew South WalesAustralia
| | - Yuben Moodley
- Centre of Research Excellence in Pulmonary FibrosisCamperdownNew South WalesAustralia
- Institute for Respiratory Health, University of Western AustraliaNedlandsWestern AustraliaAustralia
- Department of Respiratory MedicineFiona Stanley HospitalMurdochWestern AustraliaAustralia
| | - Tamera J. Corte
- Centre of Research Excellence in Pulmonary FibrosisCamperdownNew South WalesAustralia
- Department of Respiratory and Sleep MedicineRoyal Prince Alfred HospitalCamperdownNew South WalesAustralia
- University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
17
|
Kou N, Chen YB, Li XW, Xu D, Wang Y, Dong XR, Cui YL, Wang Q. Pulmonary administration of tetrandrine loaded Zinc-Alginate nanogels attenuates pulmonary fibrosis in rats. Int J Pharm 2024; 649:123625. [PMID: 37984618 DOI: 10.1016/j.ijpharm.2023.123625] [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: 09/12/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Pulmonary fibrosis is a chronic and progressive disease, current systemic administration is not fully effective with many side effects, such as gastrointestinal and liver injury. The pulmonary delivery system for pulmonary fibrosis may contribute to maximize therapeutic benefit. Natural compounds might have prominence as potential drug candidates, but the low bioavailabilities affect their clinical use. Tetrandrine is a natural alkaloid with good anti-inflammatory, antifibrogenetic and antioxidant effects, and it is used as a clinical therapeutic drug for the treatment of silicosis in China. In the present study, we explore a new strategy of pulmonary delivery system to improve low solubility and pesticide effect of tetrandrine. Tetrandrine was loaded into alginate nanogels by reverse microemulsion method. The release behavior of tetrandrine reached zero-order kinetics release and the maximum free radical clearance rates reached up to 90%. The pulmonary fibrosis rats were treated with tetrandrine nanogels by using ultrasonic atomizing inhalation. Tetrandrine nanogels decreased the development and progression of fibrosis by reducing inflammation response and bating the deposition of extra cellular matrix. In conclusion, ultrasonic atomizing inhalation of tetrandrine nanogels provided a new therapeutic strategy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Na Kou
- State Key Laboratory of Component-Based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yi-Bing Chen
- State Key Laboratory of Component-Based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xian-Wen Li
- State Key Laboratory of Component-Based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730050, China
| | - Dong Xu
- State Key Laboratory of Component-Based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Wang
- State Key Laboratory of Component-Based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xin-Ran Dong
- State Key Laboratory of Component-Based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuan-Lu Cui
- State Key Laboratory of Component-Based Chinese Medicine, Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Qiangsong Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Engineering Research Center of Pulmonary and Critical Care Medicine Technology and Device (Ministry of Education), Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300192, China.
| |
Collapse
|
18
|
Mackintosh JA, Chambers DC. Telomere length and immunosuppression in non-idiopathic pulmonary fibrosis interstitial lung disease. Eur Respir J 2024; 63:2301806. [PMID: 38237995 DOI: 10.1183/13993003.01806-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 11/02/2023] [Indexed: 01/23/2024]
Affiliation(s)
- John A Mackintosh
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
- School of Medicine, University of Queensland, Brisbane, Australia
| | - Daniel C Chambers
- Queensland Lung Transplant Service, Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
- School of Medicine, University of Queensland, Brisbane, Australia
| |
Collapse
|
19
|
Zhang D, Adegunsoye A, Oldham JM, Wolters PJ, Garcia CK, Newton CA. Reply: Telomere length and immunosuppression in non-idiopathic pulmonary fibrosis interstitial lung disease. Eur Respir J 2024; 63:2302146. [PMID: 38237997 PMCID: PMC11002962 DOI: 10.1183/13993003.02146-2023] [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: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024]
Affiliation(s)
- David Zhang
- Division of Pulmonary and Critical Care Medicine, Columbia University, New York, NY, USA
| | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care, University of Chicago, Chicago, IL, USA
| | - Justin M Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christine Kim Garcia
- Division of Pulmonary and Critical Care Medicine, Columbia University, New York, NY, USA
| | - Chad A Newton
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
20
|
Pradère P, Zajacova A, Bos S, Le Pavec J, Fisher A. Molecular monitoring of lung allograft health: is it ready for routine clinical use? Eur Respir Rev 2023; 32:230125. [PMID: 37993125 PMCID: PMC10663940 DOI: 10.1183/16000617.0125-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/16/2023] [Indexed: 11/24/2023] Open
Abstract
Maintenance of long-term lung allograft health in lung transplant recipients (LTRs) requires a fine balancing act between providing sufficient immunosuppression to reduce the risk of rejection whilst at the same time not over-immunosuppressing individuals and exposing them to the myriad of immunosuppressant drug side-effects that can cause morbidity and mortality. At present, lung transplant physicians only have limited and rather blunt tools available to assist them with this task. Although therapeutic drug monitoring provides clinically useful information about single time point and longitudinal exposure of LTRs to immunosuppressants, it lacks precision in determining the functional level of immunosuppression that an individual is experiencing. There is a significant gap in our ability to monitor lung allograft health and therefore tailor optimal personalised immunosuppression regimens. Molecular diagnostics performed on blood, bronchoalveolar lavage or lung tissue that can detect early signs of subclinical allograft injury, differentiate rejection from infection or distinguish cellular from humoral rejection could offer clinicians powerful tools in protecting lung allograft health. In this review, we look at the current evidence behind molecular monitoring in lung transplantation and ask if it is ready for routine clinical use. Although donor-derived cell-free DNA and tissue transcriptomics appear to be the techniques with the most immediate clinical potential, more robust data are required on their performance and additional clinical value beyond standard of care.
Collapse
Affiliation(s)
- Pauline Pradère
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
- Department of Respiratory Diseases, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph and Paris Saclay University, Paris, France
| | - Andrea Zajacova
- Prague Lung Transplant Program, Department of Pneumology, Motol University Hospital and 2nd Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Saskia Bos
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
- Institute of Transplantation, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| | - Jérôme Le Pavec
- Department of Respiratory Diseases, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph and Paris Saclay University, Paris, France
| | - Andrew Fisher
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
- Institute of Transplantation, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| |
Collapse
|
21
|
Del Valle KT, Carmona EM. Diagnosis and Management of Pulmonary Manifestations of Telomere Biology Disorders. Curr Hematol Malig Rep 2023:10.1007/s11899-023-00720-9. [PMID: 38159192 DOI: 10.1007/s11899-023-00720-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE OF REVIEW Telomere biology disorders (TBD) are a group of genetic disorders characterized by premature shortening of telomeres, resulting in accelerated aging of somatic cells. This often leads to major multisystem organ dysfunction, and TBDs have become increasingly recognized as a significant contributor to numerous disease processes within the past 10-15 years. Both research and clinical practice in this field are rapidly evolving. RECENT FINDINGS A subset of patients with TBD suffers from interstitial lung disease, most commonly pulmonary fibrosis. Often, the clinical presentation is indistinguishable from other forms of lung fibrosis. There are no pathognomonic radiographic or histological features, and a high level of suspicion is therefore required. Telomere evaluation is thus crucial to establishing the diagnosis. This review details the clinical presentation, objective evaluation, indicated genetic testing, and recommended management strategies for patients affected by interstitial lung disease associated with TBDs. Our goal is to empower pulmonologists and other healthcare professionals who care for these patients to provide appropriate and personalized care for this population.
Collapse
Affiliation(s)
- Kathryn T Del Valle
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Eva M Carmona
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
22
|
Tesolato S, Vicente-Valor J, Jarabo JR, Calatayud J, Sáiz-Pardo M, Nieto A, Álvaro-Álvarez D, Linares MJ, Fraile CA, Hernándo F, Iniesta P, Gómez-Martínez AM. Role of Telomere Length in Survival of Patients with Idiopathic Pulmonary Fibrosis and Other Interstitial Lung Diseases. Biomedicines 2023; 11:3257. [PMID: 38137478 PMCID: PMC10741059 DOI: 10.3390/biomedicines11123257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Interstitial lung diseases (ILDs) constitute a group of more than 200 disorders, with idiopathic pulmonary fibrosis (IPF) being one of the most frequent. Telomere length (TL) shortening causes loss of function of the lung parenchyma. However, little is known about its role as a prognostic factor in ILD patients. With the aim of investigating the role of TL and telomerase activity in the prognosis of patients affected by ILDs, we analysed lung tissue samples from 61 patients. We measured relative TL and telomerase activity by conventional procedures. Both clinical and molecular parameters were associated with overall survival by the Kaplan-Meier method. Patients with IPF had poorer prognosis than patients with other ILDs (p = 0.034). When patients were classified according to TL, those with shortened telomeres reported lower overall survival (p = 0.085); differences reached statistical significance after excluding ILD patients who developed cancer (p = 0.021). In a Cox regression analysis, TL behaved as a risk-modifying variable for death associated with rheumatic disease (RD) co-occurrence (p = 0.029). Also, in patients without cancer, ferritin was significantly increased in cases with RD and IPF co-occurrence (p = 0.032). In relation to telomerase activity, no significant differences were detected. In conclusion, TL in lung tissue emerges as a prognostic factor in ILD patients. Specifically, in cases with RD and IPF co-occurrence, TL can be considered as a risk-modifying variable for death.
