1
|
Shen Q, Zhou S, Song M, Ouyang X, Tan Y, Peng Y, Zhou Z, Peng H. Prevalence and prognostic value of malnutrition in patients with IPF using three scoring systems. Respir Med 2024; 233:107774. [PMID: 39168392 DOI: 10.1016/j.rmed.2024.107774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND To describe the nutritional status of IPF patients, report clinical associations and evaluate the prognostic value of them in IPF. METHODS 264 IPF patients diagnosed with IPF at the Second Xiangya Hospital of Central South University between January 2011 and January 2021 were recruited. Three different scoring systems, including nutritional risk index (NRI), controlling nutritional status (CONUT) score, and prognostic nutritional index (PNI) were used to describe the nutritional status of IPF patients. RESULT This study investigated the prevalence of malnutrition in 264 IPF patients, of which the percentage with malnutrition varied from 37.5 % with the NRI, to 47.4 % with the CONUT score, and to 6.4 % with the PNI. The moderate to severe malnutrition ranged from 10.2 % to 31.1 % across these indices, with PNI identifying only 4.9 % in this category. Worsening malnutrition status was associated with significantly higher incidence of all-cause mortality and IPF death regard of the malnutrition index as NRI (p < 0.05). When the normal nutrition of NRI was used as a reference, patients in the moderate to severe risk remained at a higher risk of all-cause death (HR = 2.06(1.25-3.41)) and IPF death(HR = 2.36(1.35-4.15)). The adjusted multivariate analysis, identified age(HR = 1.13(1.08-1.20)), DLCO <60, % predicted (HR = 3.31(1,24-9.42)) and the use of anti-fibrotic drugs (HR = 0.25(0.10-0.60)) as independent predictors of mortality. CONCLUSIONS Malnutrition is common among patients with IPF and the baseline as diagnosis of IPF is strongly related to increased mortality.
Collapse
Affiliation(s)
- Qinxue Shen
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; The Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Shiting Zhou
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; The Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Min Song
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; The Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Xiaoli Ouyang
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; The Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Yuexin Tan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; The Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Yating Peng
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; The Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; The Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China
| | - Hong Peng
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China; The Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, 410011, China; Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
2
|
Fu H, Wang Z, Hu Z, Zhao T, Xin H, Wu F, Hou J, Yang Y, Zhang Y, Jiang W, Wang F, Deng N, Chen J. Pilot study of home-based monitoring for early prediction of acute exacerbations in patients with fibrosing interstitial lung diseases. Sci Rep 2024; 14:21101. [PMID: 39256540 PMCID: PMC11387483 DOI: 10.1038/s41598-024-71942-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
This study aimed to assess the potential of home monitoring using a monitoring application for the early prediction of acute exacerbations (AEs) in patients with fibrosing interstitial lung diseases (F-ILDs) by tracking symptoms, peripheral blood oxygen saturation (SpO2), and heart rate (HR). Data on symptoms, SpO2, and HR before and after a 1-min sit-to-stand test (1STST) were collected using an online home monitoring application. Symptoms were recorded at least 3 times a week, including cough intensity and frequency (Cough Assessment Test scale (COAT) score), breathlessness grade (modified Medical Research Council (mMRC) score), and SpO2 and HR before and after 1STST. Eighty-five patients with stable F-ILDs were enrolled. We observed a significant increase in COAT and mMRC scores, alongside a significant decrease in SpO2 before and after 1STST, 2 weeks before the first recorded AE. Furthermore, a combination of variables-an increase in COAT (≥ 4) and mMRC(≥ 1) scores, a decrease in SpO2 at rest (≥ 5%), and a decrease in SpO2 after 1STST (≥ 4%)- proved the most effective in predicting AE onset in patients with F-ILDs at 2 weeks before the first recorded AE. Home telemonitoring of symptoms, SpO2 holds potential value for early AE detection in patients with F-ILDs.
