1
|
Petersen AG, Korntner SH, Bousamaki J, Oró D, Arraut AM, Pors SE, Salinas CG, Andersen MW, Madsen MR, Nie Y, Butts J, Roqueta‐Rivera M, Simonsen U, Hansen HH, Feigh M. Reproducible lung protective effects of a TGFβR1/ALK5 inhibitor in a bleomycin-induced and spirometry-confirmed model of IPF in male mice. Physiol Rep 2024; 12:e70077. [PMID: 39394052 PMCID: PMC11469938 DOI: 10.14814/phy2.70077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/30/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024] Open
Abstract
This study comprehensively validated the bleomycin (BLEO) induced mouse model of IPF for utility in preclinical drug discovery. To this end, the model was rigorously evaluated for reproducible phenotype and TGFβ-directed treatment outcomes. Lung disease was profiled longitudinally in male C57BL6/JRJ mice receiving a single intratracheal instillation of BLEO (n = 10-12 per group). A TGFβR1/ALK5 inhibitor (ALK5i) was profiled in six independent studies in BLEO-IPF mice, randomized/stratified to treatment according to baseline body weight and non-invasive whole-body plethysmography. ALK5i (60 mg/kg/day) or vehicle (n = 10-16 per study) was administered orally for 21 days, starting 7 days after intratracheal BLEO installation. BLEO-IPF mice recapitulated functional, histological and biochemical hallmarks of IPF, including declining expiratory/inspiratory capacity and inflammatory and fibrotic lung injury accompanied by markedly elevated TGFβ levels in bronchoalveolar lavage fluid and lung tissue. Pulmonary transcriptome signatures of inflammation and fibrosis in BLEO-IPF mice were comparable to reported data in IPF patients. ALK5i promoted reproducible and robust therapeutic outcomes on lung functional, biochemical and histological endpoints in BLEO-IPF mice. The robust lung fibrotic disease phenotype, along with the consistent and reproducible lung protective effects of ALK5i treatment, makes the spirometry-confirmed BLEO-IPF mouse model highly applicable for profiling novel drug candidates for IPF.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yaohui Nie
- Enanta PharmaceuticalsWatertownMassachusettsUSA
| | | | | | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Faculty of HealthAarhus UniversityAarhusDenmark
| | | | | |
Collapse
|
2
|
Libra A, Sciacca E, Muscato G, Sambataro G, Spicuzza L, Vancheri C. Highlights on Future Treatments of IPF: Clues and Pitfalls. Int J Mol Sci 2024; 25:8392. [PMID: 39125962 PMCID: PMC11313529 DOI: 10.3390/ijms25158392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/22/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by irreversible scarring of lung tissue, leading to death. Despite recent advancements in understanding its pathophysiology, IPF remains elusive, and therapeutic options are limited and non-curative. This review aims to synthesize the latest research developments, focusing on the molecular mechanisms driving the disease and on the related emerging treatments. Unfortunately, several phase 2 studies showing promising preliminary results did not meet the primary endpoints in the subsequent phase 3, underlying the complexity of the disease and the need for new integrated endpoints. IPF remains a challenging condition with a complex interplay of genetic, epigenetic, and pathophysiological factors. Ongoing research into the molecular keystones of IPF is critical for the development of targeted therapies that could potentially stop the progression of the disease. Future directions include personalized medicine approaches, artificial intelligence integration, growth in genetic insights, and novel drug targets.
Collapse
Affiliation(s)
- Alessandro Libra
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| | - Enrico Sciacca
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| | - Giuseppe Muscato
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| | - Gianluca Sambataro
- Artroreuma s.r.l., Rheumatology Outpatient Clinic, 95030 Mascalucia, CT, Italy;
| | - Lucia Spicuzza
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Disease, Policlinico “G. Rodolico-San Marco”, University of Catania, 95123 Catania, CT, Italy; (A.L.); (E.S.); (G.M.); (L.S.)
| |
Collapse
|
3
|
England BR, Roul P, Yang Y, Hershberger D, Sayles H, Rojas J, Cannon GW, Sauer BC, Curtis JR, Baker JF, Mikuls TR. Extracting forced vital capacity from the electronic health record through natural language processing in rheumatoid arthritis-associated interstitial lung disease. Pharmacoepidemiol Drug Saf 2024; 33:e5744. [PMID: 38112272 PMCID: PMC10872496 DOI: 10.1002/pds.5744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 11/27/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023]
Abstract
PURPOSE To develop a natural language processing (NLP) tool to extract forced vital capacity (FVC) values from electronic health record (EHR) notes in patients with rheumatoid arthritis-interstitial lung disease (RA-ILD). METHODS We selected RA-ILD patients (n = 7485) in the Veterans Health Administration (VA) between 2000 and 2020 using validated ICD-9/10 codes. We identified numeric values in proximity to FVC string patterns from clinical notes in the EHR. Subsequently, we performed processing steps to account for variability in note structure, related pulmonary function test (PFT) output, and values copied across notes, then assigned dates from linked administrative procedure records. NLP-derived FVC values were compared to values recorded directly from PFT equipment available on a subset of patients. RESULTS We identified 5911 FVC values (n = 1844 patients) from PFT equipment and 15 383 values (n = 4982 patients) by NLP. Among 2610 date-matched FVC values from NLP and PFT equipment, 95.8% of values were within 5% predicted. The mean (SD) difference was 0.09% (5.9), and values strongly correlated (r = 0.94, p < 0.001), with a precision of 0.87 (95% CI 0.86, 0.88). NLP captured more patients with longitudinal FVC values (n = 3069 vs. n = 1164). Mean (SD) change in FVC %-predicted per year was similar between sources (-1.5 [30.0] NLP vs. -0.9 [16.6] PFT equipment; standardized response mean = 0.05 for both). CONCLUSIONS NLP of EHR notes increases the capture of accurate, longitudinal FVC values by three-fold over PFT equipment. Use of this NLP tool can facilitate pharmacoepidemiologic research in RA-ILD and other lung diseases by capturing this critical measure of disease severity.
Collapse
Affiliation(s)
- Bryant R. England
- VA Nebraska-Western Iowa Health Care System & Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Punyasha Roul
- VA Nebraska-Western Iowa Health Care System & Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Yangyuna Yang
- VA Nebraska-Western Iowa Health Care System & Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Daniel Hershberger
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Harlan Sayles
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE
| | | | | | - Brian C. Sauer
- VA Salt Lake City & University of Utah, Salt Lake City, UT
| | | | - Joshua F. Baker
- Corporal Michael J. Crescenz VA & University of Pennsylvania, Philadelphia, PA
| | - Ted R. Mikuls
- VA Nebraska-Western Iowa Health Care System & Division of Rheumatology & Immunology, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
4
|
Elkassem AMA, Mresh R, Farag A, Rothenberg S, Lirette ST, Smith AD, Kulkarni T. Pulmonary Surface Irregularity Score as a New Quantitative CT Marker for Idiopathic Pulmonary Fibrosis-a Pilot Study. J Digit Imaging 2023; 36:2382-2391. [PMID: 37670182 PMCID: PMC10584743 DOI: 10.1007/s10278-023-00896-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 09/07/2023] Open
Abstract
The purpose of this study is to evaluate the accuracy and inter-observer agreement of a quantitative pulmonary surface irregularity (PSI) score on high-resolution chest CT (HRCT) for predicting transplant-free survival in patients with IPF. For this IRB-approved HIPAA-compliant retrospective single-center study, adult patients with IPF and HRCT imaging (N = 50) and an age- and gender-matched negative control group with normal HRCT imaging (N = 50) were identified. Four independent readers measured the PSI score in the midlungs on HRCT images using dedicated software while blinded to clinical data. A t-test was used to compare the PSI scores between negative control and IPF cohorts. In the IPF cohort, multivariate cox regression analysis was used to associate PSI score and clinical parameters with transplant-free survival. Inter-observer agreement for the PSI score was assessed by intraclass correlation coefficient (ICC). The technical failure rate of the midlung PSI score was 0% (0/100). The mean PSI score of 5.38 in the IPF cohort was significantly higher than 3.14 in the negative control cohort (p < .001). In the IPF cohort, patients with a high PSI score (≥ median) were 8 times more likely to die than patients with a low PSI score (HR: 8.36; 95%CI: 2.91-24.03; p < .001). In a multivariate model including age, gender, FVC, DLCO, and PSI score, only the PSI score was associated with transplant-free survival (HR:2.11 per unit increase; 95%CI: 0.26-3.51; p = .004). Inter-observer agreement for the PSI score among 4 readers was good (ICC: 0.88; 95%CI: 0.84-0.91). The PSI score had high accuracy and good inter-observer agreement on HRCT for predicting transplant-free survival in patients with IPF.
Collapse
Affiliation(s)
- Asser M Abou Elkassem
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL, 35249, USA.
| | - Rafah Mresh
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL, 35249, USA
| | - Ahmed Farag
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL, 35249, USA
| | - Steven Rothenberg
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL, 35249, USA
| | - Seth T Lirette
- Department of Data Science, University of Mississippi Medical Center, Translational Research Center, 2500 N. State St., Jackson, MS, 39216, USA
| | - Andrew D Smith
- Department of Radiology, University of Alabama at Birmingham, 619 19th Street South, Birmingham, AL, 35249, USA
| | - Tejaswini Kulkarni
- Department of Medicine, University of Alabama at Birmingham, 1900 University Blvd, Birmingham, AL, 35294, USA
| |
Collapse
|
5
|
Fernández Pérez ER, Crooks JL, Lynch DA, Humphries SM, Koelsch TL, Swigris JJ, Solomon JJ, Mohning MP, Groshong SD, Fier K. Pirfenidone in fibrotic hypersensitivity pneumonitis: a double-blind, randomised clinical trial of efficacy and safety. Thorax 2023; 78:1097-1104. [PMID: 37028940 DOI: 10.1136/thorax-2022-219795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 03/18/2023] [Indexed: 04/09/2023]
Abstract
BACKGROUND Fibrotic hypersensitivity pneumonitis (FHP) is an irreversible lung disease with high morbidity and mortality. We sought to evaluate the safety and effect of pirfenidone on disease progression in such patients. METHODS We conducted a single-centre, randomised, double-blinded, placebo-controlled trial in adults with FHP and disease progression. Patients were assigned in a 2:1 ratio to receive either oral pirfenidone (2403 mg/day) or placebo for 52 weeks. The primary end point was the mean absolute change in the per cent predicted forced vital capacity (FVC%). Secondary end points included progression-free survival (PFS, time to a relative decline ≥10% in FVC and/or diffusing capacity of the lung for carbon monoxide (DLCO), acute respiratory exacerbation, a decrease of ≥50 m in the 6 min walk distance, increase or introduction of immunosuppressive drugs or death), change in FVC slope and mean DLCO%, hospitalisations, radiological progression of lung fibrosis and safety. RESULTS After randomising 40 patients, enrolment was interrupted by the COVID-19 pandemic. There was no significant between-group difference in FVC% at week 52 (mean difference -0.76%, 95% CI -6.34 to 4.82). Pirfenidone resulted in a lower rate of decline in the adjusted FVC% at week 26 and improved PFS (HR 0.26, 95% CI 0.12 to 0.60). Results for other secondary end points showed no significant difference between groups. No deaths occurred in the pirfenidone group and one death (respiratory) occurred in the placebo group. There were no treatment-emergent serious adverse events. CONCLUSIONS The trial was underpowered to detect a difference in the primary end point. Pirfenidone was found to be safe and improved PFS in patients with FHP. TRIAL REGISTRATION MUMBER NCT02958917.