Collapse
Affiliation(s)
- Sofía Tesolato
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University, Ramón y Cajal Sq. (University City), 28040 Madrid, Spain; (S.T.); (J.V.-V.)
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (J.-R.J.); (J.C.); (C.-A.F.); (F.H.); (A.-M.G.-M.)
| | - Juan Vicente-Valor
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University, Ramón y Cajal Sq. (University City), 28040 Madrid, Spain; (S.T.); (J.V.-V.)
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (J.-R.J.); (J.C.); (C.-A.F.); (F.H.); (A.-M.G.-M.)
| | - Jose-Ramón Jarabo
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (J.-R.J.); (J.C.); (C.-A.F.); (F.H.); (A.-M.G.-M.)
- Department of Surgery, Faculty of Medicine, Complutense University, Ramón y Cajal Sq. (University City), 28040 Madrid, Spain
- Thoracic Surgery Service of the San Carlos Hospital, 28040 Madrid, Spain
| | - Joaquín Calatayud
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (J.-R.J.); (J.C.); (C.-A.F.); (F.H.); (A.-M.G.-M.)
- Department of Surgery, Faculty of Medicine, Complutense University, Ramón y Cajal Sq. (University City), 28040 Madrid, Spain
- Thoracic Surgery Service of the San Carlos Hospital, 28040 Madrid, Spain
| | - Melchor Sáiz-Pardo
- Pathological Anatomy Service of the San Carlos Hospital, 28040 Madrid, Spain;
| | - Asunción Nieto
- Pulmonology Service of the San Carlos Hospital, 28040 Madrid, Spain;
| | | | - María-Jesús Linares
- Pulmonology Service of Alcorcon Foundation University Hospital, 28922 Madrid, Spain;
| | - Carlos-Alfredo Fraile
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (J.-R.J.); (J.C.); (C.-A.F.); (F.H.); (A.-M.G.-M.)
- Department of Surgery, Faculty of Medicine, Complutense University, Ramón y Cajal Sq. (University City), 28040 Madrid, Spain
- Thoracic Surgery Service of the San Carlos Hospital, 28040 Madrid, Spain
| | - Florentino Hernándo
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (J.-R.J.); (J.C.); (C.-A.F.); (F.H.); (A.-M.G.-M.)
- Department of Surgery, Faculty of Medicine, Complutense University, Ramón y Cajal Sq. (University City), 28040 Madrid, Spain
- Thoracic Surgery Service of the San Carlos Hospital, 28040 Madrid, Spain
| | - Pilar Iniesta
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University, Ramón y Cajal Sq. (University City), 28040 Madrid, Spain; (S.T.); (J.V.-V.)
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (J.-R.J.); (J.C.); (C.-A.F.); (F.H.); (A.-M.G.-M.)
| | - Ana-María Gómez-Martínez
- San Carlos Health Research Institute (IdISSC), 28040 Madrid, Spain; (J.-R.J.); (J.C.); (C.-A.F.); (F.H.); (A.-M.G.-M.)
- Department of Surgery, Faculty of Medicine, Complutense University, Ramón y Cajal Sq. (University City), 28040 Madrid, Spain
- Thoracic Surgery Service of the San Carlos Hospital, 28040 Madrid, Spain
| |
Collapse
|
23
|
Banaszak LG, Smith-Simmer K, Shoger K, Lovrien L, Malik A, Sandbo N, Sultan S, Guzy R, Lowery EM, Churpek JE. Implementation of a prospective screening strategy to identify adults with a telomere biology disorder among those undergoing lung transplant evaluation for interstitial lung disease. Respir Med 2023; 220:107464. [PMID: 37951311 DOI: 10.1016/j.rmed.2023.107464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 11/13/2023]
Abstract
INTRODUCTION Patients with interstitial lung disease (ILD) secondary to telomere biology disorders (TBD) experience increased morbidity after lung transplantation. Identifying patients with TBD may allow for personalized management to facilitate better outcomes. However, establishing a TBD diagnosis in adults is challenging. METHODS A TBD screening questionnaire was introduced prospectively into the lung transplant evaluation. Patients with ILD screening positive were referred for comprehensive TBD phenotyping and concurrent telomere length measurement and germline genetic testing. RESULTS Of 98 patients, 32 (33%) screened positive. Eight patients (8% of total; 25% of patients with a positive screen) met strict TBD diagnostic criteria, requiring either critically short lymphocyte telomeres (<1st percentile) (n = 4), a pathogenic variant in a TBD-associated gene (n = 1), or both (n = 3) along with a TBD clinical phenotype. Additional patients not meeting strict diagnostic criteria had histories consistent with TBD along with telomere lengths <10th percentile and/or rare variants in TBD-associated genes, highlighting a critical need to refine TBD diagnostic criteria for this patient population. CONCLUSION A TBD phenotype screening questionnaire in patients with ILD undergoing lung transplant evaluation has a diagnostic yield of 25%. Additional gene discovery, rare variant functional testing, and refined TBD diagnostic criteria are needed to realize the maximum benefit of testing for TBD in patients undergoing lung transplantation.
Collapse
Affiliation(s)
- Lauren G Banaszak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Kelcy Smith-Simmer
- Oncology Genetics, University of Wisconsin Carbone Cancer Center, UW Health, Madison, WI, 53705, USA
| | - Kyle Shoger
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Lauren Lovrien
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Amy Malik
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Nathan Sandbo
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Samir Sultan
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Robert Guzy
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Erin M Lowery
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jane E Churpek
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
24
|
Zhang D, Adegunsoye A, Oldham JM, Kozlitina J, Garcia N, Poonawalla M, Strykowski R, Linderholm AL, Ley B, Ma SF, Noth I, Strek ME, Wolters PJ, Garcia CK, Newton CA. Telomere length and immunosuppression in non-idiopathic pulmonary fibrosis interstitial lung disease. Eur Respir J 2023; 62:2300441. [PMID: 37591536 PMCID: PMC10695771 DOI: 10.1183/13993003.00441-2023] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Studies suggest a harmful pharmacogenomic interaction exists between short leukocyte telomere length (LTL) and immunosuppressants in idiopathic pulmonary fibrosis (IPF). It remains unknown if a similar interaction exists in non-IPF interstitial lung disease (ILD). METHODS A retrospective, multicentre cohort analysis was performed in fibrotic hypersensitivity pneumonitis (fHP), unclassifiable ILD (uILD) and connective tissue disease (CTD)-ILD patients from five centres. LTL was measured by quantitative PCR for discovery and replication cohorts and expressed as age-adjusted percentiles of normal. Inverse probability of treatment weights based on propensity scores were used to assess the association between mycophenolate or azathioprine exposure and age-adjusted LTL on 2-year transplant-free survival using weighted Cox proportional hazards regression incorporating time-dependent immunosuppressant exposure. RESULTS The discovery and replication cohorts included 613 and 325 patients, respectively. In total, 40% of patients were exposed to immunosuppression and 22% had LTL <10th percentile of normal. fHP and uILD patients with LTL <10th percentile experienced reduced survival when exposed to either mycophenolate or azathioprine in the discovery cohort (mortality hazard ratio (HR) 4.97, 95% CI 2.26-10.92; p<0.001) and replication cohort (mortality HR 4.90, 95% CI 1.74-13.77; p=0.003). Immunosuppressant exposure was not associated with differential survival in patients with LTL ≥10th percentile. There was a significant interaction between LTL <10th percentile and immunosuppressant exposure (discovery pinteraction=0.013; replication pinteraction=0.011). Low event rate and prevalence of LTL <10th percentile precluded subgroup analyses for CTD-ILD. CONCLUSION Similar to IPF, fHP and uILD patients with age-adjusted LTL <10th percentile may experience reduced survival when exposed to immunosuppression.