Collapse
Affiliation(s)
- Hongyan Fu
- Department of Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi, 710049, People's Republic of China
| | - Zhaojun Wang
- Department of Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Zhengyu Hu
- Department of Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
- School of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Tingting Zhao
- Department of Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Hongxia Xin
- Department of Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Fan Wu
- Guangzhou Institute of Respiratory Health and State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease and National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, People's Republic of China
| | - Jia Hou
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Yanjuan Yang
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Yanan Zhang
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Wangshu Jiang
- Ministry of Education Key Laboratory of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Faxuan Wang
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Ning Deng
- Ministry of Education Key Laboratory of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Juan Chen
- Department of Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China.
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China.
| |
Collapse
|
3
|
Adegunsoye A, Kropski JA, Behr J, Blackwell TS, Corte TJ, Cottin V, Glanville AR, Glassberg MK, Griese M, Hunninghake GM, Johannson KA, Keane MP, Kim JS, Kolb M, Maher TM, Oldham JM, Podolanczuk AJ, Rosas IO, Martinez FJ, Noth I, Schwartz DA. Genetics and Genomics of Pulmonary Fibrosis: Charting the Molecular Landscape and Shaping Precision Medicine. Am J Respir Crit Care Med 2024; 210:401-423. [PMID: 38573068 PMCID: PMC11351799 DOI: 10.1164/rccm.202401-0238so] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/04/2024] [Indexed: 04/05/2024] Open
Abstract
Recent genetic and genomic advancements have elucidated the complex etiology of idiopathic pulmonary fibrosis (IPF) and other progressive fibrotic interstitial lung diseases (ILDs), emphasizing the contribution of heritable factors. This state-of-the-art review synthesizes evidence on significant genetic contributors to pulmonary fibrosis (PF), including rare genetic variants and common SNPs. The MUC5B promoter variant is unusual, a common SNP that markedly elevates the risk of early and established PF. We address the utility of genetic variation in enhancing understanding of disease pathogenesis and clinical phenotypes, improving disease definitions, and informing prognosis and treatment response. Critical research gaps are highlighted, particularly the underrepresentation of non-European ancestries in PF genetic studies and the exploration of PF phenotypes beyond usual interstitial pneumonia/IPF. We discuss the role of telomere length, often critically short in PF, and its link to progression and mortality, underscoring the genetic complexity involving telomere biology genes (TERT, TERC) and others like SFTPC and MUC5B. In addition, we address the potential of gene-by-environment interactions to modulate disease manifestation, advocating for precision medicine in PF. Insights from gene expression profiling studies and multiomic analyses highlight the promise for understanding disease pathogenesis and offer new approaches to clinical care, therapeutic drug development, and biomarker discovery. Finally, we discuss the ethical, legal, and social implications of genomic research and therapies in PF, stressing the need for sound practices and informed clinical genetic discussions. Looking forward, we advocate for comprehensive genetic testing panels and polygenic risk scores to improve the management of PF and related ILDs across diverse populations.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Pulmonary/Critical Care, and
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Juergen Behr
- Department of Medicine V, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, member of the German Center for Lung Research (DZL), Munich, Germany
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Tamera J. Corte
- Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases (OrphaLung), Louis Pradel Hospital, Hospices Civils de Lyon, ERN-LUNG (European Reference Network on Rare Respiratory Diseases), Lyon, France
- Claude Bernard University Lyon, Lyon, France
| | - Allan R. Glanville
- Lung Transplant Unit, St. Vincent’s Hospital Sydney, Sydney, New South Wales, Australia
| | - Marilyn K. Glassberg
- Department of Medicine, Loyola Chicago Stritch School of Medicine, Chicago, Illinois
| | - Matthias Griese
- Department of Pediatric Pneumology, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Munich, Germany
| | - Gary M. Hunninghake
- Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Michael P. Keane
- Department of Respiratory Medicine, St. Vincent’s University Hospital and School of Medicine, University College Dublin, Dublin, Ireland
| | - John S. Kim
- Department of Medicine, School of Medicine, and
| | - Martin Kolb
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Toby M. Maher
- Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York; and
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - David A. Schwartz
- Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
4
|
Oğuz MS, Bingöl Z, Pıhtılı A, Karaca Özer P, Sarıtaş Arslan M, Kılıçaslan Z, Bilge AK, Kıyan E, Okumuş G. Oxygen saturation recovery after 6-minute walk test in patients with idiopathic pulmonary fibrosis. BMC Pulm Med 2024; 24:373. [PMID: 39085811 PMCID: PMC11292883 DOI: 10.1186/s12890-024-03188-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/29/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The six-minute walk test (6MWT) is widely used to assess functional capacity and prognosis in patients with idiopathic pulmonary fibrosis (IPF). However, studies on oxygen saturation recovery after the 6MWT in patients with IPF are rare. In our study, we investigated the relationship between oxygen saturation recovery time and dyspnea, fatigue, quality of life, prognostic markers and pulmonary hypertension (PH). METHODS In this cross-sectional study, IPF patients diagnosed according to current guidelines and followed up in our Interstitial Lung Disease Outpatient Clinic between 2021 and 2022 were included. Demographics, data from spirometry, diffusion capacity measurement, arterial blood gas analysis, transthoracic echocardiography and the 6MWT were recorded. The oxygen saturation recovery time, distance saturation product (DSP), gender-age-physiology (GAP) index and composite physiological index (CPI) scores were calculated. Dyspnea severity was assessed by the modified Medical Research Council (mMRC) and Dyspnoea-12 (D-12) scales, fatigue severity by the Multidimensional Fatigue Inventory (MFI-20) and quality of life by the St George's Respiratory Questionnaire (SGRQ). RESULTS Fifty IPF patients (34 men, 16 women, age: 66.8 ± 7.3 years) were included in the study. The mean FVC was 77.8 ± 19.3%, the DLCO was 52.9 ± 17.1%, the 6-minute walk distance (6MWD) was 385.7 ± 90.6 m, the GAP index was 3.5 ± 1.5, and the CPI was 43.7 ± 14.1. Oxygen saturation after the 6MWT reached pretest values at an average of 135.6 ± 73.5 s. The oxygen saturation recovery time was longer in patients with higher GAP index scores (Rs = 0.870, p < 0.001), CPI scores (Rs = 0.906, p < 0.001), desaturation (Rs = 0.801, p < 0.001), FVC%/DLCO% (Rs = 0.432, p = 0.002), sPAP (Rs = 0.492, p = 0.001), TRV (Rs = 0.504, p = 0.001), mMRC (Rs = 0.913, p < 0.001), MFI-20 (Rs = 0.944, p < 0.001), D-12 scale (Rs = 0.915, p < 0.001) and SGRQ scores (Rs = 0.927, p < 0.001); lower FVC (%) (Rs=-0.627, p < 0.001), DLCO (%) (Rs=-0.892, p < 0.001), PaO2 (Rs=-0.779, p < 0.001), DSP (Rs=-0.835, p < 0.001), and 6MWD (Rs=-0.763, p < 0.001). A total of twenty patients (40%) exhibited an increased risk of PH. According to our multiple regression analysis, oxygen saturation recovery time was independently associated with the GAP index (p = 0.036), the lowest oxygen saturation occurring during the 6MWT (p = 0.011) and the SGRQ score (p < 0.001). CONCLUSIONS Our results showed that oxygen saturation recovery time is associated with dyspnea, fatigue, quality of life, increased risk of PH and prognostic markers in IPF. Therefore, we recommend continuous measurement of oxygen saturation after 6MWT until pretest values are reached.