Collapse
Affiliation(s)
| | - James L Crooks
- Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, USA
| | - David A Lynch
- Radiology, National Jewish Health, Denver, Colorado, USA
| | | | | | - Jeffrey J Swigris
- Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Joshua J Solomon
- Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Michael P Mohning
- Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | | | - Kaitlin Fier
- Clinical and Translational Research Unit, National Jewish Health, Denver, Colorado, USA
| |
Collapse
|
6
|
Soumagne T, Quétant S, Guillien A, Falque L, Hess D, Aguilaniu B, Degano B. Insight into the relationship between forced vital capacity and transfer of the lungs for carbon monoxide in patients with idiopathic pulmonary fibrosis. Respir Med Res 2023; 84:101042. [PMID: 37734234 DOI: 10.1016/j.resmer.2023.101042] [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: 11/22/2022] [Revised: 04/17/2023] [Accepted: 07/11/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND Forced vital capacity (FVC) is routinely used to quantify the severity and identify the progression of idiopathic pulmonary fibrosis (IPF). Although less commonly used, lung transfer of carbon monoxide (TLCO) correlates better with the severity of IPF than does FVC. METHODS Aiming at studying how FVC behaves in relation to TLCO, we analysed cross-sectional data from 430 IPF patients, of which 221 had at least 2 assessments (performed 2.4 ± 1.9 years apart) available for longitudinal analyses. Thresholds for identifying "abnormal" FVC and TLCO values were the statistically-defined lower limits of normal (LLN). For patients with longitudinal data, mean annual absolute declines of FVC and TLCO were calculated. RESULTS The correlation between FVC and TLCO (%predicted) was weak (R2=0.21). FVC was "abnormal" (i.e., CONCLUSION In IPF, a "normal" FVC should be viewed with caution as it is most often associated with an "abnormal" TLCO, a parameter that is strongly correlated with the morphological extent of the disease. Only 1/3 of the patients with a FVC-based progression criterion also had a TLCO progression criterion. In contrast, 2/3 of patients with a TLCO progression criterion also had a FVC progression criterion.
Collapse
Affiliation(s)
- Thibaud Soumagne
- Service de Pneumologie et Soins intensifs respiratoires, Hôpital Européen Georges-Pompidou, Assistance Publique des hôpitaux de Paris, Paris, France
| | - Sébastien Quétant
- Service Hospitalier Universitaire Pneumologie Physiologie, Pôle Thorax et Vaisseaux, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Alicia Guillien
- Épidemiologie environnementale appliquée à la reproduction et à la santé respiratoire, INSERM, CNRS, Université Grenoble Alpes, Institut pour l'Avancée des Biosciences (IAB), U1209, Grenoble, France
| | - Loïc Falque
- Service Hospitalier Universitaire Pneumologie Physiologie, Pôle Thorax et Vaisseaux, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - David Hess
- Programme Colibri, Association pour la complémentarité des connaissances et des pratiques de la pneumologie (aCCPP), 19 avenue Marcelin Berthelot, 38100, Grenoble, France
| | - Bernard Aguilaniu
- Programme Colibri, Association pour la complémentarité des connaissances et des pratiques de la pneumologie (aCCPP), 19 avenue Marcelin Berthelot, 38100, Grenoble, France; Université Grenoble Alpes, Grenoble, France
| | - Bruno Degano
- Service Hospitalier Universitaire Pneumologie Physiologie, Pôle Thorax et Vaisseaux, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France; Laboratoire HP2, INSERM U1300, Université Grenoble Alpes, Grenoble, France.
| |
Collapse
|
7
|
Richeldi L, Azuma A, Cottin V, Kreuter M, Maher TM, Martinez FJ, Oldham JM, Valenzuela C, Gordat M, Liu Y, Stowasser S, Zoz DF, Wijsenbeek MS. Design of a phase III, double-blind, randomised, placebo-controlled trial of BI 1015550 in patients with idiopathic pulmonary fibrosis (FIBRONEER-IPF). BMJ Open Respir Res 2023; 10:e001563. [PMID: 37597969 PMCID: PMC10441083 DOI: 10.1136/bmjresp-2022-001563] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 07/31/2023] [Indexed: 08/21/2023] Open
Abstract
IntroductionThere is an unmet need for new treatments for idiopathic pulmonary fibrosis (IPF). The oral preferential phosphodiesterase 4B inhibitor, BI 1015550, prevented a decline in forced vital capacity (FVC) in a phase II study in patients with IPF. This study design describes the subsequent pivotal phase III study of BI 1015550 in patients with IPF (FIBRONEER-IPF). METHODS AND ANALYSIS In this placebo-controlled, double-blind, phase III trial, patients are being randomised in a 1:1:1 ratio to receive 9 mg or 18 mg of BI 1015550 or placebo two times per day over at least 52 weeks, stratified by use of background antifibrotics (nintedanib/pirfenidone vs neither). The primary endpoint is the absolute change in FVC at week 52. The key secondary endpoint is a composite of time to first acute IPF exacerbation, hospitalisation due to respiratory cause or death over the duration of the trial. ETHICS AND DISSEMINATION The trial is being carried out in compliance with the ethical principles of the Declaration of Helsinki, in accordance with the International Council on Harmonisation Guideline for Good Clinical Practice and other local ethics committees. The results of the study will be disseminated at scientific congresses and in peer-reviewed publications. TRIAL REGISTRATION NUMBER NCT05321069.
Collapse
Affiliation(s)
- Luca Richeldi
- Unità Operativa Complessa di Pneumologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Arata Azuma
- Pulmonary Medicine and Oncology, Nippon Medical School, Tokyo, Japan
- Respiratory Medicine and Clinical Research Centre, Meisei Hospital, Saitama, Japan
| | - Vincent Cottin
- Hôpital Louis Pradel, Centre Coordonnateur National de référence des Maladies Pulmonaires Rares, Hospices Civils de Lyon, UMR754, INRAE, Université Claude Bernard Lyon 1, Member of ERN-LUNG, Lyon, France
| | - Michael Kreuter
- Centre for Interstitial and Rare Lung Diseases, Department of Pneumology, Thoraxklinik, University of Heidelberg, German Center for Lung Research, Heidelberg, Germany
- Department of Pneumology, RKH Clinic Ludwigsburg, Ludwigsburg, Germany
| | - Toby M Maher
- Department of Pulmonary, Critical Care and Sleep Medicine, USC Keck School of Medicine, Los Angeles, California, USA
- Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Justin M Oldham
- Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Claudia Valenzuela
- ILD Unit, Pulmonology Department, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maud Gordat
- Clinical Development & Operation Department, Boehringer Ingelheim, Reims, France
| | - Yi Liu
- Department of Biostatistics and Data Sciences, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Susanne Stowasser
- TA Inflammation Med, Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Donald F Zoz
- Global Clinical Development and Medical Affairs, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut, USA
| | - Marlies S Wijsenbeek
- Centre for Interstitial Lung Diseases and Sarcoidosis, Department of Respiratory Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
8
|
Bernardinello N, Cocconcelli E, Giraudo C, Daverio M, Castelli G, Petrarulo S, Bovo M, Fichera G, Cavinato S, Cattelan AM, Saetta M, Spagnolo P, Balestro E. Predictors of pulmonary sequelae after COVID-19 pneumonia: A 12-month follow-up study. Front Med (Lausanne) 2023; 10:1084002. [PMID: 36817777 PMCID: PMC9932038 DOI: 10.3389/fmed.2023.1084002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Background Since the beginning of the SARS-CoV-2 pandemic, over 550 million people have been infected worldwide. Despite these large numbers, the long-term pulmonary consequences of COVID-19 remain unclear. Aims The aim of this single-center observational cohort study was to identify and characterize pulmonary sequelae of COVID-19 at 12 months from hospitalization and to reveal possible predictors for the persistence of long-term lung consequences. Methods Based on the persistence or absence of radiological changes after 12 months from hospitalization, the whole population was categorized into NOT-RECOVERED (NOT-REC) and RECOVERED (REC) groups, respectively. Clinical and pulmonary function data tests and clinical data were also collected and compared in the two groups. In the NOT-REC group, high resolution computed tomography (HRCT) images were semiquantitatively scored analyzing ground-glass opacities (GGO), interstitial thickening (IT), consolidations (CO), linear and curvilinear band opacities, and bronchiectasis for each lung lobe. Logistic regression analyses served to detect the factors associated with 12-month radiological consequences. Results Out of the 421 patients followed after hospitalization for SARS-CoV-2 pneumonia, 347 met inclusion and exclusion criteria and were enrolled in the study. The NOT-REC patients (n = 24; 6.9%) were significantly older [67 (62-76) years vs. 63 (53-71) years; p = 0.02], more frequently current smokers [4 (17%) vs. 12 (4%); p = 0.02], and with more severe respiratory failure at the time of hospitalization [PaO2/FiO2 at admission: 201 (101-314) vs. 295 (223-343); p = 0.01] compared to REC group (n = 323; 93.1%). On multivariable analysis, being a current smoker resulted in an independent predictor for lung sequelae after 12 months from hospitalization [5.6 OR; 95% CI (1.41-22.12); p = 0.01]. Conclusion After 12 months from hospital admission, a limited number of patients displayed persistent pulmonary sequelae with minimal extension. Being a current smoker at the time of SARS-CoV-2 infection is an independent predictive factor to lung consequences, regardless of the disease severity.
Collapse
Affiliation(s)
- Nicol Bernardinello
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, Padova, Italy
| | - Elisabetta Cocconcelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, Padova, Italy
| | - Chiara Giraudo
- Department of Medicine, Institute of Radiology, University of Padova and Padova City Hospital, Padova, Italy
| | - Matteo Daverio
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, Padova, Italy
| | - Gioele Castelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, Padova, Italy
| | - Simone Petrarulo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, Padova, Italy
| | - Matteo Bovo
- School of Medicine and Surgery, University of Padova, Padova, Italy
| | - Giulia Fichera
- Pediatric Radiology Unit, University of Padova and Padova City Hospital, Padova, Italy
| | - Silvia Cavinato
- Division of Infectious and Tropical Diseases, University of Padova and Padova City Hospital, Padova, Italy
| | - Anna Maria Cattelan
- Division of Infectious and Tropical Diseases, University of Padova and Padova City Hospital, Padova, Italy
| | - Marina Saetta
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, Padova, Italy
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, Padova, Italy
| | - Elisabetta Balestro
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova and Padova City Hospital, Padova, Italy,*Correspondence: Elisabetta Balestro,
| |
Collapse
|
9
|
Mai TH, Han LW, Hsu JC, Kamath N, Pan L. Idiopathic pulmonary fibrosis therapy development: a clinical pharmacology perspective. Ther Adv Respir Dis 2023; 17:17534666231181537. [PMID: 37392011 PMCID: PMC10333628 DOI: 10.1177/17534666231181537] [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: 11/08/2022] [Accepted: 05/26/2023] [Indexed: 07/02/2023] Open
Abstract
Drug development for idiopathic pulmonary fibrosis (IPF) has been challenging due to poorly understood disease etiology, unpredictable disease progression, highly heterogeneous patient populations, and a lack of robust pharmacodynamic biomarkers. Moreover, because lung biopsy is invasive and dangerous, making the extent of fibrosis as a direct longitudinal measurement of IPF disease progression unfeasible, most clinical trials studying IPF can only assess progression of fibrosis indirectly through surrogate measures. This review discusses current state-of-art practices, identifies knowledge gaps, and brainstorms development opportunities for preclinical to clinical translation, clinical populations, pharmacodynamic endpoints, and dose optimization strategies. This article highlights clinical pharmacology perspectives in leveraging real-world data as well as modeling and simulation, special population considerations, and patient-centric approaches for designing future studies.