Collapse
Affiliation(s)
- David Zhang
- Division of Pulmonary and Critical Care Medicine, Columbia University, New York, NY, USA
- These two authors contributed equally to this work
| | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, USA
- These two authors contributed equally to this work
| | - Justin M Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nicole Garcia
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Maria Poonawalla
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Rachel Strykowski
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Angela L Linderholm
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California Davis, Sacramento, CA, USA
| | - Brett Ley
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Shwu-Fan Ma
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA, USA
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA, USA
| | - Mary E Strek
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christine Kim Garcia
- Division of Pulmonary and Critical Care Medicine, Columbia University, New York, NY, USA
| | - Chad A Newton
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
25
|
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
|
26
|
Amati F, Spagnolo P, Ryerson CJ, Oldham JM, Gramegna A, Stainer A, Mantero M, Sverzellati N, Lacedonia D, Richeldi L, Blasi F, Aliberti S. Walking the path of treatable traits in interstitial lung diseases. Respir Res 2023; 24:251. [PMID: 37872563 PMCID: PMC10594881 DOI: 10.1186/s12931-023-02554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/05/2023] [Indexed: 10/25/2023] Open
Abstract
Interstitial lung diseases (ILDs) are complex and heterogeneous diseases. The use of traditional diagnostic classification in ILD can lead to suboptimal management, which is worsened by not considering the molecular pathways, biological complexity, and disease phenotypes. The identification of specific "treatable traits" in ILDs, which are clinically relevant and modifiable disease characteristics, may improve patient's outcomes. Treatable traits in ILDs may be classified into four different domains (pulmonary, aetiological, comorbidities, and lifestyle), which will facilitate identification of related assessment tools, treatment options, and expected benefits. A multidisciplinary care team model is a potential way to implement a "treatable traits" strategy into clinical practice with the aim of improving patients' outcomes. Multidisciplinary models of care, international registries, and the use of artificial intelligence may facilitate the implementation of the "treatable traits" approach into clinical practice. Prospective studies are needed to test potential therapies for a variety of treatable traits to further advance care of patients with ILD.
Collapse
Affiliation(s)
- Francesco Amati
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padua, Italy
| | - Christopher J Ryerson
- Department of Medicine, University of British Columbia and Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Justin M Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrea Gramegna
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Anna Stainer
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Marco Mantero
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Nicola Sverzellati
- Unit of Scienze Radiologiche, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Donato Lacedonia
- Department of Medical and Occupational Sciences, Institute of Respiratory Disease, Università degli Studi di Foggia, Foggia, Italy
| | - Luca Richeldi
- Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Blasi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Respiratory Unit and Cystic Fibrosis Adult Center, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy.
- IRCCS Humanitas Research Hospital, Respiratory Unit, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| |
Collapse
|
27
|
Tomos I, Roussis I, Matthaiou AM, Dimakou K. Molecular and Genetic Biomarkers in Idiopathic Pulmonary Fibrosis: Where Are We Now? Biomedicines 2023; 11:2796. [PMID: 37893169 PMCID: PMC10604739 DOI: 10.3390/biomedicines11102796] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) represents a chronic progressive fibrotic interstitial lung disease of unknown cause with an ominous prognosis. It remains an unprecedent clinical challenge due to its delayed diagnosis and unpredictable clinical course. The need for accurate diagnostic, prognostic and predisposition biomarkers in everyday clinical practice becomes more necessary than ever to ensure prompt diagnoses and early treatment. The identification of such blood biomarkers may also unravel novel drug targets against IPF development and progression. So far, the role of diverse blood biomarkers, implicated in various pathogenetic pathways, such as in fibrogenesis (S100A4), extracellular matrix remodelling (YKL-40, MMP-7, ICAM-1, LOXL2, periostin), chemotaxis (CCL-18, IL-8), epithelial cell injury (KL-6, SP-A, SP-D), autophagy and unfolded protein response has been investigated in IPF with various results. Moreover, the recent progress in genetics in IPF allows for a better understanding of the underlying disease mechanisms. So far, the causative mutations in pulmonary fibrosis include mutations in telomere-related genes and in surfactant-related genes, markers that could act as predisposition biomarkers in IPF. The aim of this review is to provide a comprehensive overview from the bench to bedside of current knowledge and recent insights on biomarkers in IPF, and to suggest future directions for research. Large-scale studies are still needed to confirm the exact role of these biomarkers.
Collapse
Affiliation(s)
- Ioannis Tomos
- 5th Department of Respiratory Medicine, ‘SOTIRIA’ Chest Diseases Hospital of Athens, 11527 Athens, Greece; (I.R.); (A.M.M.); (K.D.)
| | - Ioannis Roussis
- 5th Department of Respiratory Medicine, ‘SOTIRIA’ Chest Diseases Hospital of Athens, 11527 Athens, Greece; (I.R.); (A.M.M.); (K.D.)
| | - Andreas M. Matthaiou
- 5th Department of Respiratory Medicine, ‘SOTIRIA’ Chest Diseases Hospital of Athens, 11527 Athens, Greece; (I.R.); (A.M.M.); (K.D.)
- Laboratory of Molecular and Cellular Pneumonology, Medical School, University of Crete, 714 09 Heraklion, Greece
- Respiratory Physiology Laboratory, Medical School, University of Cyprus, Nicosia 2029, Cyprus
| | - Katerina Dimakou
- 5th Department of Respiratory Medicine, ‘SOTIRIA’ Chest Diseases Hospital of Athens, 11527 Athens, Greece; (I.R.); (A.M.M.); (K.D.)
| |
Collapse
|
28
|
Hannan SJ, Iasella CJ, Sutton RM, Popescu ID, Koshy R, Burke R, Chen X, Zhang Y, Pilewski JM, Hage CA, Sanchez PG, Im A, Farah R, Alder JK, McDyer JF. Lung transplant recipients with telomere-mediated pulmonary fibrosis have increased risk for hematologic complications. Am J Transplant 2023; 23:1590-1602. [PMID: 37392813 PMCID: PMC11062487 DOI: 10.1016/j.ajt.2023.06.014] [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: 01/18/2023] [Revised: 05/31/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Idiopathic pulmonary fibrosis lung transplant recipients (IPF-LTRs) are enriched for short telomere length (TL) and telomere gene rare variants. A subset of patients with nontransplant short-TL are at increased risk for bone marrow (BM) dysfunction. We hypothesized that IPF-LTRs with short-TL and/or rare variants would be at increased risk for posttransplant hematologic complications. Data were extracted from a retrospective cohort of 72 IPF-LTRs and 72 age-matched non-IPF-LTR controls. Genetic assessment was done using whole genome sequencing or targeted sequence panel. TL was measured using flow cytometry and fluorescence in-situ hybridization (FlowFISH) and TelSeq software. The majority of the IPF-LTR cohort had short-TL, and 26% of IPF-LTRs had rare variants. Compared to non-IPF controls, short-TL IPF-LTRs were more likely to have immunosuppression agents discontinued due to cytopenias (P = .0375), and BM dysfunction requiring BM biopsy was more prevalent (29% vs 4%, P = .0003). IPF-LTRs with short-TL and rare variants had increased requirements for transfusion and growth factor support. Multivariable logistic regression demonstrated that short-TL, rare variants, and lower pretransplant platelet counts were associated with BM dysfunction. Pretransplant TL measurement and genetic testing for rare telomere gene variants identified IPF-LTRs at increased risk for hematologic complications. Our findings support stratification for telomere-mediated pulmonary fibrosis in lung transplant candidates.
Collapse
Affiliation(s)
- Stefanie J Hannan
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Carlo J Iasella
- Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rachel M Sutton
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Iulia D Popescu
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ritchie Koshy
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robin Burke
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiaoping Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joseph M Pilewski
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chadi A Hage
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Pablo G Sanchez
- Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Division of Lung Transplant and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Annie Im
- Hillman Cancer Center, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rafic Farah
- Hillman Cancer Center, University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jonathan K Alder
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John F McDyer
- Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Lung Transplant Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
29
|
Wang M, Cao L. Hydrolysable tannins as a potential therapeutic drug for the human fibrosis-associated disease. Drug Dev Res 2023; 84:1096-1113. [PMID: 37386756 DOI: 10.1002/ddr.22089] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/22/2023] [Accepted: 06/06/2023] [Indexed: 07/01/2023]
Abstract
Fibrosis is a pathological change with abnormal tissue regeneration due to a response to persistent injury, which is extensively related to organ damage and failure, leading to high morbidity and mortality worldwide. Although the pathogenesis of fibrosis has been comprehensively elucidated, there are few effective therapies for treating fibrotic diseases. Natural products are increasingly regarded as an effective strategy for fibrosis with numerous favorable functions. Hydrolysable tannins (HT) are a type of natural products that have the potential to treat the fibrotic disease. In this review, we describe some biological activities and the therapeutic prospects of HT in organ fibrosis. Furthermore, the underlying mechanisms of inhibition of HT on fibrotic organs in relation to inflammation, oxidative stress, epithelial-mesenchymal transition, fibroblast activation and proliferation, and extracellular matrix accumulation are discussed. Understanding the mechanism of HT against fibrotic diseases will provide a new strategy for the prevention and attenuation of fibrosis progression.