Collapse
Affiliation(s)
- Tu H. Mai
- Genentech Inc., South San Francisco, CA,
USA
| | | | - Joy C. Hsu
- Genentech Inc., South San Francisco, CA,
USA
| | | | - Lin Pan
- Genentech, Inc., 1 DNA Way, South San
Francisco, CA 94008, USA
| |
Collapse
|
10
|
Harari S, Wells AU, Wuyts WA, Nathan SD, Kirchgaessler KU, Bengus M, Behr J. The 6-min walk test as a primary end-point in interstitial lung disease. Eur Respir Rev 2022; 31:31/165/220087. [PMID: 36002171 DOI: 10.1183/16000617.0087-2022] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/14/2022] [Indexed: 12/23/2022] Open
Abstract
There is a need for clinical trial end-points to better assess how patients feel and function, so that interventions can be developed which alleviate symptoms and improve quality of life. Use of 6-min walk test (6MWT) outcomes as a primary end-point in interstitial lung disease (ILD) trials is growing, particularly for drugs targeting concurrent pulmonary hypertension. However, 6MWT outcomes may be influenced differentially by interstitial lung and pulmonary vascular components of ILD, making interpretation complicated. We propose that using 6MWT outcomes, including 6-min walk distance or oxygen desaturation, as primary end-points should depend upon the study population (how advanced the ILD is; whether vasculopathy is significant), the degree of disease progression, and, importantly, the effect of study treatment expected. We argue that the 6MWT as a single outcome measure is suitable as a primary end-point if the treatment goal is to improve functional performance or prevent disease progression within a study population of patients with advanced ILD or those with ILD and co-existent vasculopathy. In addition, we discuss the potential of composite primary end-points incorporating 6MWT outcomes, outlining important considerations to ensure that they are appropriate for the study population and treatment goals.
Collapse
Affiliation(s)
- Sergio Harari
- Dept of Clinical Sciences and Community Health, University of Milan, Milan, Italy.,U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Ospedale San Guiseppe, MultiMedica IRCCS, Milan, Italy
| | - Athol U Wells
- Interstitial Lung Disease Unit, Royal Brompton Hospital, London, UK
| | - Wim A Wuyts
- Unit for Interstitial Lung Diseases, University of Leuven, Leuven, Belgium
| | - Steven D Nathan
- The Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, VA, USA
| | | | | | - Jürgen Behr
- Dept of Medicine V, University Hospital, LMU Munich, Member of the German Center for Lung Research (DZL), Munich, Germany .,Asklepios Fachkliniken München-Gauting, Comprehensive Pneumology Center, Munich, Germany
| |
Collapse
|
11
|
Xie J, Hu Y, Sun D, Liu C, Li Z, Zhu J. Targeting non-coding RNA H19: A potential therapeutic approach in pulmonary diseases. Front Pharmacol 2022; 13:978151. [PMID: 36188624 PMCID: PMC9523668 DOI: 10.3389/fphar.2022.978151] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Non-coding RNA is still one of the most popular fields in biology research. In recent years, people paid more attention to the roles of H19 in lung diseases, which expressed abnormally in various pathological process. Therefore, this review focus on the regulatory role of H19 in asthma, pulmonary arterial hypertension (PAH), idiopathic pulmonary fibrosis (IPF), lung injury, pneumonia, lung cancer, etc. And the potential therapeutic agents and molecular treatments of H19 are collected. The aim is to demonstrate its underlying mechanism in pulmonary diseases and to guide the basic research targeting H19 into clinical drug translation.
Collapse
Affiliation(s)
- Jinghui Xie
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yuedi Hu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Dengdi Sun
- The Key Laboratory of Intelligent Computing and Signal Processing (ICSP), Ministry of Education, School of Artificial Intelligence, Anhui University, Hefei, China
| | - Changan Liu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Zegeng Li
- Institute of Traditional Chinese Medicine Prevention and Control on Respiratory Disease, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Department of Respiratory Medicine, First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Jie Zhu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Institute of Traditional Chinese Medicine Prevention and Control on Respiratory Disease, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Jouneau S, Rousseau C, Lederlin M, Lescoat A, Kerjouan M, Chauvin P, Luque-Paz D, Guillot S, Oger E, Vernhet L, Thibault. R. Malnutrition and decreased food intake at diagnosis are associated with hospitalization and mortality of idiopathic pulmonary fibrosis patients. Clin Nutr 2022; 41:1335-1342. [DOI: 10.1016/j.clnu.2022.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/06/2022] [Accepted: 05/02/2022] [Indexed: 11/03/2022]
|
13
|
Morrow LE, Hilleman D, Malesker MA. Management of patients with fibrosing interstitial lung diseases. Am J Health Syst Pharm 2022; 79:129-139. [PMID: 34608488 PMCID: PMC8881211 DOI: 10.1093/ajhp/zxab375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
PURPOSE This article summarizes the appropriate use and pharmacology of treatments for fibrosing interstitial lung diseases, with a specific focus on the antifibrotic agents nintedanib and pirfenidone. SUMMARY The interstitial lung diseases are a heterogenous group of parenchymal lung disorders with a common feature-infiltration of the interstitial space with derangement of the normal capillary-alveolar anatomy. Diseases characterized by fibrosis of the interstitial space are referred to as the fibrosing interstitial lung diseases and often show progression over time: idiopathic pulmonary fibrosis is the most common fibrotic interstitial lung disease. Historically, therapies for fibrosing lung diseases have been limited in number, questionable in efficacy, and associated with potential harms. Food and Drug Administration (FDA) approval of the antifibrotic agents nintedanib and pirfenidone for idiopathic pulmonary fibrosis in 2014 heralded an era of reorganization of therapy for the fibrotic interstitial lung diseases. Subsequent investigations have led to FDA approval of nintedanib for systemic sclerosis-associated interstitial lung disease and interstitial lung diseases with a progressive phenotype. Although supportive care and pulmonary rehabilitation should be provided to all patients, the role(s) of immunomodulators and/or immune suppressing agents vary by the underlying disease state. Several agents previously used to treat fibrotic lung diseases (N-acetylcysteine, anticoagulation, and pulmonary vasodilators) lack efficacy or cause harm. CONCLUSION With the introduction of effective pharmacotherapy for fibrosing interstitial lung disease, pharmacists have an increasingly important role in the interdisciplinary team managing these patients.
Collapse
Affiliation(s)
- Lee E Morrow
- Creighton University School of Medicine, Omaha, NE
- Creighton University School of Pharmacy and Health Professions, Omaha, NE, USA
| | - Daniel Hilleman
- Creighton University School of Pharmacy and Health Professions, Omaha, NE
- Creighton University School of Medicine, Omaha, NE, USA
| | - Mark A Malesker
- Creighton University School of Pharmacy and Health Professions, Omaha, NE
- Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
14
|
Liu H, Li Y, Zou Y, Zhang X, Shi X, Yin Z, Lin Y. Influence of miRNA-30a-5p on Pulmonary Fibrosis in Mice with Streptococcus pneumoniae Infection through Regulation of Autophagy by Beclin-1. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9963700. [PMID: 34604389 PMCID: PMC8486528 DOI: 10.1155/2021/9963700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/06/2021] [Indexed: 11/17/2022]
Abstract
The study is aimed at observing the influence of microribonucleic acid- (miRNA-) 30a-50p on the pulmonary fibrosis in mice with Streptococcus pneumoniae infection through the regulation of autophagy by Beclin-1. Specific pathogen-free mice were instilled with Streptococcus pneumoniae through the trachea to establish the pulmonary fibrosis model. Then, they were divided into the miRNA-30a-50p mimics group (mimics group, n = 10) and miRNA-30a-5p inhibitors group (inhibitors group, n = 10), with the control group (n = 10) also set. Pulmonary tissue wet weight/dry weight (W/D) was detected. The content of tumor necrosis factor-α (TNF-α), interleukin- (IL-) 6, and myeloperoxidase (MPO) was determined using enzyme-linked immunosorbent assay (ELISA). Besides, the changes in the pulmonary function index dynamic lung compliance (Cdyn), plateau pressure (Pplat), and peak airway pressure (Ppeak) were monitored, and the gene and protein expression levels were measured via quantitative PCR (qPCR) and Western blotting. The expression level of miRNA-30a-5p was substantially raised in the mimics group (p < 0.05), but extremely low in the inhibitors group (p < 0.05). The mimics group had obviously raised levels of serum aminotransferase (AST), glutamic-pyruvic transaminase (GPT), alkaline phosphatase (ALP), and pulmonary tissue W/D (p < 0.05). Additionally, the expression levels of TNF-α, IL-6, and MPO were notably elevated in the mimics group, while their expression levels showed the opposite conditions in the inhibitors group (p < 0.05). According to the HE staining results, the inhibitors group had arranged orderly cells, while the mimics group exhibited lung injury, pulmonary edema, severe inflammatory response, and alveolar congestion. In the inhibitors group, Cdyn was remarkably elevated, but Pplat and Ppeak declined considerably (p < 0.05). Besides, the inhibitors group exhibited elevated messenger RNA (mRNA) levels of Beclin-1 and LC3, lowered mRNA levels of α-SMA and p62, a raised protein level of Beclin-1, and a markedly decreased protein level of p62 (p < 0.05). Silencing miRNA-30a-5p expression can promote the expression of Beclin-1 to accelerate the occurrence of autophagy, thereby treating pulmonary fibrosis in mice with Streptococcus pneumoniae infection.
Collapse
Affiliation(s)
- Hanyu Liu
- Department of Laboratory, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, China
| | - Yabo Li
- Department of Laboratory, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, China
| | - Yingdong Zou
- Department of Laboratory, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, China
| | - Xingzong Zhang
- Department of Laboratory, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, China
| | - Xiongfei Shi
- Department of Laboratory, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, China
| | - Zhiping Yin
- Department of Laboratory, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, China
| | - Yun Lin
- Department of Laboratory, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, China
| |
Collapse
|
15
|
Farrand E, Limper AH. Clinical Trials for Idiopathic Pulmonary Fibrosis and the Role of Health Systems. Clin Chest Med 2021; 42:287-294. [PMID: 34024404 DOI: 10.1016/j.ccm.2021.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We are in the midst of transformative innovation in health care delivery and clinical trials in idiopathic pulmonary fibrosis (IPF). Health systems are uniquely positioned at the crossroad of these shifting paradigms, equipped with the resources to expand the research pipeline in IPF through visionary leadership and targeted investments. The authors hope that by prioritizing development of health information technology, supporting a broader range of clinical trial designs, and cultivating broad stakeholder engagement, health systems will generate data to address knowledge-evidence-practice gaps in IPF. This will continue to improve the ability to deliver high-quality, safe, and effective care.