Collapse
Affiliation(s)
- Meiwei Wang
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Linghui Cao
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| |
Collapse
|
30
|
Stanel SC, Callum J, Rivera-Ortega P. Genetic and environmental factors in interstitial lung diseases: current and future perspectives on early diagnosis of high-risk cohorts. Front Med (Lausanne) 2023; 10:1232655. [PMID: 37601795 PMCID: PMC10435297 DOI: 10.3389/fmed.2023.1232655] [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: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Within the wide scope of interstitial lung diseases (ILDs), familial pulmonary fibrosis (FPF) is being increasingly recognized as a specific entity, with earlier onset, faster progression, and suboptimal responses to immunosuppression. FPF is linked to heritable pathogenic variants in telomere-related genes (TRGs), surfactant-related genes (SRGs), telomere shortening (TS), and early cellular senescence. Telomere abnormalities have also been identified in some sporadic cases of fibrotic ILD. Air pollution and other environmental exposures carry additive risk to genetic predisposition in pulmonary fibrosis. We provide a perspective on how these features impact on screening strategies for relatives of FPF patients, interstitial lung abnormalities, ILD multi-disciplinary team (MDT) discussion, and disparities and barriers to genomic testing. We also describe our experience with establishing a familial interstitial pneumonia (FIP) clinic and provide guidance on how to identify patients with telomere dysfunction who would benefit most from genomic testing.
Collapse
Affiliation(s)
- Stefan Cristian Stanel
- Interstitial Lung Disease Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jack Callum
- Interstitial Lung Disease Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Pilar Rivera-Ortega
- Interstitial Lung Disease Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
31
|
Adams TN, Batra K, Kypreos M, Glazer CS. Impact of radiographic honeycombing on transplant free survival and efficacy of immunosuppression in fibrotic hypersensitivity pneumonitis. BMC Pulm Med 2023; 23:224. [PMID: 37349780 DOI: 10.1186/s12890-023-02523-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/17/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND The distinction between hypersensitivity pneumonitis (HP) and idiopathic pulmonary fibrosis (IPF) was thought to be important due to the difference in mortality between the conditions as well as the response to treatment. However, recent work suggests that the clinical diagnosis may matter less than certain radiographic features, namely usual interstitial pneumonia (UIP) pattern. The purpose of this study is to evaluate whether radiographic honeycombing is more predictive of transplant-free survival (TFS) than other clinical, radiographic, or histologic findings that distinguish HP from IPF in the current guidelines and to evaluate the impact of radiographic honeycombing on the efficacy of immunosuppression in fibrotic HP. METHODS We retrospectively identified IPF and fibrotic HP patients evaluated between 2003 and 2019. Univariable and multivariable logistic regression was performed for patients with fibrotic HP and IPF to evaluate TFS. To assess the impact of treatment with immunosuppression on TFS in fibrotic HP, a cox proportional hazard model adjusted for known predictors of survival in HP including age, gender, and baseline pulmonary function testing results was constructed, and p-interaction for the presence of honeycombing on high resolution computed tomography and use of immunosuppression was calculated. RESULTS Our cohort included 178 with IPF and 198 with fibrotic HP. In a multivariable analysis, the presence of honeycombing had a greater impact on the TFS than the diagnosis of HP vs. IPF. Among the criteria used in the HP diagnostic guidelines, only typical HP scan impacted survival in a multivariable model, while identification of antigen and surgical lung biopsy findings had no impact on survival. We identified a trend toward worse survival on immunosuppression in those with HP with radiographic honeycombing. CONCLUSION Our data suggests that honeycombing and baseline pulmonary function testing have a greater impact on TFS than the clinical diagnosis of IPF vs. fibrotic HP and that radiographic honeycombing is a predictor of poor TFS in fibrotic HP. We suggest that invasive diagnostic testing including surgical lung biopsy may not be useful in predicting mortality in HP patients with honeycombing and may potentially increase risk of immunosuppression.
Collapse
Affiliation(s)
- Traci N Adams
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75219, USA.
| | - Kiran Batra
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75219, USA
| | - Margaret Kypreos
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75219, USA
| | - Craig S Glazer
- Division of Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75219, USA
| |
Collapse
|
32
|
Kim JS, Manichaikul AW, Hoffman EA, Balte P, Anderson MR, Bernstein EJ, Madahar P, Oelsner EC, Kawut SM, Wysoczanski A, Laine AF, Adegunsoye A, Ma JZ, Taub MA, Mathias RA, Rich SS, Rotter JI, Noth I, Garcia CK, Barr RG, Podolanczuk AJ. MUC5B, telomere length and longitudinal quantitative interstitial lung changes: the MESA Lung Study. Thorax 2023; 78:566-573. [PMID: 36690926 PMCID: PMC9899287 DOI: 10.1136/thorax-2021-218139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 07/11/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND The MUC5B promoter variant (rs35705950) and telomere length are linked to pulmonary fibrosis and CT-based qualitative assessments of interstitial abnormalities, but their associations with longitudinal quantitative changes of the lung interstitium among community-dwelling adults are unknown. METHODS We used data from participants in the Multi-Ethnic Study of Atherosclerosis with high-attenuation areas (HAAs, Examinations 1-6 (2000-2018)) and MUC5B genotype (n=4552) and telomere length (n=4488) assessments. HAA was defined as the per cent of imaged lung with attenuation of -600 to -250 Hounsfield units. We used linear mixed-effects models to examine associations of MUC5B risk allele (T) and telomere length with longitudinal changes in HAAs. Joint models were used to examine associations of longitudinal changes in HAAs with death and interstitial lung disease (ILD). RESULTS The MUC5B risk allele (T) was associated with an absolute change in HAAs of 2.60% (95% CI 0.36% to 4.86%) per 10 years overall. This association was stronger among those with a telomere length below an age-adjusted percentile of 5% (p value for interaction=0.008). A 1% increase in HAAs per year was associated with 7% increase in mortality risk (rate ratio (RR)=1.07, 95% CI 1.02 to 1.12) for overall death and 34% increase in ILD (RR=1.34, 95% CI 1.20 to 1.50). Longer baseline telomere length was cross-sectionally associated with less HAAs from baseline scans, but not with longitudinal changes in HAAs. CONCLUSIONS Longitudinal increases in HAAs were associated with the MUC5B risk allele and a higher risk of death and ILD.
Collapse
Affiliation(s)
- John S Kim
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Ani W Manichaikul
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Iowa City, Iowa, USA
| | - Pallavi Balte
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Michaela R Anderson
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Elana J Bernstein
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Purnema Madahar
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Elizabeth C Oelsner
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics and Epidemiology, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Artur Wysoczanski
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Andrew F Laine
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | | | - Jennie Z Ma
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Margaret A Taub
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Rasika A Mathias
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stephen S Rich
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jerome I Rotter
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California, USA
- The Institute for Translational Genomics and Population Sciences, The Lundquist Institute, Harbor-UCLA Medical Center, Torrance, California, USA
| | - Imre Noth
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Christine Kim Garcia
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - R Graham Barr
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Anna J Podolanczuk
- Division of Pulmonary and Critical Care, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
33
|
Brazee PL, Knipe RS. Vascular BMPRs Strike Out the Rolling Ball of Fibrosis. Am J Respir Crit Care Med 2023; 207:1419-1421. [PMID: 36952237 PMCID: PMC10263134 DOI: 10.1164/rccm.202303-0382ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Affiliation(s)
- Patricia L Brazee
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital Harvard Medical School Boston, Massachusetts
| | - Rachel S Knipe
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital Harvard Medical School Boston, Massachusetts
| |
Collapse
|
34
|
Adegunsoye A, Newton CA, Oldham JM, Ley B, Lee CT, Linderholm AL, Chung JH, Garcia N, Zhang D, Vij R, Guzy R, Jablonski R, Bag R, Voogt RS, Ma SF, Sperling AI, Raghu G, Martinez FJ, Strek ME, Wolters PJ, Garcia CK, Pierce BL, Noth I. Telomere length associates with chronological age and mortality across racially diverse pulmonary fibrosis cohorts. Nat Commun 2023; 14:1489. [PMID: 36932145 PMCID: PMC10023792 DOI: 10.1038/s41467-023-37193-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Pulmonary fibrosis (PF) is characterized by profound scarring and poor survival. We investigated the association of leukocyte telomere length (LTL) with chronological age and mortality across racially diverse PF cohorts. LTL measurements among participants with PF stratified by race/ethnicity were assessed in relation to age and all-cause mortality, and compared to controls. Generalized linear models were used to evaluate the age-LTL relationship, Cox proportional hazards models were used for hazard ratio estimation, and the Cochran-Armitage test was used to assess quartiles of LTL. Standardized LTL shortened with increasing chronological age; this association in controls was strengthened in PF (R = -0.28; P < 0.0001). In PF, age- and sex-adjusted LTL below the median consistently predicted worse mortality across all racial groups (White, HR = 2.21, 95% CI = 1.79-2.72; Black, HR = 2.22, 95% CI = 1.05-4.66; Hispanic, HR = 3.40, 95% CI = 1.88-6.14; and Asian, HR = 2.11, 95% CI = 0.55-8.23). LTL associates uniformly with chronological age and is a biomarker predictive of mortality in PF across racial groups.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA.