Collapse
Affiliation(s)
- Erica Farrand
- 505 Parnassus Avenue, Room M1083, Box 0111, San Francisco, CA 94117, USA.
| | | |
Collapse
|
16
|
Kalluri M, Luppi F, Vancheri A, Vancheri C, Balestro E, Varone F, Mogulkoc N, Cacopardo G, Bargagli E, Renzoni E, Torrisi S, Calvello M, Libra A, Pavone M, Bonella F, Cottin V, Valenzuela C, Wijsenbeek M, Bendstrup E. Patient-reported outcomes and patient-reported outcome measures in interstitial lung disease: where to go from here? Eur Respir Rev 2021; 30:30/160/210026. [PMID: 34039675 PMCID: PMC9488962 DOI: 10.1183/16000617.0026-2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/02/2021] [Indexed: 12/03/2022] Open
Abstract
Patient-reported outcome measures (PROMs), tools to assess patient self-report of health status, are now increasingly used in research, care and policymaking. While there are two well-developed disease-specific PROMs for interstitial lung diseases (ILD) and idiopathic pulmonary fibrosis (IPF), many unmet and urgent needs remain. In December 2019, 64 international ILD experts convened in Erice, Italy to deliberate on many topics, including PROMs in ILD. This review summarises the history of PROMs in ILD, shortcomings of the existing tools, challenges of development, validation and implementation of their use in clinical trials, and the discussion held during the meeting. Development of disease-specific PROMs for ILD including IPF with robust methodology and validation in concordance with guidance from regulatory authorities have increased user confidence in PROMs. Minimal clinically important difference for bidirectional changes may need to be developed. Cross-cultural validation and linguistic adaptations are necessary in addition to robust psychometric properties for effective PROM use in multinational clinical trials. PROM burden of use should be reduced through appropriate use of digital technologies and computerised adaptive testing. Active patient engagement in all stages from development, testing, choosing and implementation of PROMs can help improve probability of success and further growth. PROMs are essential tools for research and care in ILD and IPF. They report patient perceptions of the impact of disease and its treatments on whole-person wellbeing and can guide research to make care more patient-centred.https://bit.ly/3s7Y0a8
Collapse
Affiliation(s)
- Meena Kalluri
- Division of Pulmonary Medicine, University of Alberta, Edmonton, AB, Canada .,Shared first and last authorship
| | - Fabrizio Luppi
- Respiratory Diseases Unit, University of Milano-Bicocca. "S. Gerardo" Hospital, Monza, Italy.,Shared first and last authorship
| | - Ada Vancheri
- Regional Referral Center for Rare Lung Diseases, University - Hospital "Policlinico G. Rodolico - San Marco", Catania, Italy
| | - Carlo Vancheri
- Dept of Clinical and Experimental Medicine, Regional Referral Centre for Rare Lung Diseases, University - Hospital "Policlinico G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Elisabetta Balestro
- Dept of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Francesco Varone
- UOC Pneumologia, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Nesrin Mogulkoc
- Dept of Pulmonology, Ege University Hospiral, Bornova, Izmir, Turkey
| | - Giulia Cacopardo
- UOSD UTIR, A.R.N.A.S. Ospedali Civico Di Cristina Benfratelli, Palermo, Italy
| | - Elena Bargagli
- Respiratory Diseases Unit, Siena University, Siena, Italy
| | - Elizabeth Renzoni
- Interstitial Lung Disease Unit, Royal Brompton Hospital, Imperial College, London, UK
| | - Sebastiano Torrisi
- Dept of Clinical and Experimental Medicine, Regional Referral Centre for Rare Lung Diseases, University - Hospital "Policlinico G. Rodolico - San Marco", University of Catania, Catania, Italy
| | | | - Alessandro Libra
- Regional Referral Center for Rare Lung Diseases, University - Hospital "Policlinico G. Rodolico - San Marco", Catania, Italy
| | - Mauro Pavone
- Dept of Clinical and Experimental Medicine, Regional Referral Centre for Rare Lung Diseases, University - Hospital "Policlinico G. Rodolico - San Marco", University of Catania, Catania, Italy
| | - Francesco Bonella
- Pneumology Dept, Centre for Interstitial and Rare Lung Disease, Ruhrlandklinik University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Vincent Cottin
- Dept of Respiratory Medicine, National Reference Coordinating Center for Rare Pulmonary Diseases, Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France.,University of Lyon, INRAE, IVPC, Lyon, France
| | - Claudia Valenzuela
- Pulmonology Dept, Hospital Universitario de la Princesa, Universidad Autonoma Madrid, Madrid, Spain.,Shared first and last authorship
| | - Marlies Wijsenbeek
- Centre of excellence, Interstitial Lung Diseases and Sarcoidosis, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.,Shared first and last authorship
| | - Elisabeth Bendstrup
- Dept of Respiratory Diseases and Allergy, Centre for Rare Lung Diseases, Aarhus University Hospital, Aarhus N, Denmark.,Shared first and last authorship
| | | |
Collapse
|
17
|
Lancaster L, Fieuw A, Meulemans J, Ford P, Nathan SD. Standardization of the 6-min walk test in clinical trials of idiopathic pulmonary fibrosis. Contemp Clin Trials 2020; 100:106227. [PMID: 33246099 DOI: 10.1016/j.cct.2020.106227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 11/27/2022]
Abstract
The 6-min walk test (6MWT) is an important measure of functional capacity in idiopathic pulmonary fibrosis (IPF) and has been an endpoint of several IPF clinical trials. However, current guidance for the 6MWT offers insufficient advice on standardization, particularly oxygen supplementation, for clinical trials. Three physicians experienced with the 6MWT and IPF developed a standardized protocol for the 6MWT based on existing clinical guidelines and published literature. The protocol comprises guidance on test conditions, pre-defined parameters to measure at specified timepoints, and step-by-step instructions on conducting the test. The standardized test will be evaluated in the large-scale phase 3 ISABELA trials (NCT03711162; NCT03733444). The test is conducted indoors, using standardized equipment, along a flat, straight, 30-m unobstructed corridor; tests for each individual are performed by the same administrators at the same time of day; warm-up prior to testing is prohibited; supplemental oxygen tanks are permitted and moved by the patient in the same manner for each test; precise wording is used to instruct and encourage patients. Contraindications and stopping criteria are specified. Key assessments include: 6-min walk distance, distance walked at 1 and 3 min, the Borg CR10 scale, heart rate, blood pressure, and oxygen desaturation levels. A standardized 6MWT for IPF will enable more reliable comparisons between clinical trials and limit variability, optimizing use as an endpoint. Application of the standardized 6MWT in the ISABELA program will allow its correlation with other clinically important endpoints and may lead to novel composite endpoints for use in future trials. Submission category: Study Design, Statistical Design, Study Protocols. Submission classifications: Clinical study methodology; Clinical trial design; Clinical trials; Pulmonary disease; Pulmonary disease clinical trial; Respiratory medicine.
Collapse
Affiliation(s)
- Lisa Lancaster
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Ann Fieuw
- Clinical Research, Galapagos NV, Mechelen, Belgium
| | | | - Paul Ford
- Clinical Development, Galapagos NV, Mechelen, Belgium
| | - Steven D Nathan
- Advanced Lung Disease and Transplant Program, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, VA, USA
| |
Collapse
|
18
|
Barratt SL, Davis R, Sharp C, Pauling JD. The prognostic value of cardiopulmonary exercise testing in interstitial lung disease: a systematic review. ERJ Open Res 2020; 6:00027-2020. [PMID: 32832530 PMCID: PMC7430148 DOI: 10.1183/23120541.00027-2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/22/2020] [Indexed: 01/05/2023] Open
Abstract
The heterogeneity of interstitial lung disease (ILD) results in prognostic uncertainty concerning end-of-life discussions and optimal timing for transplantation. Effective prognostic markers and prediction models are needed. Cardiopulmonary exercise testing (CPET) provides a comprehensive assessment of the physiological changes in the respiratory, cardiovascular and musculoskeletal systems in a controlled laboratory environment. It has shown promise as a prognostic factor for other chronic respiratory conditions. We sought to evaluate the prognostic value of CPET in predicting outcomes in longitudinal studies of ILD. MEDLINE, Embase and the Cochrane Database of Systematic Reviews were used to identify studies reporting the prognostic value of CPET in predicting outcomes in longitudinal studies of ILD. Study quality was assessed using the Quality in Prognosis Study risk of bias tool. Thirteen studies were included that reported the prognostic value of CPET in ILD. All studies reported at least one CPET parameter predicting clinical outcomes in ILD, with survival being the principal outcome assessed. Maximum oxygen consumption, reduced ventilatory efficiency and exercise-induced hypoxaemia were all reported to have prognostic value in ILD. Issues with study design (primarily due to inherent problems of retrospective studies, patient selection and presentation of numerous CPET parameters), insufficient adjustment for important confounders and inadequate statistical analyses limit the strength of the conclusions that can be drawn at this stage. There is insufficient evidence to confirm the value of CPET in facilitating “real-world” clinical decisions in ILD. Additional prospective studies are required to validate the putative prognostic associations reported in previous studies in carefully phenotyped patient populations. There is presently insufficient evidence to confirm the value of CPET in facilitating “real-world” clinical decisions in ILD. Additional prospective studies are required to validate the putative prognostic associations reported in previous studies.https://bit.ly/3dfp5kq
Collapse
Affiliation(s)
- Shaney L Barratt
- Academic Respiratory Unit, School of Clinical Sciences, University of Bristol, Bristol, UK.,Bristol Interstitial Lung Disease Service, North Bristol NHS Trust, Westbury-on-Trym, Bristol, UK
| | - Richard Davis
- Academic Respiratory Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Charles Sharp
- Gloucestershire Hospitals NHS Foundation Trust, Gloucester, UK
| | - John D Pauling
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK.,Royal National Hospital for Rheumatic Diseases, Royal United Hospitals NHS Foundation Trust, Bath, UK
| |
Collapse
|
19
|
Hoyer N, Thomsen LH, Wille MMW, Wilcke T, Dirksen A, Pedersen JH, Saghir Z, Ashraf H, Shaker SB. Increased respiratory morbidity in individuals with interstitial lung abnormalities. BMC Pulm Med 2020; 20:67. [PMID: 32188453 PMCID: PMC7081690 DOI: 10.1186/s12890-020-1107-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/03/2020] [Indexed: 01/26/2023] Open
Abstract
Background Interstitial lung abnormalities (ILA) are common in participants of lung cancer screening trials and broad population-based cohorts. They are associated with increased mortality, but less is known about disease specific morbidity and healthcare utilisation in individuals with ILA. Methods We included all participants from the screening arm of the Danish Lung Cancer Screening Trial with available baseline CT scan data (n = 1990) in this cohort study. The baseline scan was scored for the presence of ILA and patients were followed for up to 12 years. Data about all hospital admissions, primary healthcare visits and medicine prescriptions were collected from the Danish National Health Registries and used to determine the participants’ disease specific morbidity and healthcare utilisation using Cox proportional hazards models. Results The 332 (16.7%) participants with ILA were more likely to be diagnosed with one of several respiratory diseases, including interstitial lung disease (HR: 4.9, 95% CI: 1.8–13.3, p = 0.008), COPD (HR: 1.7, 95% CI: 1.2–2.3, p = 0.01), pneumonia (HR: 2.0, 95% CI: 1.4–2.7, p < 0.001), lung cancer (HR: 2.7, 95% CI: 1.8–4.0, p < 0.001) and respiratory failure (HR: 1.8, 95% CI: 1.1–3.0, p = 0.03) compared with participants without ILA. These findings were confirmed by increased hospital admission rates with these diagnoses and more frequent prescriptions for inhalation medicine and antibiotics in participants with ILA. Conclusions Individuals with ILA are more likely to receive a diagnosis and treatment for several respiratory diseases, including interstitial lung disease, COPD, pneumonia, lung cancer and respiratory failure during long-term follow-up.