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL, USA.
| | - Chad A Newton
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX, USA
| | - Justin M Oldham
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Brett Ley
- Section of Pulmonary & Critical Care Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Cathryn T Lee
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Angela L Linderholm
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Jonathan H Chung
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | - Nicole Garcia
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Da Zhang
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University Medical Center, New York, NY, USA
| | - Rekha Vij
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Robert Guzy
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Renea Jablonski
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Remzi Bag
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Rebecca S Voogt
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Shwu-Fan Ma
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Anne I Sperling
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Ganesh Raghu
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington Medical Center, Seattle, WA, USA
| | - Fernando J Martinez
- Pulmonary Critical Care Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Mary E Strek
- Section of Pulmonary & Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Paul J Wolters
- Section of Pulmonary & Critical Care Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Christine Kim Garcia
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University Medical Center, New York, NY, USA
| | - Brandon L Pierce
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Imre Noth
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
35
|
Borie R, Kannengiesser C, Antoniou K, Bonella F, Crestani B, Fabre A, Froidure A, Galvin L, Griese M, Grutters JC, Molina-Molina M, Poletti V, Prasse A, Renzoni E, van der Smagt J, van Moorsel CHM. European Respiratory Society statement on familial pulmonary fibrosis. Eur Respir J 2023; 61:13993003.01383-2022. [PMID: 36549714 DOI: 10.1183/13993003.01383-2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
Genetic predisposition to pulmonary fibrosis has been confirmed by the discovery of several gene mutations that cause pulmonary fibrosis. Although genetic sequencing of familial pulmonary fibrosis (FPF) cases is embedded in routine clinical practice in several countries, many centres have yet to incorporate genetic sequencing within interstitial lung disease (ILD) services and proper international consensus has not yet been established. An international and multidisciplinary expert Task Force (pulmonologists, geneticists, paediatrician, pathologist, genetic counsellor, patient representative and librarian) reviewed the literature between 1945 and 2022, and reached consensus for all of the following questions: 1) Which patients may benefit from genetic sequencing and clinical counselling? 2) What is known of the natural history of FPF? 3) Which genes are usually tested? 4) What is the evidence for telomere length measurement? 5) What is the role of common genetic variants (polymorphisms) in the diagnostic workup? 6) What are the optimal treatment options for FPF? 7) Which family members are eligible for genetic sequencing? 8) Which clinical screening and follow-up parameters may be considered in family members? Through a robust review of the literature, the Task Force offers a statement on genetic sequencing, clinical management and screening of patients with FPF and their relatives. This proposal may serve as a basis for a prospective evaluation and future international recommendations.
Collapse
Affiliation(s)
- Raphael Borie
- Université Paris Cité, Inserm, PHERE, Hôpital Bichat, AP-HP, Service de Pneumologie A, Centre Constitutif du Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | | | - Katerina Antoniou
- Laboratory of Molecular and Cellular Pneumonology, Department of Respiratory Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Francesco Bonella
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik, University Hospital, University of Essen, European Reference Network (ERN)-LUNG, ILD Core Network, Essen, Germany
| | - Bruno Crestani
- Université Paris Cité, Inserm, PHERE, Hôpital Bichat, AP-HP, Service de Pneumologie A, Centre Constitutif du Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | - Aurélie Fabre
- Department of Histopathology, St Vincent's University Hospital and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Antoine Froidure
- Pulmonology Department, Cliniques Universitaires Saint-Luc and Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Liam Galvin
- European Pulmonary Fibrosis Federation, Blackrock, Ireland
| | - Matthias Griese
- Dr von Haunersches Kinderspital, University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Jan C Grutters
- ILD Center of Excellence, St Antonius Hospital, Nieuwegein, The Netherlands
- Division of Heart and Lungs, UMC Utrecht, Utrecht, The Netherlands
| | - Maria Molina-Molina
- Interstitial Lung Disease Unit, Respiratory Department, University Hospital of Bellvitge, IDIBELL, Hospitalet de Llobregat (Barcelona), CIBERES, Barcelona, Spain
| | - Venerino Poletti
- Department of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
- Department of Experimental, Diagnostics and Speciality Medicine, University of Bologna, Bologna, Italy
| | - Antje Prasse
- Department of Pulmonology, Hannover Medical School, German Center for Lung Research (DZL), BREATH, Hannover, Germany
- Fraunhofer ITEM, Hannover, Germany
| | - Elisabetta Renzoni
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Clinical Group, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jasper van der Smagt
- Division of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
36
|
Nelson N, Feurstein S, Niaz A, Truong J, Holien JK, Lucas S, Fairfax K, Dickinson J, Bryan TM. Functional genomics for curation of variants in telomere biology disorder associated genes: A systematic review. Genet Med 2023; 25:100354. [PMID: 36496180 DOI: 10.1016/j.gim.2022.11.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Patients with an underlying telomere biology disorder (TBD) have variable clinical presentations, and they can be challenging to diagnose clinically. A genomic diagnosis for patients presenting with TBD is vital for optimal treatment. Unfortunately, many variants identified during diagnostic testing are variants of uncertain significance. This complicates management decisions, delays treatment, and risks nonuptake of potentially curative therapies. Improved application of functional genomic evidence may reduce variants of uncertain significance classifications. METHODS We systematically searched the literature for published functional assays interrogating TBD gene variants. When possible, established likely benign/benign and likely pathogenic/pathogenic variants were used to estimate the assay sensitivity, specificity, positive predictive value, negative predictive value, and odds of pathogenicity. RESULTS In total, 3131 articles were screened and 151 met inclusion criteria. Sufficient data to enable a PS3/BS3 recommendation were available for TERT variants only. We recommend that PS3 and BS3 can be applied at a moderate and supportive level, respectively. PS3/BS3 application was limited by a lack of assay standardization and limited inclusion of benign variants. CONCLUSION Further assay standardization and assessment of benign variants are required for optimal use of the PS3/BS3 criterion for TBD gene variant classification.
Collapse
Affiliation(s)
- Niles Nelson
- The Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, Tasmania, Australia; Department of Molecular Medicine, The Royal Hobart Hospital, Hobart, Tasmania, Australia; Department of Molecular Haematology, The Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
| | - Simone Feurstein
- Section of Hematology, Oncology, and Rheumatology, Department of Internal Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Aram Niaz
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, New South Wales, Australia
| | - Jia Truong
- School of Science, STEM College, RMIT University, Bundoora, Victoria, Australia
| | - Jessica K Holien
- School of Science, STEM College, RMIT University, Bundoora, Victoria, Australia
| | - Sionne Lucas
- The Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, Tasmania, Australia
| | - Kirsten Fairfax
- The Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, Tasmania, Australia
| | - Joanne Dickinson
- The Menzies Institute for Medical Research, College of Health and Medicine, The University of Tasmania, Hobart, Tasmania, Australia
| | - Tracy M Bryan
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, New South Wales, Australia
| |
Collapse
|
37
|
Usual interstitial pneumonia as a stand-alone diagnostic entity: the case for a paradigm shift? THE LANCET. RESPIRATORY MEDICINE 2023; 11:188-196. [PMID: 36640788 DOI: 10.1016/s2213-2600(22)00475-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 01/13/2023]
Abstract
Usual interstitial pneumonia (UIP) is characterised by a distinctive morphological and radiological appearance that was considered the pathognomonic hallmark of idiopathic pulmonary fibrosis (IPF). However, this peculiar lung remodelling pattern is also seen in other fibrotic interstitial lung diseases, including hypersensitivity pneumonitis, and connective tissue diseases. In this Personal View, we advocate the designation of a UIP pattern as a single, discrete diagnostic entity, amalgamating its primary form and secondary processes in disorders such as hypersensitivity pneumonitis (hypersensitivity pneumonitis with UIP), rheumatoid arthritis (rheumatoid arthritis with UIP), and others. The current separation between primary and secondary UIP is in keeping with the view that every individual interstitial lung disease must be viewed as a separate entity but does not reflect striking similarities between primary and secondary UIP in the morphological or radiological appearance, clinical behaviour, pathogenic pathways, and the efficacy of anti-fibrotic therapy. We believe that the unification of UIP as a single diagnostic entity has undeniable advantages.