Collapse
Affiliation(s)
- Nils Hoyer
- Department of Respiratory Medicine, Herlev and Gentofte Hospital, Kildegårdsvej 28, 2900 Hellerup, Copenhagen, Denmark.
| | - Laura H Thomsen
- Department of Respiratory Medicine, Amager and Hvidovre Hospital, Copenhagen, Denmark
| | | | - Torgny Wilcke
- Department of Respiratory Medicine, Herlev and Gentofte Hospital, Kildegårdsvej 28, 2900 Hellerup, Copenhagen, Denmark
| | - Asger Dirksen
- Department of Respiratory Medicine, Herlev and Gentofte Hospital, Kildegårdsvej 28, 2900 Hellerup, Copenhagen, Denmark
| | - Jesper H Pedersen
- Department of Cardiothoracic Surgery RT, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Zaigham Saghir
- Department of Respiratory Medicine, Herlev and Gentofte Hospital, Kildegårdsvej 28, 2900 Hellerup, Copenhagen, Denmark
| | - Haseem Ashraf
- Department of Radiology, Akershus University Hospital, Loerenskog, Norway.,Division of Medicine and Laboratory Sciences, University of Oslo, Oslo, Norway
| | - Saher B Shaker
- Department of Respiratory Medicine, Herlev and Gentofte Hospital, Kildegårdsvej 28, 2900 Hellerup, Copenhagen, Denmark
| |
Collapse
|
20
|
Hagmeyer L, Herkenrath S, Anduleit N, Treml M, Randerath W. Cardiopulmonary Exercise Testing Allows Discrimination Between Idiopathic Non-specific Interstitial Pneumonia and Idiopathic Pulmonary Fibrosis in Mild to Moderate Stages of the Disease. Lung 2019; 197:721-726. [PMID: 31676976 DOI: 10.1007/s00408-019-00282-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 10/15/2019] [Indexed: 10/25/2022]
Abstract
It is unclear whether there are cardiopulmonary exercise testing (CPET) parameters which may indicate poor prognosis in the early course of fibrosing interstitial lung disease. 27 untreated consecutive subjects (13 idiopathic non-specific interstitial pneumonia (iNSIP), 14 idiopathic pulmonary fibrosis (IPF); 19 male; age 69 ± 10 years) were enrolled in this observational pilot study. Subjects underwent routine pulmonary function testing and CPET. Statistically, the t test and the Mann-Whitney-U test were applied in the presence of normal and non-normal distribution (according to Shapiro-Wilk), respectively. Analyzing the whole cohort, only mild functional impairments were determined. Comparison of iNSIP and IPF groups detected significant differences for the CPET parameters V'O2Peak[%pred] (p = 0.011), V'O2/kgPeak (p = 0.033), Watt[%pred] (p = 0.048), V'E/V'CO2 (Rest: p = 0.016; AT: p = 0.011; Peak: p = 0.019; Slope: p = 0.040), V'E/V'O2 (Rest: p = 0.033 AT: p = 0.014; Peak: p = 0.035). CPET parameters may indicate IPF-specific impairments even in mild disease. It may be hypothesized that these parameters are early biomarkers of poor prognosis.
Collapse
Affiliation(s)
- Lars Hagmeyer
- Hospital Bethanien Solingen, Institute of Pneumology, University of Cologne, Solingen, Germany. .,Hospital Bethanien Solingen, Clinic of Pneumology and Allergology, Center for Sleep Medicine and Respiratory Care, Aufderhöher Straße 169-175, 42699, Solingen, Germany.
| | - Simon Herkenrath
- Hospital Bethanien Solingen, Institute of Pneumology, University of Cologne, Solingen, Germany.,Hospital Bethanien Solingen, Clinic of Pneumology and Allergology, Center for Sleep Medicine and Respiratory Care, Aufderhöher Straße 169-175, 42699, Solingen, Germany
| | - Norbert Anduleit
- Hospital Bethanien Solingen, Institute of Pneumology, University of Cologne, Solingen, Germany
| | - Marcel Treml
- Hospital Bethanien Solingen, Institute of Pneumology, University of Cologne, Solingen, Germany
| | - Winfried Randerath
- Hospital Bethanien Solingen, Institute of Pneumology, University of Cologne, Solingen, Germany.,Hospital Bethanien Solingen, Clinic of Pneumology and Allergology, Center for Sleep Medicine and Respiratory Care, Aufderhöher Straße 169-175, 42699, Solingen, Germany
| |
Collapse
|
21
|
Mycophenolate mofetil as an alternative treatment in sarcoidosis. Pulm Pharmacol Ther 2019; 58:101840. [PMID: 31518648 DOI: 10.1016/j.pupt.2019.101840] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION In sarcoidosis although no better drug therapy than corticosteroids (CS) has emerged, alternative immunosuppressive agents are used when indicated. Mycophenolate mofetil (MMF) presents rapid action, a considerable safety profile and absence of lung toxicity. Few data exist so far on its use in patients with sarcoidosis. This is a retrospective study on the effectiveness and safety of MMF in patients with sarcoidosis. MATERIALS AND METHODS All patients with biopsy proven sarcoidosis treated for at least 1 year with MMF from 2008 to 2017 in our department are evaluated. RESULTS Eight patients with both pulmonary and extrapulmonary disease are included in the analysis. During follow-up, symptoms and chest radiological findings improved in all. A statistically significant improvement of FEV1 and FVC is reported (p = 0.010 and p = 0.021 respectively). Cardiac and renal disease resolved during treatment while dermal disease significantly improved. MMF permitted CS dose reduction from 15.0 (10.0, 35.0) to 2.5 (0.0, 5.0) mg prednisolone (or equivalent), p = 0.016. All patients but one, tolerated well MMF. CONCLUSION MMF as an alternative drug in systemic sarcoidosis, proved safe and effective, permitting the reduction of the dose of oral CS and leading to clinical, functional and radiological improvement.
Collapse
|
22
|
Harari S, Caminati A, Confalonieri M, Poletti V, Vancheri C, Pesci A, Rogliani P, Luppi F, Agostini C, Rottoli P, Sanduzzi Zamparelli A, Sebastiani A, Della Porta R, Salton F, Messore B, Tomassetti S, Rosso R, Biffi A, Puxeddu E, Cerri S, Cinetto F, Refini RM, Bocchino M, Di Michele L, Specchia C, Albera C. The prognostic role of Gender-Age-Physiology system in idiopathic pulmonary fibrosis patients treated with pirfenidone. CLINICAL RESPIRATORY JOURNAL 2019; 13:166-173. [PMID: 30675755 DOI: 10.1111/crj.12999] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 01/05/2019] [Accepted: 01/15/2019] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Gender, age, physiology (GAP) system have proven to be an easy tool for predicting disease stages and survival in idiopathic pulmonary fibrosis (IPF) patients. OBJECTIVE To validate mortality risk as determined by the GAP system in a real-life multicentre IPF population treated with pirfenidone. METHODS The study included patients who received pirfenidone for at least 6 months. The GAP calculator and the GAP index were determined. The primary outcome was all-cause mortality. The prognostic accuracy of the GAP system was evaluated with respect to calibration and discrimination. RESULTS AND CONCLUSION Sixty-eight IPF patients were enrolled in the study. The median follow-up was 2.4 years (range 0.1-7.4 years). A total of 22 deaths as first event (32%) and of 10 lung transplantation (15%) were recorded. The cumulative incidence of mortality at 1, 2 and 3 years was 10.4%, 22.4% and 38.4%, respectively. The differences between the predicted and observed mortality were not significant for the GAP index while the observed mortality become comparable to that predicted by the GAP calculator only in the third year of follow-up. The C-index for the GAP index was 0.74 (95% CI 0.57-0.93) while the C-statistic value for the GAP calculator was 0.77 (95% CI 0.59-0.95).
Collapse
Affiliation(s)
- Sergio Harari
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria - Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare. Ospedale San Giuseppe - MultiMedica, IRCCS, Milano, Italy
| | - Antonella Caminati
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria - Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare. Ospedale San Giuseppe - MultiMedica, IRCCS, Milano, Italy
| | - Marco Confalonieri
- Department of Pulmonology, University Hospital of Cattinara, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Trieste, Trieste, Italy
| | - Venerino Poletti
- U.O. di Pneumologia Dipartimento dell'Apparato Respiratorio e del Torace, Ospedale G.P. Morgagni -L. Pierantoni, Forlì, Italy.,Department of Respiratory Diseases & Allergy, Aarhus University Hospital (DK), Aarthus, Denmark
| | - Carlo Vancheri
- Regional Referral Centre for Rare Lung Disease, University of Catania, A.O.U. Policlinico-Vittorio Emanuele, Catania, Italy
| | - Alberto Pesci
- Respiratory Unit, Department of Health Science, University of Milano Bicocca, AO San Gerardo, Monza, Italy
| | - Paola Rogliani
- Respiratory Unit Policlinico Tor Vergata, Department of "Systems Medicine, University of Rome "Tor Vergata", Roma, Italy
| | - Fabrizio Luppi
- Center for Rare Lung Diseases, University Hospital Policlinico di Modena, Modena, Italy
| | - Carlo Agostini
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Paola Rottoli
- Respiratory Diseases and Lung Transplant Unit, Department of Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Alessandro Sanduzzi Zamparelli
- UOC II Pneumotisiologia, Scuola di specializzazione in malattie respiratorie Università degli Studi di Napoli Federico II A.O.R.N. Monaldi-Cotugno-CTO Piazzale Ettore Ruggieri, Napoli, Italy
| | - Alfredo Sebastiani
- UOS Interstiziopatie Polmonari Az Osp. S. Camillo-Forlanini, Roma, Italy
| | - Rossana Della Porta
- Department of Pulmonology, University Hospital of Cattinara, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Trieste, Trieste, Italy
| | - Francesco Salton
- Department of Pulmonology, University Hospital of Cattinara, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Trieste, Trieste, Italy
| | - Barbara Messore
- Department of Clinical and Biological Sciences, Interstitial and Rare Lung Disease Unit AOU San Luigi Gonzaga, Orbassano, University of Turin, Turin, Italy
| | - Sara Tomassetti
- U.O. di Pneumologia Dipartimento dell'Apparato Respiratorio e del Torace, Ospedale G.P. Morgagni -L. Pierantoni, Forlì, Italy
| | - Roberta Rosso
- Regional Referral Centre for Rare Lung Disease, University of Catania, A.O.U. Policlinico-Vittorio Emanuele, Catania, Italy
| | - Alice Biffi
- Respiratory Unit, Department of Health Science, University of Milano Bicocca, AO San Gerardo, Monza, Italy
| | - Ermanno Puxeddu
- Respiratory Unit Policlinico Tor Vergata, Department of "Systems Medicine, University of Rome "Tor Vergata", Roma, Italy
| | - Stefania Cerri
- Center for Rare Lung Diseases, University Hospital Policlinico di Modena, Modena, Italy
| | | | - Rosa Metella Refini
- Respiratory Diseases and Lung Transplant Unit, Department of Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Marialuisa Bocchino
- UOC II Pneumotisiologia, Scuola di specializzazione in malattie respiratorie Università degli Studi di Napoli Federico II A.O.R.N. Monaldi-Cotugno-CTO Piazzale Ettore Ruggieri, Napoli, Italy
| | - Loreta Di Michele
- UOS Interstiziopatie Polmonari Az Osp. S. Camillo-Forlanini, Roma, Italy
| | - Claudia Specchia
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,IRCCS MultiMedica, Milano, Italy
| | - Carlo Albera
- Department of Clinical and Biological Sciences, Interstitial and Rare Lung Disease Unit AOU San Luigi Gonzaga, Orbassano, University of Turin, Turin, Italy
| | | |
Collapse
|
23
|
Ratwani AP, Ahmad KI, Barnett SD, Nathan SD, Brown AW. Connective tissue disease-associated interstitial lung disease and outcomes after hospitalization: A cohort study. Respir Med 2019; 154:1-5. [PMID: 31176795 DOI: 10.1016/j.rmed.2019.05.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/16/2019] [Accepted: 05/28/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND The impact of hospitalization on patient outcomes is increasingly recognized and considered in the prognostication of many pulmonary disorders. We sought to evaluate the impact of hospitalization on survival in connective tissue disease-interstitial lung disease (CTD-ILD) patients. METHODS A chart review of patients with CTD-ILD followed at a tertiary care center was performed. Patients were stratified into two groups based on hospitalization status. Outcomes of the groups were compared using Kaplan-Meier survival analyses as well as multivariate competing risk analysis. RESULTS There were 137 patients identified with confirmed CTD-ILD. Patients who underwent hospitalization for any reason had a significant decrease in transplant-free survival compared to the never hospitalized cohort (3-year survival 60% vs. 94%; p = 0.0001). Hospitalization for ≥7 days was associated with worse outcomes than those hospitalized for <7 days (median survival 1.59 years vs. 7.17 years, p = 0.0012). Based on multivariate competing risk analysis, factors associated with death, with lung transplantation as a competing risk, were age (HR = 1.05 [95% 1.01-1.09]; P = 0.0443), male gender (HR = 4.94 [95% CI: 1.58-15.41]; P = 0.006), and all cause hospitalization (HR = 11.97 [95% CI: 1.36-105.49]; P = 0.0253). CONCLUSION This study highlights the impact of hospitalization on subsequent outcomes in the CTD-ILD population with a significantly reduced transplant-free survival demonstrated, especially after cardiopulmonary hospitalization events.