Collapse
|
38
|
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: 0] [Impact Index Per Article: 0] [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
|
39
|
Stanel SC, Rivera-Ortega P. Present and future perspectives in early diagnosis and monitoring for progressive fibrosing interstitial lung diseases. Front Med (Lausanne) 2023; 10:1114722. [PMID: 36873896 PMCID: PMC9975385 DOI: 10.3389/fmed.2023.1114722] [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: 12/02/2022] [Accepted: 01/26/2023] [Indexed: 02/17/2023] Open
Abstract
Progressive fibrosing interstitial lung diseases (PF-ILDs) represent a group of conditions of both known and unknown origin which continue to worsen despite standard treatments, leading to respiratory failure and early mortality. Given the potential to slow down progression by initiating antifibrotic therapies where appropriate, there is ample opportunity to implement innovative strategies for early diagnosis and monitoring with the goal of improving clinical outcomes. Early diagnosis can be facilitated by standardizing ILD multidisciplinary team (MDT) discussions, implementing machine learning algorithms for chest computed-tomography quantitative analysis and novel magnetic-resonance imaging techniques, as well as measuring blood biomarker signatures and genetic testing for telomere length and identification of deleterious mutations in telomere-related genes and other single-nucleotide polymorphisms (SNPs) linked to pulmonary fibrosis such as rs35705950 in the MUC5B promoter region. Assessing disease progression in the post COVID-19 era also led to a number of advances in home monitoring using digitally-enabled home spirometers, pulse oximeters and other wearable devices. While validation for many of these innovations is still in progress, significant changes to current clinical practice for PF-ILDs can be expected in the near future.
Collapse
Affiliation(s)
- Stefan Cristian Stanel
- Interstitial Lung Disease (ILD) Unit, North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe, United Kingdom.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Pilar Rivera-Ortega
- Interstitial Lung Disease (ILD) Unit, North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe, United Kingdom
| |
Collapse
|
40
|
Genetic associations and lung function in IPF: unexpected answers, persistent questions. THE LANCET. RESPIRATORY MEDICINE 2023; 11:5-6. [PMID: 35985356 DOI: 10.1016/s2213-2600(22)00270-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/27/2022]
|
41
|
Cocconcelli E, Bernardinello N, Giraudo C, Castelli G, Greco C, Polverosi R, Saetta M, Spagnolo P, Balestro E. Radiological Assessment in Idiopathic Pulmonary Fibrosis (IPF) Patients According to MUC5B Polymorphism. Int J Mol Sci 2022; 23:ijms232415890. [PMID: 36555528 PMCID: PMC9784960 DOI: 10.3390/ijms232415890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
The MUC5B rs35705950 mutant T allele is the strongest genetic risk factor for familial and sporadic IPF. We sought to determine whether MUC5B genotype influences radiological patterns of IPF at diagnosis, as well as their change over time, in patients on antifibrotic therapy. Among eighty-eight IPF patients, previously genotyped for MUC5B rs35705950, we considered seventy-eight patients who were evaluated for radiological quantification of the following features both at treatment initiation (HRCT1) and after 1 year (HRCT2): ground glass opacities (AS), reticulations (IS) and honeycombing (HC). Of the evaluated patients, 69% carried at least one copy of the T allele (TT/TG). Carriers of the T allele displayed similar FVC loss in the first year of treatment as GG carriers, but overall survival at the end of follow-up was longer in the TT/TG group, compared to the GG group. In the GG group, both the AS and HC increased significantly, whereas in the TT/TG group only HC increased over the first year of treatment. MUC5B rs35705950 GG carriers are associated with increased ground glass and honeycombing extent over time and worse survival than T allele carriers. Longitudinal HRCT may help define the prognostic role of the MUC5B rs35705950 genotype.
Collapse
Affiliation(s)
- Elisabetta Cocconcelli
- Respiratory Disease Unit, Department of Cardiac Thoracic Vascular Sciences, Public Health University of Padova, 35128 Padova, Italy
| | - Nicol Bernardinello
- Respiratory Disease Unit, Department of Cardiac Thoracic Vascular Sciences, Public Health University of Padova, 35128 Padova, Italy
| | - Chiara Giraudo
- Department of Medicine-DIMED, Padova University Hospital, 35128 Padova, Italy
| | - Gioele Castelli
- Respiratory Disease Unit, Department of Cardiac Thoracic Vascular Sciences, Public Health University of Padova, 35128 Padova, Italy
| | | | | | - Marina Saetta
- Respiratory Disease Unit, Department of Cardiac Thoracic Vascular Sciences, Public Health University of Padova, 35128 Padova, Italy
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac Thoracic Vascular Sciences, Public Health University of Padova, 35128 Padova, Italy
| | - Elisabetta Balestro
- Respiratory Disease Unit, Department of Cardiac Thoracic Vascular Sciences, Public Health University of Padova, 35128 Padova, Italy
- Correspondence:
| |
Collapse
|
42
|
Zhang D, Newton CA, Wang B, Povysil G, Noth I, Martinez FJ, Raghu G, Goldstein D, Garcia CK. Utility of whole genome sequencing in assessing risk and clinically relevant outcomes for pulmonary fibrosis. Eur Respir J 2022; 60:2200577. [PMID: 36028256 PMCID: PMC10038316 DOI: 10.1183/13993003.00577-2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/12/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Whole genome sequencing (WGS) can detect variants and estimate telomere length. The clinical utility of WGS in estimating risk, progression and survival of pulmonary fibrosis patients is unknown. METHODS In this observational cohort study, we performed WGS on 949 patients with idiopathic pulmonary fibrosis or familial pulmonary fibrosis to determine rare and common variant genotypes, estimate telomere length and assess the association of genomic factors with clinical outcomes. RESULTS WGS estimates of telomere length correlated with quantitative PCR (R=0.65) and Southern blot (R=0.71) measurements. Rare deleterious qualifying variants were found in 14% of the total cohort, with a five-fold increase in those with a family history of disease versus those without (25% versus 5%). Most rare qualifying variants (85%) were found in telomere-related genes and were associated with shorter telomere lengths. Rare qualifying variants had a greater effect on telomere length than a polygenic risk score calculated using 20 common variants previously associated with telomere length. The common variant polygenic risk score predicted telomere length only in sporadic disease. Reduced transplant-free survival was associated with rare qualifying variants, shorter quantitative PCR-measured telomere lengths and absence of the MUC5B promoter (rs35705950) single nucleotide polymorphism, but not with WGS-estimated telomere length or the common variant polygenic risk score. Disease progression was associated with both measures of telomere length (quantitative PCR measured and WGS estimated), rare qualifying variants and the common variant polygenic risk score. CONCLUSION As a single test, WGS can inform pulmonary fibrosis genetic-mediated risk, evaluate the functional effect of telomere-related variants by estimating telomere length, and prognosticate clinically relevant disease outcomes.
Collapse
Affiliation(s)
- David Zhang
- Dept of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Chad A Newton
- Dept of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Binhan Wang
- Dept of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Gundula Povysil
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Imre Noth
- Dept of Medicine, University of Virginia, Charlottesville, VA, USA
| | | | - Ganesh Raghu
- Dept of Medicine, University of Washington Medical Center, Seattle, WA, USA
| | - David Goldstein
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Christine Kim Garcia
- Dept of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
43
|
Papiris SA, Kannengiesser C, Borie R, Kolilekas L, Kallieri M, Apollonatou V, Ba I, Nathan N, Bush A, Griese M, Dieude P, Crestani B, Manali ED. Genetics in Idiopathic Pulmonary Fibrosis: A Clinical Perspective. Diagnostics (Basel) 2022; 12:2928. [PMID: 36552935 PMCID: PMC9777433 DOI: 10.3390/diagnostics12122928] [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: 10/14/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Unraveling the genetic background in a significant proportion of patients with both sporadic and familial IPF provided new insights into the pathogenic pathways of pulmonary fibrosis. AIM The aim of the present study is to overview the clinical significance of genetics in IPF. PERSPECTIVE It is fascinating to realize the so-far underestimated but dynamically increasing impact that genetics has on aspects related to the pathophysiology, accurate and early diagnosis, and treatment and prevention of this devastating disease. Genetics in IPF have contributed as no other in unchaining the disease from the dogma of a "a sporadic entity of the elderly, limited to the lungs" and allowed all scientists, but mostly clinicians, all over the world to consider its many aspects and "faces" in all age groups, including its co-existence with several extra pulmonary conditions from cutaneous albinism to bone-marrow and liver failure. CONCLUSION By providing additional evidence for unsuspected characteristics such as immunodeficiency, impaired mucus, and surfactant and telomere maintenance that very often co-exist through the interaction of common and rare genetic variants in the same patient, genetics have created a generous and pluralistic yet unifying platform that could lead to the understanding of the injurious and pro-fibrotic effects of many seemingly unrelated extrinsic and intrinsic offending factors. The same platform constantly instructs us about our limitations as well as about the heritability, the knowledge and the wisdom that is still missing.