Collapse
Affiliation(s)
- Ankush P Ratwani
- Department of Internal Medicine, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Kareem I Ahmad
- Advanced Lung Disease and Transplant Program. Inova Fairfax Hospital, Falls Church, VA, USA
| | - Scott D Barnett
- Advanced Lung Disease and Transplant Program. Inova Fairfax Hospital, Falls Church, VA, USA
| | - Steven D Nathan
- Advanced Lung Disease and Transplant Program. Inova Fairfax Hospital, Falls Church, VA, USA
| | - A Whitney Brown
- Advanced Lung Disease and Transplant Program. Inova Fairfax Hospital, Falls Church, VA, USA.
| |
Collapse
|
24
|
|
25
|
Mancuzo EV, Soares MR, Pereira CADC. Six-minute walk distance and survival time in patients with idiopathic pulmonary fibrosis in Brazil. ACTA ACUST UNITED AC 2019; 44:267-272. [PMID: 30328926 PMCID: PMC6326705 DOI: 10.1590/s1806-37562018000000049] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/08/2018] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To determine the cut-off point for the six-minute walk distance (6MWD) that indicates lower survival time in patients with idiopathic pulmonary fibrosis (IPF) in Brazil. METHODS This was retrospective study carried out in two referral centers for IPF. The 6MWT was performed twice, considering the highest value of the 6MWD. Various cut-off points were estimated, in absolute values and in percentage of predicted values, using ROC curves, the Kaplan-Meier method, and data from other studies. RESULTS The sample comprised 70 patients with IPF. The mean age was 71.9 ± 6.4 years, and 50 patients (71.4%) were male. The mean FVC was 76.6 ± 18.2% of predicted value. The mean SpO2 at rest before and after 6MWT were 93.8 ± 2.5% and 85.3 ± 6.5%, respectively. The median survival time was 44 months (95% CI: 37-51 months). The mean 6MWD was 381 ± 115 m (79.2 ± 24.0% of predicted). After the analyses, the best cut-off points for estimating survival were 6MWD < 330 m and < 70% of predicted. The median survival time of patients with a 6MWD < 330 m was 24 months (95% CI: 3-45 months), whereas that of those with a 6MWD ≥ 330 m was 59 months (95% CI: 41-77 months; p = 0.009). Similarly, the median survival times of those with a 6MWD < 70% and ≥ 70% of predicted, respectively, were 24 months (95% CI: 13-35 months) and 59 months (95% CI: 38-80 months; p = 0.013). Cox multivariate regression models including age, sex, smoking status, SpO2 at the end of the 6MWT, and FVC% showed that 6MWD remained significantly associated with survival (p = 0.003). CONCLUSIONS Values of 6MWD < 330 m and < 70% of predicted value were associated with lower survival time in IPF patients in Brazil.
Collapse
Affiliation(s)
- Eliane Viana Mancuzo
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte (MG) Brasil.,Ambulatório de Doenças Pulmonares Intersticiais, Hospital das Clínicas, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte (MG) Brasil
| | | | - Carlos Alberto de Castro Pereira
- Programa de Pós-Graduação, Assistência e Pesquisa, Ambulatório de Doenças Pulmonares Intersticiais, Faculdade de Medicina, Universidade Federal de São Paulo, São Paulo (SP) Brasil
| |
Collapse
|
26
|
Nawroth JC, Barrile R, Conegliano D, van Riet S, Hiemstra PS, Villenave R. Stem cell-based Lung-on-Chips: The best of both worlds? Adv Drug Deliv Rev 2019; 140:12-32. [PMID: 30009883 PMCID: PMC7172977 DOI: 10.1016/j.addr.2018.07.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/06/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023]
Abstract
Pathologies of the respiratory system such as lung infections, chronic inflammatory lung diseases, and lung cancer are among the leading causes of morbidity and mortality, killing one in six people worldwide. Development of more effective treatments is hindered by the lack of preclinical models of the human lung that can capture the disease complexity, highly heterogeneous disease phenotypes, and pharmacokinetics and pharmacodynamics observed in patients. The merger of two novel technologies, Organs-on-Chips and human stem cell engineering, has the potential to deliver such urgently needed models. Organs-on-Chips, which are microengineered bioinspired tissue systems, recapitulate the mechanochemical environment and physiological functions of human organs while concurrent advances in generating and differentiating human stem cells promise a renewable supply of patient-specific cells for personalized and precision medicine. Here, we discuss the challenges of modeling human lung pathophysiology in vitro, evaluate past and current models including Organs-on-Chips, review the current status of lung tissue modeling using human pluripotent stem cells, explore in depth how stem-cell based Lung-on-Chips may advance disease modeling and drug testing, and summarize practical consideration for the design of Lung-on-Chips for academic and industry applications.
Collapse
Affiliation(s)
| | | | | | - Sander van Riet
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, the Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, the Netherlands
| | | |
Collapse
|
27
|
Abstract
This is a time of substantial progress in the evaluation and care of patients with idiopathic pulmonary fibrosis (IPF). In addition to the approval and widespread availability of the first IPF-specific therapies, there have been improvements in imaging interpretation and lung biopsy methods to enable more expeditious and more accurate diagnosis. Recent advances in identifying genetic factors that underlie susceptibility to IPF and affect prognosis have raised the possibility of personalized therapeutic approaches in the future. Further, evolving work is elucidating novel mechanisms influencing epithelial, mesenchymal, and inflammatory cell responses during the injury-repair process, thus advancing understanding of disease pathogenesis. As analytic approaches mature, the field is now poised to harness the power of rapidly advancing "omics" technologies to further accelerate progress.
Collapse
Affiliation(s)
- Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA; ,
- Department of Veterans Affairs Medical Center, Nashville, Tennessee 37212, USA
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA; ,
- Department of Veterans Affairs Medical Center, Nashville, Tennessee 37212, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37212, USA
| |
Collapse
|
28
|
|
29
|
Mimics of Idiopathic Pulmonary Fibrosis. Respir Med 2019. [DOI: 10.1007/978-3-319-99975-3_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
30
|
Future Directions for IPF Research. Respir Med 2019. [DOI: 10.1007/978-3-319-99975-3_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
31
|
|
32
|
Goldin JG. Predicting Outcome in Idiopathic Pulmonary Fibrosis Using Automated Computed Tomography Analysis. Am J Respir Crit Care Med 2018; 198:701-702. [DOI: 10.1164/rccm.201804-0657ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Jonathan G. Goldin
- David Geffen School of Medicine at UCLAUniversity of California, Los AngelesLos Angeles, California
| |
Collapse
|
33
|
Humphries SM, Swigris JJ, Brown KK, Strand M, Gong Q, Sundy JS, Raghu G, Schwarz MI, Flaherty K, Sood R, O'Riordan TG, Lynch DA. Quantitative high-resolution computed tomography fibrosis score: performance characteristics in idiopathic pulmonary fibrosis. Eur Respir J 2018; 52:13993003.01384-2018. [DOI: 10.1183/13993003.01384-2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 01/05/2023]
Abstract
We evaluated performance characteristics and estimated the minimal clinically important difference (MCID) of data-driven texture analysis (DTA), a high-resolution computed tomography (HRCT)-derived measurement of lung fibrosis, in subjects with idiopathic pulmonary fibrosis (IPF).The study population included 141 subjects with IPF from two interventional clinical trials who had both baseline and nominal 54- or 60-week follow-up HRCT. DTA scores were computed and compared with forced vital capacity (FVC), diffusing capacity of the lung for carbon monoxide, distance covered during a 6-min walk test and St George's Respiratory Questionnaire scores to assess the method's reliability, validity and responsiveness. Anchor- and distribution-based methods were used to estimate its MCID.DTA had acceptable reliability in subjects appearing stable according to anchor variables at follow-up. Correlations between the DTA score and other clinical measurements at baseline were moderate to weak and in the hypothesised directions. Acceptable responsiveness was demonstrated by moderate to weak correlations (in the directions hypothesised) between changes in the DTA score and changes in other parameters. Using FVC as an anchor, MCID was estimated to be 3.4%.Quantification of lung fibrosis extent on HRCT using DTA is reliable, valid and responsive, and an increase of ∼3.4% represents a clinically important change.
Collapse
|
34
|
Conforti F, Davies ER, Calderwood CJ, Thatcher TH, Jones MG, Smart DE, Mahajan S, Alzetani A, Havelock T, Maher TM, Molyneaux PL, Thorley AJ, Tetley TD, Warner JA, Packham G, Ganesan A, Skipp PJ, Marshall BJ, Richeldi L, Sime PJ, O'Reilly KMA, Davies DE. The histone deacetylase inhibitor, romidepsin, as a potential treatment for pulmonary fibrosis. Oncotarget 2018; 8:48737-48754. [PMID: 28467787 PMCID: PMC5564721 DOI: 10.18632/oncotarget.17114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 11/25/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease that usually affects elderly people. It has a poor prognosis and there are limited therapies. Since epigenetic alterations are associated with IPF, histone deacetylase (HDAC) inhibitors offer a novel therapeutic strategy to address the unmet medical need. This study investigated the potential of romidepsin, an FDA-approved HDAC inhibitor, as an anti-fibrotic treatment and evaluated biomarkers of target engagement that may have utility in future clinical trials. The anti-fibrotic effects of romidepsin were evaluated both in vitro and in vivo together with any harmful effect on alveolar type II cells (ATII). Bronchoalveolar lavage fluid (BALF) from IPF or control donors was analyzed for the presence of lysyl oxidase (LOX). In parallel with an increase in histone acetylation, romidepsin potently inhibited fibroblast proliferation, myofibroblast differentiation and LOX expression. ATII cell numbers and their lamellar bodies were unaffected. In vivo, romidepsin inhibited bleomycin-induced pulmonary fibrosis in association with suppression of LOX expression. LOX was significantly elevated in BALF of IPF patients compared to controls. These data show the anti-fibrotic effects of romidepsin, supporting its potential use as novel treatment for IPF with LOX as a companion biomarker for evaluation of early on-target effects.
Collapse
Affiliation(s)
- Franco Conforti
- The Brooke Laboratory, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK.,NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
| | - Elizabeth R Davies
- The Brooke Laboratory, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK.,NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
| | - Claire J Calderwood
- The Brooke Laboratory, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - Thomas H Thatcher
- Department of Medicine/Pulmonary & Critical Care, University of Rochester, Rochester, NY, USA
| | - Mark G Jones
- The Brooke Laboratory, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK.,NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
| | - David E Smart
- The Brooke Laboratory, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK.,NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
| | - Sumeet Mahajan
- Institute for Life Sciences, University of Southampton, Highfield, UK
| | | | - Tom Havelock
- NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK.,University Hospital Southampton, Southampton, UK
| | - Toby M Maher
- NIHR Respiratory Biomedical Research Unit, Royal Brompton Hospital, London, UK.,National Heart & Lung Institute, Faculty of Medicine, Imperial College, London, UK
| | - Philip L Molyneaux
- NIHR Respiratory Biomedical Research Unit, Royal Brompton Hospital, London, UK.,National Heart & Lung Institute, Faculty of Medicine, Imperial College, London, UK
| | - Andrew J Thorley
- National Heart & Lung Institute, Faculty of Medicine, Imperial College, London, UK
| | - Teresa D Tetley
- National Heart & Lung Institute, Faculty of Medicine, Imperial College, London, UK
| | - Jane A Warner
- The Brooke Laboratory, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK.,NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK.,Institute for Life Sciences, University of Southampton, Highfield, UK
| | - Graham Packham
- Cancer Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - A Ganesan
- School of Pharmacy, University of East Anglia, Norwich, UK
| | - Paul J Skipp
- Institute for Life Sciences, University of Southampton, Highfield, UK
| | | | - Luca Richeldi
- The Brooke Laboratory, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK.,NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK.,University Hospital Southampton, Southampton, UK
| | - Patricia J Sime
- Department of Medicine/Pulmonary & Critical Care, University of Rochester, Rochester, NY, USA
| | - Katherine M A O'Reilly
- NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK.,Respiratory Medicine, Mater Misericordiae University Hospital, Dublin, Ireland.,School of Medicine and Medical Science, University College, Dublin, Ireland
| | - Donna E Davies
- The Brooke Laboratory, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UK.,NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK.,Institute for Life Sciences, University of Southampton, Highfield, UK
| |
Collapse
|
35
|
Torrisi SE, Pavone M, Vancheri A, Vancheri C. When to start and when to stop antifibrotic therapies. Eur Respir Rev 2017; 26:26/145/170053. [PMID: 28974541 PMCID: PMC9488637 DOI: 10.1183/16000617.0053-2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/12/2017] [Indexed: 11/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterised by progressive changes of the lung architecture causing cough and dyspnoea and ultimately leading to lung failure and death. Today, for the first time, two drugs that may reduce the inexorable progression of the disease are available, suggesting that treatment with specific drugs for IPF should be started as soon as diagnosis is made. This applies to any disease and particularly to IPF, which is marked by a 5-year survival comparable or even worse than many cancers. However, despite common sense and even worse, in spite of scientific data coming from clinical trials, post hoc analysis, long-term safety studies and real-life experiences, the question of when to start and when to stop treatment with antifibrotics is still debated. In IPF, particularly when the disease is diagnosed at an early stage, “wait and watch” behaviour is not rare to observe. This is largely due to the lack of awareness of both patients and clinicians regarding the progression of the disease and its prognosis. Another important issue is when treatment should be stopped. In general, there are two main reasons to stop a therapy: unbearable side-effects and/or lack of efficacy. According to current (although preliminary) evidence, antifibrotic drugs should not be discontinued except for safety issues. Today, for the first time, we can use two effective drugs able to reduce the inexorable progression of IPFhttp://ow.ly/MCS930eOp2b
Collapse
Affiliation(s)
- Sebastiano Emanuele Torrisi
- Regional Referral Centre for Rare Lung Diseases, A.O.U. Policlinico-Vittorio Emanuele, University of Catania, Catania, Italy
| | - Mauro Pavone
- Regional Referral Centre for Rare Lung Diseases, A.O.U. Policlinico-Vittorio Emanuele, University of Catania, Catania, Italy
| | - Ada Vancheri
- Regional Referral Centre for Rare Lung Diseases, A.O.U. Policlinico-Vittorio Emanuele, University of Catania, Catania, Italy
| | - Carlo Vancheri
- Regional Referral Centre for Rare Lung Diseases, A.O.U. Policlinico-Vittorio Emanuele, University of Catania, Catania, Italy
| |
Collapse
|
36
|
Ley B, Swigris J, Day BM, Stauffer JL, Raimundo K, Chou W, Collard HR. Pirfenidone Reduces Respiratory-related Hospitalizations in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2017; 196:756-761. [PMID: 28471697 DOI: 10.1164/rccm.201701-0091oc] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
RATIONALE Respiratory-related hospitalizations of patients with idiopathic pulmonary fibrosis (IPF) are more frequent than those for acute IPF exacerbations and are associated with poor outcomes. OBJECTIVES To compare the risk of nonelective hospitalization by type (all-cause, respiratory related, and non-respiratory related) and death after hospitalization with use of pirfenidone versus placebo over 52 weeks using data derived from three phase III IPF clinical trials. METHODS Individual patient data was pooled from three phase III randomized, placebo-controlled studies of pirfenidone for IPF (the two CAPACITY [Clinical Studies Assessing Pirfenidone in IPF: Research of Efficacy and Safety Outcomes] trials and the ASCEND [Assessment of Pirfenidone to Confirm Efficacy and Safety in Idiopathic Pulmonary Fibrosis] trial), including all patients randomized to pirfenidone 2,403 mg/d (n = 623) or placebo (n = 624). The risk of hospitalization over 52 weeks was compared using standard time-to-event methods. Among those hospitalized, the risk of death after hospitalization was compared with adjustment for treatment group propensity. MEASUREMENTS AND MAIN RESULTS A total of 1,247 patients (692 from the CAPACITY trials and 555 from the ASCEND trial) were included in the pooled analysis. Pirfenidone was associated with lower risk of respiratory-related hospitalization than placebo (7% vs. 12%; hazard ratio [HR], 0.52; 95% confidence interval [CI], 0.36-0.77; P = 0.001), but all-cause (HR, 0.91; 95% CI, 0.70-1.19; P = 0.528) or non-respiratory-related hospitalization (HR, 1.32; 95% CI, 0.92-1.88; P = 0.145) was not. Among those hospitalized for any reason, treatment with pirfenidone was associated with lower risk of death after hospitalization up to 52 weeks after randomization, but this association was no longer significant with longer follow-up. CONCLUSIONS In a pooled analysis of three phase III IPF clinical trials, patients receiving pirfenidone had a lower risk of nonelective respiratory-related hospitalization over the course of 1 year. The effect of pirfenidone on death after hospitalization is uncertain.
Collapse
Affiliation(s)
- Brett Ley
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Jeffrey Swigris
- 2 Interstitial Lung Disease Program, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado; and
| | - Bann-Mo Day
- 3 Genentech, Inc., South San Francisco, California
| | | | | | - Willis Chou
- 3 Genentech, Inc., South San Francisco, California
| | - Harold R Collard
- 1 Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| |
Collapse
|
37
|
Abstract
INTRODUCTION While many pharmacologic therapies for the treatment of idiopathic pulmonary fibrosis (IPF) have been evaluated via randomized, placebo-controlled clinical trials (RCTs) conducted over the past two decades, most therapies have been shown to be ineffective or even potentially harmful. However, a number of recently completed RCTs have shown significant efficacy for pirfenidone and nintedanib for the treatment of IPF. Areas covered: This manuscript reviews recent advances in the management of IPF and other forms of fibrosing interstitial lung disease (ILD) with an emphasis on IPF. The material upon which this discussion is based was obtained from various published texts and manuscripts identified via literature searching (e.g. PubMed). Expert commentary: Anti-fibrotic drugs are now available for clinical use and perceived as standard-of-care therapies that have the potential to blunt disease progression for many patients with IPF. However, these agents do not necessarily stop disease progression or have a significant impact on mortality, and more effective pharmacologic therapies are needed for patients with IPF. Additionally, whether anti-fibrotic agents can be effective therapies for other forms of pulmonary fibrosis, which often have radiologic and histopathologic manifestations that mimic IPF, is being evaluated in a number of RCTs.
Collapse
Affiliation(s)
- Keith C Meyer
- a Department of Medicine , University of Wisconsin School of Medicine and Public Health - Medicine , K4/910 Clinical Science Center 600 Highland Avenue Madison, Madison , WI , 53792-9988 , USA
| |
Collapse
|
38
|
Abstract
INTRODUCTION Many forms of interstitial lung disease (ILD) can progress to extensive fibrosis and respiratory failure. Idiopathic pulmonary fibrosis (IPF), which generally has a poor prognosis, has been thoroughly studied over the past two decades, and many important discoveries have been made that pertain to genetic predisposition, epidemiology, disease pathogenesis, diagnosis, and management. Additionally, non-IPF forms of ILD can have radiologic and histopathologic manifestations that mimic IPF, and making an accurate diagnosis is key to providing personalized medicine to patients with pulmonary fibrosis. Areas covered: This manuscript discusses current knowledge pertaining to the genetics, epidemiology, pathogenesis, and diagnosis of pulmonary fibrosis with an emphasis on IPF. The material upon which this discussion is based was obtained from various published texts and manuscripts identified via literature searching (e.g. PubMed). Expert commentary: Many genetic variants have been identified that are associated with risk of developing pulmonary fibrosis, and an improved understanding of the influence of both genomic and epigenomic factors in the development of pulmonary fibrosis is rapidly evolving. Because many forms of fibrosing ILD can have similar radiologic and histopathologic patterns yet have different responses to therapeutic interventions, making an accurate diagnosis of specific forms of pulmonary fibrosis is increasingly important.
Collapse
Affiliation(s)
- Keith C Meyer
- a Department of Medicine , University of Wisconsin School of Medicine and Public Health - Medicine , Madison , WI , United States
| |
Collapse
|
39
|
Idiopathic Interstitial Pneumonia Associated With Autoantibodies: A Large Case Series Followed Over 1 Year. Chest 2017; 152:103-112. [PMID: 28300570 DOI: 10.1016/j.chest.2017.03.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 02/01/2017] [Accepted: 03/01/2017] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Some patients with autoimmune characteristics and idiopathic interstitial pneumonia, particularly usual interstitial pneumonia (UIP), do not fit neatly into the category of connective tissue disease-associated interstitial lung disease (CTD-ILD), idiopathic pulmonary fibrosis (IPF), or recently proposed yet to be validated criteria for interstitial pneumonia with autoimmune features (IPAF). Outcomes of these patients are unknown. METHODS This was a retrospective single-center study. Analyses of variance compared differences in mean change in FVC and diffusion capacity (Dlco) over 1 year among 124 well-defined patients (20 patients with positive autoantibodies with or without symptoms of connective tissue disease [AI-ILD], 15 patients with IPAF, 36 patients with CTD-ILD, and 53 patients with IPF with negative CTD serologies [Lone-IPF]). RESULTS Of the patients, 75% with AI-ILD, 33% with IPAF, and 33% with CTD-ILD had UIP. Initial FVC and Dlco were similarly moderately reduced across groups. Mean change in FVC over 12 months was as follows: -60 mL (IPAF), -110 mL (AI-ILD), -10 mL (CTD-ILD), and -90 mL (Lone-IPF) (P = .52). Mean change in Dlco was as follows: 2.39 mL/mm Hg/min (IPAF), -1.15 mL/mm Hg/min (AI-ILD), -0.27 mL/mm Hg/min (CTD-ILD), and -1.05 mL/mm Hg/min (Lone-IPF) (P < .001). By pattern of disease, the mean change in FVC was as follows: -140 mL (UIP), 10 mL (nonspecific interstitial pneumonia), and 12 mL (unclassifiable/other) (P = .001). CONCLUSIONS No clinically significant differences in pulmonary function to distinguish between patients with AI-ILD, IPAF, CTD-ILD, and Lone-IPF were observed after 1 year. Longer periods of follow-up are needed to understand the outcomes of these patients. It is not yet clear whether AI-ILD is a distinct phenotype or a variant of the newly proposed entity IPAF.
Collapse
|
40
|
Bonini M, Fiorenzano G. Exertional dyspnoea in interstitial lung diseases: the clinical utility of cardiopulmonary exercise testing. Eur Respir Rev 2017; 26:26/143/160099. [DOI: 10.1183/16000617.0099-2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/01/2016] [Indexed: 01/08/2023] Open
Abstract
Interstitial lung diseases (ILDs) represent a heterogeneous group of pathologies characterised by alveolar and interstitial damage, pulmonary inflammation (usually associated with fibrosis), decreased lung function and impaired gas exchange, which can be attributed to either a known or an unknown aetiology. Dyspnoea is one of the most common and disabling symptoms in patients with ILD, significantly impacting quality of life. The mechanisms causing dyspnoea are complex and not yet fully understood. However, it is recognised that dyspnoea occurs when there is an imbalance between the central respiratory efferent drive and the response of the respiratory musculature. The respiratory derangement observed in ILD patients at rest is even more evident during exercise. Pathophysiological mechanisms responsible for exertional dyspnoea and reduced exercise tolerance include altered respiratory mechanics, impaired gas exchange, cardiovascular abnormalities and peripheral muscle dysfunction.This review describes the respiratory physiology of ILD, both at rest and during exercise, and aims to provide comprehensive and updated evidence on the clinical utility of the cardiopulmonary exercise test in the assessment and management of these pathological entities. In addition, the role of exercise training and pulmonary rehabilitation programmes in the ILD population is addressed.