Collapse
Affiliation(s)
- Spyros A. Papiris
- 2nd Pulmonary Medicine Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Caroline Kannengiesser
- Département de Génétique, APHP Hôpital Bichat, Université de Paris, 75018 Paris, France
- INSERM UMR 1152, Université de Paris, 75018 Paris, France
| | - Raphael Borie
- Service de Pneumologie A, INSERM UMR_1152, Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, APHP Hôpital Bichat, Sorbonne Université, 75018 Paris, France
| | - Lykourgos Kolilekas
- 7th Pulmonary Department, Athens Chest Hospital “Sotiria”, 11527 Athens, Greece
| | - Maria Kallieri
- 2nd Pulmonary Medicine Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Vasiliki Apollonatou
- 2nd Pulmonary Medicine Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Ibrahima Ba
- Département de Génétique, APHP Hôpital Bichat, Université de Paris, 75018 Paris, France
| | - Nadia Nathan
- Peditric Pulmonology Department and Reference Centre for Rare Lung Diseases RespiRare, INSERM UMR_S933 Laboratory of Childhood Genetic Diseases, Armand Trousseau Hospital, Sorbonne University and APHP, 75012 Paris, France
| | - Andrew Bush
- Paediatrics and Paediatric Respirology, Imperial College, Imperial Centre for Paediatrics and Child Health, Royal Brompton Harefield NHS Foundation Trust, London SW3 6NP, UK
| | - Matthias Griese
- Department of Pediatric Pneumology, Dr von Hauner Children’s Hospital, Ludwig-Maximilians-University, German Center for Lung Research, 80337 Munich, Germany
| | - Philippe Dieude
- Department of Rheumatology, INSERM U1152, APHP Hôpital Bichat-Claude Bernard, Université de Paris, 75018 Paris, France
| | - Bruno Crestani
- Service de Pneumologie A, INSERM UMR_1152, Centre de Référence des Maladies Pulmonaires Rares, FHU APOLLO, APHP Hôpital Bichat, Sorbonne Université, 75018 Paris, France
| | - Effrosyni D. Manali
- 2nd Pulmonary Medicine Department, General University Hospital “Attikon”, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
44
|
Idiopathic Pulmonary Fibrosis and Telomeres. J Clin Med 2022; 11:jcm11236893. [PMID: 36498467 PMCID: PMC9740997 DOI: 10.3390/jcm11236893] [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: 10/26/2022] [Revised: 11/10/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis is an interstitial lung disease of unknown etiology with a highly compromised prognosis and a significant mortality rate within a few years of diagnosis. Despite being idiopathic, it has been shown that telomeric shortening could play an important role in its etiopathogenesis. Mutations in telomere-related genes have been identified, but they are not always present despite telomere shortening. On the other hand, this telomeric shortening has been linked to a worse prognosis of the disease independently of other clinical factors, implying it may serve as a biomarker.
Collapse
|
45
|
Piotrowski WJ, Martusewicz-Boros MM, Białas AJ, Barczyk A, Batko B, Błasińska K, Boros PW, Górska K, Grzanka P, Jassem E, Jastrzębski D, Kaczyńska J, Kowal-Bielecka O, Kucharz E, Kuś J, Kuźnar-Kamińska B, Kwiatkowska B, Langfort R, Lewandowska K, Mackiewicz B, Majewski S, Makowska J, Miłkowska-Dymanowska J, Puścińska E, Siemińska A, Sobiecka M, Soroka-Dąda RA, Szołkowska M, Wiatr E, Ziora D, Śliwiński P. Guidelines of the Polish Respiratory Society on the Diagnosis and Treatment of Progressive Fibrosing Interstitial Lung Diseases Other than Idiopathic Pulmonary Fibrosis. Adv Respir Med 2022; 90:425-450. [PMID: 36285980 PMCID: PMC9717335 DOI: 10.3390/arm90050052] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 08/24/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2023]
Abstract
The recommendations were developed as answers to previously formulated questions concerning everyday diagnostic and therapeutic challenges. They were developed based on a review of the current literature using the GRADE methodology. The experts suggest that PF-ILD be diagnosed based on a combination of different criteria, such as the aggravation of symptoms, progression of radiological lesions, and worsening of lung function test parameters. The experts recommend a precise diagnosis of an underlying disease, with serological testing for an autoimmune disease always being included. The final diagnosis should be worked out by a multidisciplinary team (MDT). Patients with an interstitial lung disease other than IPF who do not meet the criteria for the progressive fibrosis phenotype should be monitored for progression, and those with systemic autoimmune diseases should be regularly monitored for signs of interstitial lung disease. In managing patients with interstitial lung disease associated with autoimmune diseases, an opinion of an MDT should be considered. Nintedanib rather than pirfenidon should be introduced in the event of the ineffectiveness of the therapy recommended for the treatment of the underlying disease, but in some instances, it is possible to start antifibrotic treatment without earlier immunomodulatory therapy. It is also admissible to use immunomodulatory and antifibrotic drugs simultaneously. No recommendations were made for or against termination of anti-fibrotic therapy in the case of noted progression during treatment of a PF-ILD other than IPF. The experts recommend that the same principles of non-pharmacological and palliative treatment and eligibility for lung transplantation should be applied to patients with an interstitial lung disease other than IPF with progressive fibrosis as in patients with IPF.
Collapse
Affiliation(s)
| | - Magdalena M. Martusewicz-Boros
- 3rd Lung Diseases and Oncology Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Adam J. Białas
- Department of Pathobiology of Respiratory Diseases, Medical University of Lodz, 90-153 Lodz, Poland
| | - Adam Barczyk
- Department of Pneumonology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Bogdan Batko
- Department of Rheumatology and Immunology, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University, 30-705 Krakow, Poland
| | - Katarzyna Błasińska
- Department of Radiology, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Piotr W. Boros
- Lung Pathophysiology Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Katarzyna Górska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Piotr Grzanka
- Department of Radiology, Voivodeship Hospital in Opole, 45-061 Opole, Poland
| | - Ewa Jassem
- Department of Allergology and Pneumonology, Medical University of Gdansk, 80-214 Gdańsk, Poland
| | - Dariusz Jastrzębski
- Department of Lung Diseases and Tuberculosis, Medical University of Silesia, 41-803 Zabrze, Poland
| | | | - Otylia Kowal-Bielecka
- Department of Rheumatology and Internal Medicine, Medical University of Białystok, 15-276 Białystok, Poland
| | - Eugeniusz Kucharz
- Department of Internal Medicine, Rheumatology and Clinical Immunology, Medical University of Silesia, 40-635 Katowice, Poland
| | - Jan Kuś
- 1st Lung Diseases Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Barbara Kuźnar-Kamińska
- Department of Pulmonology, Allergology and Respiratory Oncology, University of Medical Sciences in Poznan, 61-701 Poznan, Poland
| | - Brygida Kwiatkowska
- Department of Rheumatology, Eleonora Reicher Rheumatology Institute, 02-637 Warszawa, Poland
| | - Renata Langfort
- Department of Pathology, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warszawa, Poland
| | - Katarzyna Lewandowska
- 1st Lung Diseases Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Barbara Mackiewicz
- Department of Pneumonology, Oncology and Allergology, Medical University, Lublin, 20-090 Lublin, Poland
| | - Sebastian Majewski
- Department of Pneumology, Medical University of Lodz, 90-153 Lodz, Poland
| | - Joanna Makowska
- Department of Rheumatology, Medical University of Lodz, 92-213 Lodz, Poland
| | | | - Elżbieta Puścińska
- 2nd Department of Respiratory Medicine, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Alicja Siemińska
- Department of Allergology, Medical University of Gdańsk, 80-214 Gdansk, Poland
| | - Małgorzata Sobiecka
- 1st Lung Diseases Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | | | - Małgorzata Szołkowska
- Department of Pathology, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warszawa, Poland
| | - Elżbieta Wiatr
- 3rd Lung Diseases and Oncology Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Dariusz Ziora
- Department of Lung Diseases and Tuberculosis, Medical University of Silesia, 41-803 Zabrze, Poland
| | - Paweł Śliwiński
- 2nd Department of Respiratory Medicine, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| |
Collapse
|
46
|
Wijsenbeek M, Suzuki A, Maher TM. Interstitial lung diseases. Lancet 2022; 400:769-786. [PMID: 35964592 DOI: 10.1016/s0140-6736(22)01052-2] [Citation(s) in RCA: 144] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 03/14/2022] [Accepted: 06/03/2022] [Indexed: 02/07/2023]
Abstract
Over 200 interstitial lung diseases, from ultra rare to relatively common, are recognised. Most interstitial lung diseases are characterised by inflammation or fibrosis within the interstitial space, the primary consequence of which is impaired gas exchange, resulting in breathlessness, diminished exercise tolerance, and decreased quality of life. Outcomes vary considerably for each of the different interstitial lung diseases. In some conditions, spontaneous reversibility or stabilisation can occur, but unfortunately in many people with interstitial lung disease, especially in those manifesting progressive pulmonary fibrosis, respiratory failure and death are a sad reality. Over the past 3 years, the field of interstitial lung disease has had important advances, with the approval of drugs to treat systemic sclerosis-associated interstitial lung disease, interstitial lung disease-associated pulmonary hypertension, and different forms of progressive pulmonary fibrosis. This Seminar provides an update on epidemiology, pathogenesis, presentation, diagnosis, disease course, and management of the interstitial lung diseases that are most frequently encountered in clinical practice. Furthermore, we describe how developments have led to a shift in the classification and treatment of interstitial lung diseases that exhibit progressive pulmonary fibrosis and summarise the latest practice-changing guidelines. We conclude with an outline of controversies, uncertainties, and future directions.
Collapse
Affiliation(s)
- Marlies Wijsenbeek
- Center for Interstitial Lung Diseases and Sarcoidosis, Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.
| | - Atsushi Suzuki
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Toby M Maher
- Hastings Centre for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
47
|
Kam M, Caliez J, Nunes H, Gille T. The road to hell is paved with good intentions: a look back at the PANTHER-IPF trial. Breathe (Sheff) 2022; 18:220074. [PMID: 36340823 PMCID: PMC9584592 DOI: 10.1183/20734735.0074-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/18/2022] [Indexed: 11/05/2022] Open
Abstract
The PANTHER-IPF trial was a turning point in treatment of idiopathic pulmonary fibrosis (#IPF) highlighting the importance of randomised controlled trials in determining treatment strategies, even for rare diseases and/or potentially fatal acute events https://bit.ly/3Oi0KwD.
Collapse
Affiliation(s)
- Michelle Kam
- Dept of Respiratory and Critical Care Medicine, Singapore General Hospital, Singapore,Both authors contributed equally to this manuscript,Corresponding author: Michelle Kam ()
| | - Julien Caliez
- Dept of Pulmonology, Reference Center for Rare Pulmonary Diseases, Avicenne University Hospital, Hôpitaux Universitaires de Paris Seine-Saint-Denis (HUPSSD), Assistance Publique - Hôpitaux de Paris (AP-HP), Bobigny, France,Both authors contributed equally to this manuscript
| | - Hilario Nunes
- Dept of Pulmonology, Reference Center for Rare Pulmonary Diseases, Avicenne University Hospital, Hôpitaux Universitaires de Paris Seine-Saint-Denis (HUPSSD), Assistance Publique - Hôpitaux de Paris (AP-HP), Bobigny, France,Inserm UMR 1272 “Hypoxia & the Lung”, UFR Santé, Médecine, Biologie Humaine (SMBH), Université Sorbonne Paris Nord (USPN), Bobigny, France
| | - Thomas Gille
- Inserm UMR 1272 “Hypoxia & the Lung”, UFR Santé, Médecine, Biologie Humaine (SMBH), Université Sorbonne Paris Nord (USPN), Bobigny, France,Dept of Physiology and Functional Explorations, Avicenne University Hospital, Hôpitaux Universitaires de Paris Seine-Saint-Denis (HUPSSD), Assistance Publique - Hôpitaux de Paris (AP-HP), Bobigny, France,Dept of Physiology and Functional Explorations, Jean Verdier University Hospital, Hôpitaux Universitaires de Paris Seine-Saint-Denis (HUPSSD), Assistance Publique - Hôpitaux de Paris (AP-HP), Bondy, France
| |
Collapse
|
48
|
Newton CA, Oldham JM, Applegate C, Carmichael N, Powell K, Dilling D, Schmidt SL, Scholand MB, Armanios M, Garcia CK, Kropski JA, Talbert J. The Role of Genetic Testing in Pulmonary Fibrosis. Chest 2022; 162:394-405. [PMID: 35337808 PMCID: PMC9424324 DOI: 10.1016/j.chest.2022.03.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 11/25/2022] Open
Abstract
Patients with familial pulmonary fibrosis represent a subset of patients with pulmonary fibrosis in whom inherited gene variation predisposes them to disease development. In the appropriate setting, genetic testing allows for personalized assessment of disease, recognition of clinically relevant extrapulmonary manifestations, and assessing susceptibility in unaffected relatives. However currently, the use of genetic testing is inconsistent, partly because of the lack of guidance regarding high-yield scenarios in which the results of genetic testing can inform clinical decision-making. To address this, the Pulmonary Fibrosis Foundation commissioned a genetic testing work group comprising pulmonologists, geneticists, and genetic counselors from the United States to provide guidance on genetic testing in patients with pulmonary fibrosis. This CHEST special feature presents a concise review of these proceedings and reviews pulmonary fibrosis susceptibility, clinically available genetic testing methods, and clinical scenarios in which genetic testing should be considered.
Collapse
|
49
|
Mackintosh JA, Chambers DC. Towards truly 'idiopathic' pulmonary fibrosis. Respirology 2022; 27:682-683. [PMID: 35764391 DOI: 10.1111/resp.14320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 11/30/2022]
Affiliation(s)
- John A Mackintosh
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Queensland Lung Transplant Program, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Daniel C Chambers
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Queensland Lung Transplant Program, The Prince Charles Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
50
|
Abbasi A, Chen C, Gandhi CK, Wu R, Pardo A, Selman M, Floros J. Single Nucleotide Polymorphisms (SNP) and SNP-SNP Interactions of the Surfactant Protein Genes Are Associated With Idiopathic Pulmonary Fibrosis in a Mexican Study Group; Comparison With Hypersensitivity Pneumonitis. Front Immunol 2022; 13:842745. [PMID: 35720392 PMCID: PMC9201215 DOI: 10.3389/fimmu.2022.842745] [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: 12/24/2021] [Accepted: 05/09/2022] [Indexed: 01/12/2023] Open
Abstract
Surfactant proteins (SPs) are important for normal lung function and innate immunity of the lungs and their genes have been identified with significant genetic variability. Changes in quantity or quality of SPs due to genetic mutations or natural genetic variability may alter their functions and contribute to the host susceptibility for particular diseases. Alternatively, SP single nucleotide polymorphisms (SNPs) can serve as markers to identify disease risk or response to therapies, as shown for other genes in a number of other studies. In the current study, we evaluated associations of SFTP SNPs with idiopathic pulmonary fibrosis (IPF) by studying novel computational models where the epistatic effects (dominant, additive, recessive) of SNP-SNP interactions could be evaluated, and then compared the results with a previously published hypersensitivity pneumonitis (HP) study where the same novel models were used. Mexican Hispanic patients (IPF=84 & HP=75) and 194 healthy control individuals were evaluated. The goal was to identify SP SNPs and SNP-SNP interactions that associate with IPF as well as SNPs and interactions that may be unique to each of these interstitial diseases or common between them. We observed: 1) in terms of IPF, i) three single SFTPA1 SNPs to associate with decreased IPF risk, ii) three SFTPA1 haplotypes to associate with increased IPF risk, and iii) a number of three-SNP interactions to associate with IPF susceptibility. 2) Comparison of IPF and HP, i) three SFTPA1 and one SFTPB SNP associated with decreased risk in IPF but increased risk in HP, and one SFTPA1 SNP associated with decreased risk in both IPF and HP, ii) a number of three-SNP interactions with the same or different effect pattern associated with IPF and/or HP susceptibility, iii) one of the three-SNP interactions that involved SNPs of SFTPA1, SFTPA2, and SFTPD, with the same effect pattern, was associated with a disease-specific outcome, a decreased and increased risk in HP and IPF, respectively. This is the first study that compares the SP gene variants in these two phenotypically similar diseases. Our findings indicate that SNPs of all SFTPs may play an important role in the genetic susceptibility to IPF and HP. Importantly, IPF and HP share some SP genetic variants, suggesting common pathophysiological mechanisms and pathways regarding surfactant biogenesis, but also some differences, highlighting the diverse underlying pathogenic mechanisms between an inflammatory-driven fibrosis (HP) and an epithelial-driven fibrosis (IPF). Alternatively, the significant SNPs identified here, along with SNPs of other genes, could serve as markers to distinguish these two devastating diseases.
Collapse
Affiliation(s)
- Ata Abbasi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.,Department of Pathology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Chixiang Chen
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chintan K Gandhi
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Rongling Wu
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Moises Selman
- Unidad de Investigación, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Joanna Floros
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics & Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|