Collapse
|
41
|
Meyer KC. Great expectations for simtuzumab in IPF fall short. THE LANCET RESPIRATORY MEDICINE 2016; 5:2-3. [PMID: 27939077 DOI: 10.1016/s2213-2600(16)30420-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 11/03/2016] [Indexed: 11/15/2022]
Affiliation(s)
- Keith C Meyer
- University of Wisconsin Lung Transplant and Advanced Lung Disease Program, Section of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, K4/910 Clinical Sciences Center, Madison, WI 53792, USA.
| |
Collapse
|
42
|
|
43
|
Tang Y, He R, An J, Deng P, Huang L, Yang W. The effect of H19-miR-29b interaction on bleomycin-induced mouse model of idiopathic pulmonary fibrosis. Biochem Biophys Res Commun 2016; 479:417-423. [DOI: 10.1016/j.bbrc.2016.09.028] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 09/05/2016] [Indexed: 11/16/2022]
|
44
|
POINT: Should All Patients With Idiopathic Pulmonary Fibrosis, Even Those With More Than Moderate Impairment, Be Treated With Nintedanib or Pirfenidone? Yes. Chest 2016; 150:273-5. [DOI: 10.1016/j.chest.2016.04.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 04/19/2016] [Indexed: 11/18/2022] Open
|
45
|
Albera C, Costabel U, Fagan EA, Glassberg MK, Gorina E, Lancaster L, Lederer DJ, Nathan SD, Spirig D, Swigris JJ. Efficacy of pirfenidone in patients with idiopathic pulmonary fibrosis with more preserved lung function. Eur Respir J 2016; 48:843-51. [DOI: 10.1183/13993003.01966-2015] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/25/2016] [Indexed: 11/05/2022]
Abstract
This post hoc analysis examined the differences in idiopathic pulmonary fibrosis disease progression and the effects of pirfenidone in patients stratified by more preserved versus less preserved baseline lung function status using forced vital capacity (FVC) or GAP (gender, age and physiology) index stage.Efficacy outcomes, i.e. FVC, 6-min walking distance (6MWD) and dyspnoea (University of California San Diego Shortness of Breath Questionnaire (UCSD SOBQ)), were analysed at 12 months in patients randomised to pirfenidone 2403 mg·day−1 or placebo in the pooled phase 3 CAPACITY/ASCEND population (n=1247), with subgroups stratified by baseline FVC ≥80% versus <80% or GAP stage I versus II–III. Treatment-by-subgroup interaction was tested based on a rank ANCOVA model; factors in the model included study, region, treatment, subgroup and treatment-by-subgroup interaction term.Patients with both more preserved (FVC ≥80% or GAP stage I) and less preserved (FVC <80% or GAP stage II–III) lung function at baseline demonstrated clinically significant disease progression at 12 months in terms of categorical decline in FVC, 6MWD and UCSD SOBQ. The magnitude of pirfenidone treatment effect was comparable between subgroups, regardless of whether lung function was classified using FVC or GAP index stage.These findings support the initiation of treatment with pirfenidone, irrespective of stage of baseline lung function in this patient population.
Collapse
|
46
|
Affiliation(s)
- Susan K Mathai
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, CO, USA
| | - David A Schwartz
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Aurora, CO, USA Dept of Medicine, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
47
|
Sgalla G, Cocconcelli E, Tonelli R, Richeldi L. Novel drug targets for idiopathic pulmonary fibrosis. Expert Rev Respir Med 2016; 10:393-405. [PMID: 26854382 DOI: 10.1586/17476348.2016.1152186] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a progressive, fatal lung disorder of unknown cause with a highly variable and unpredictable clinical course. The advances made in deciphering IPF pathobiology over the last decades have led to the approval of two anti-fibrotic molecules, pirfenidone and nintedanib, that showed to be effective in significantly reducing the rate of progression of the disease. Such pharmacological breakthroughs represent a dramatic change in the management of these patients and are reflected in updated international guidelines. However, the need to find a cure for this devastating disease remains unmet and the development of novel therapeutic agents remains hurdled by several factors. Here, we review the latest insights into therapeutic approaches for IPF and the available evidence for the most promising novel compounds currently under development, and discuss the challenges and evolution of IPF clinical research over the next few years.
Collapse
Affiliation(s)
- Giacomo Sgalla
- a Southampton NIHR Respiratory Biomedical Research Unit , University Hospital Southampton , Southampton , UK
| | - Elisabetta Cocconcelli
- b Department of Cardiologic, Thoracic and Vascular Sciences, Section of Respiratory Diseases , University of Padova , Padova , Italy
| | - Roberto Tonelli
- c Department of Respiratory Diseases , University Hospital of Modena , Modena , Italy
| | - Luca Richeldi
- a Southampton NIHR Respiratory Biomedical Research Unit , University Hospital Southampton , Southampton , UK.,d Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine , University Hospital Southampton , Southampton , UK
| |
Collapse
|
48
|
Mathai SK, Yang IV, Schwarz MI, Schwartz DA. Incorporating genetics into the identification and treatment of Idiopathic Pulmonary Fibrosis. BMC Med 2015; 13:191. [PMID: 26400796 PMCID: PMC4581155 DOI: 10.1186/s12916-015-0434-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 07/24/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis, the most common form of idiopathic interstitial pneumonia, is characterized by progressive, irreversible scarring of the lung parenchyma. Idiopathic pulmonary fibrosis has a poor prognosis, and there are no medical therapies available that have been shown to improve survival. It is usually sporadic, but there is evidence of familial clustering of pulmonary fibrosis, suggesting a genetic basis for this disease. More recently, studies have confirmed that specific genetic variants are associated with both familial and sporadic forms of pulmonary fibrosis. DISCUSSION Although there are common and rare genetic variants that have been associated with the risk of developing pulmonary fibrosis, the genotyping of patients is not a generally accepted strategy. Better understanding of the interplay between genetic risk and environmental exposure is likely needed to inform both treatment and disease prevention. Several identified disease-associated genetic variants have implications for disease progression and survival, but systematic studies of known genetic variants and their influence on therapeutic efficacy are lacking. Future investigations should focus on understanding phenotypic differences between patients carrying different risk alleles, and clinical studies should be designed to control for the influence of different genetic risk variants on patient outcomes. Inherited genetic factors play a significant role in the risk of developing pulmonary fibrosis. Future studies will be needed to characterize patient phenotypes and to understand how these genetic factors will influence clinical decision-making for both diagnosis and treatment of idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Susan K Mathai
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, 12631 East 17th Avenue, Aurora, CO, 80045, USA.
| | - Ivana V Yang
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, 12631 East 17th Avenue, Aurora, CO, 80045, USA.
| | - Marvin I Schwarz
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, 12631 East 17th Avenue, Aurora, CO, 80045, USA.
| | - David A Schwartz
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, 12631 East 17th Avenue, Aurora, CO, 80045, USA.
| |
Collapse
|
49
|
Meyer KC, Danoff SK, Lancaster LH, Nathan SD. Management of Idiopathic Pulmonary Fibrosis in the Elderly Patient: Addressing Key Questions. Chest 2015; 148:242-252. [PMID: 26149553 DOI: 10.1378/chest.14-2475] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is strongly associated with advanced age. Making an accurate diagnosis of IPF is critical, as it remains only one of many potential diagnoses for an elderly patient with newly recognized interstitial lung disease. Optimal management of IPF, especially in older-aged patients, hinges on such factors as balancing the application of standard-of-care measures with the patient's overall health status (robustness vs frailty) and considering the patient's wishes, desires, and expectations. IPF is known to be associated with certain comorbidities that tend to be more prevalent in the elderly population. Until recently, options for the pharmacologic management of IPF were limited and included therapies such as immunosuppressive agents, which may pose substantial risk to the elderly patient. However, the antifibrotic agents pirfenidone and nintedanib have now become commercially available in the United States for the treatment of IPF. The monitoring and treatment of patients with IPF, especially elderly patients with comorbid medical conditions, require consideration of adverse side effects, the avoidance of potential drug-drug interactions, treatment of comorbidities, and the timely implementation of supportive and palliative measures. Individualized counseling to guide decision-making and enhance quality of life is also integral to optimal management of the elderly patient with IPF.
Collapse
Affiliation(s)
- Keith C Meyer
- Department of Medicine (Dr Meyer), Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| | - Sonye K Danoff
- Division of Allergy, Pulmonary, and Critical Care Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lisa H Lancaster
- Division of Allergy, Pulmonary, and Critical Care Medicine, the Vanderbilt University Medical Center, Nashville, TN
| | - Steven D Nathan
- Advanced Lung Disease and Transplant Program, Department of Medicine, Inova Health Systems, Falls Church, VA
| |
Collapse
|
50
|
Ji X, Wu B, Fan J, Han R, Luo C, Wang T, Yang J, Han L, Zhu B, Wei D, Chen J, Ni C. The Anti-fibrotic Effects and Mechanisms of MicroRNA-486-5p in Pulmonary Fibrosis. Sci Rep 2015; 5:14131. [PMID: 26370615 PMCID: PMC4569899 DOI: 10.1038/srep14131] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 07/30/2015] [Indexed: 01/25/2023] Open
Abstract
To identify microRNAs (miRNAs, miRs) with potential roles in lung fibrogenesis, we performed genome-wide profiling of miRNA expression in lung tissues from a silica-induced mouse model of pulmonary fibrosis using microarrays. Seventeen miRNAs were selected for validation via qRT-PCR based on the fold changes between the silica and the control group. The dysregulation of five miRNAs, including miR-21, miR-455, miR-151-3p, miR-486-5p and miR-3107, were confirmed by qRT-PCRs in silica-induced mouse model of pulmonary fibrosis and were also confirmed in a bleomycin (BLM)-induced mouse lung fibrosis. Notably, miR-486-5p levels were decreased in the serum samples of patients with silicosis, as well as in the lung tissues of patients with silicosis and idiopathic pulmonary fibrosis (IPF). In addition, as determined by luciferase assays and Western blotting, SMAD2, a crucial mediator of pulmonary fibrosis, was identified to be one of target genes of miR-486-5p. To test the potential therapeutic significance of this miRNA, we overexpressed miR-486-5p in animal models. At day 28, miR-486-5p expression significantly decreased both the distribution and severity of lung lesions compared with the silica group (P < 0.01). In addition, miR-486-5p had a similar effect in the BLM group (P < 0.001). These results indicate that miR-486-5p may inhibit fibrosis.
Collapse
Affiliation(s)
- Xiaoming Ji
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Baiqun Wu
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jingjing Fan
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ruhui Han
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chen Luo
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ting Wang
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jingjin Yang
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lei Han
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, China
| | - Baoli Zhu
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, China
| | - Dong Wei
- Nanjing Medical University, Affiliated Wuxi People's Hospital, Lung Transplantation Center, Jiangsu Key Laboratory of Organ Transplantation, China
| | - Jingyu Chen
- Nanjing Medical University, Affiliated Wuxi People's Hospital, Lung Transplantation Center, Jiangsu Key Laboratory of Organ Transplantation, China
| | - Chunhui Ni
- Department of Occupational Medicine and Environmental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|