Collapse
Affiliation(s)
- Merve Sinem Oğuz
- Department of Pulmonary Diseases, Istanbul Medical Faculty, Istanbul University, Capa-Fatih, Istanbul, Turkey
| | - Züleyha Bingöl
- Department of Pulmonary Diseases, Istanbul Medical Faculty, Istanbul University, Capa-Fatih, Istanbul, Turkey
| | - Aylin Pıhtılı
- Department of Pulmonary Diseases, Istanbul Medical Faculty, Istanbul University, Capa-Fatih, Istanbul, Turkey
| | - Pelin Karaca Özer
- Department of Cardiology, Istanbul Medical Faculty, Istanbul University, Capa-Fatih, Istanbul, Turkey
| | - Melike Sarıtaş Arslan
- Department of Pulmonary Diseases, Istanbul Medical Faculty, Istanbul University, Capa-Fatih, Istanbul, Turkey
| | - Zeki Kılıçaslan
- Department of Pulmonary Diseases, Istanbul Medical Faculty, Istanbul University, Capa-Fatih, Istanbul, Turkey
| | - Ahmet Kaya Bilge
- Department of Cardiology, Istanbul Medical Faculty, Istanbul University, Capa-Fatih, Istanbul, Turkey
| | - Esen Kıyan
- Department of Pulmonary Diseases, Istanbul Medical Faculty, Istanbul University, Capa-Fatih, Istanbul, Turkey
| | - Gülfer Okumuş
- Department of Pulmonary Diseases, Istanbul Medical Faculty, Istanbul University, Capa-Fatih, Istanbul, Turkey.
| |
Collapse
|
5
|
Pugashetti JV, Kim JS, Combs MP, Ma SF, Adegunsoye A, Linderholm AL, Strek ME, Chen CH, Dilling DF, Whelan TPM, Flaherty KR, Martinez FJ, Noth I, Oldham JM. A multidimensional classifier to support lung transplant referral in patients with pulmonary fibrosis. J Heart Lung Transplant 2024; 43:1174-1182. [PMID: 38556070 PMCID: PMC11451110 DOI: 10.1016/j.healun.2024.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/13/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Lung transplantation remains the sole curative option for patients with idiopathic pulmonary fibrosis (IPF), but donor organs remain scarce, and many eligible patients die before transplant. Tools to optimize the timing of transplant referrals are urgently needed. METHODS Least absolute shrinkage and selection operator was applied to clinical and proteomic data generated as part of a prospective cohort study of interstitial lung disease (ILD) to derive clinical, proteomic, and multidimensional logit models of near-term death or lung transplant within 18 months of blood draw. Model-fitted values were dichotomized at the point of maximal sensitivity and specificity, and decision curve analysis was used to select the best-performing classifier. We then applied this classifier to independent IPF and non-IPF ILD cohorts to determine test performance characteristics. Cohorts were restricted to patients aged ≤72 years with body mass index 18 to 32 to increase the likelihood of transplant eligibility. RESULTS IPF derivation, IPF validation, and non-IPF ILD validation cohorts consisted of 314, 105, and 295 patients, respectively. A multidimensional model comprising 2 clinical variables and 20 proteins outperformed stand-alone clinical and proteomic models. Following dichotomization, the multidimensional classifier predicted near-term outcome with 70% sensitivity and 92% specificity in the IPF validation cohort and 70% sensitivity and 80% specificity in the non-IPF ILD validation cohort. CONCLUSIONS A multidimensional classifier of near-term outcomes accurately discriminated this end-point with good test performance across independent IPF and non-IPF ILD cohorts. These findings support refinement and prospective validation of this classifier in transplant-eligible individuals.
Collapse
Affiliation(s)
- Janelle Vu Pugashetti
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - John S Kim
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Michael P Combs
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shwu-Fan Ma
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Angela L Linderholm
- Department of Internal Medicine, University of California Davis, Davis, California
| | - Mary E Strek
- Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Ching-Hsien Chen
- Department of Internal Medicine, University of California Davis, Davis, California
| | - Daniel F Dilling
- Division of Pulmonary and Critical Care Medicine, Loyola University Chicago, Stritch School of Medicine, Chicago, Illinois
| | - Timothy P M Whelan
- Division of Pulmonary and Critical Care, Medical University of South Carolina, Charleston, South Carolina; Pulmonary Fibrosis Foundation, Chicago, Illinois
| | - Kevin R Flaherty
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; Pulmonary Fibrosis Foundation, Chicago, Illinois
| | - Fernando J Martinez
- Division of Pulmonary and Critical Care, Weill Cornell Medical Center, New York, New York
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Justin M Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan; Department of Epidemiology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
6
|
Polat G, Özdemir Ö, Ermin S, Serçe Unat D, Demirci Üçsular F. Predictive factors of mortality in patients with idiopathic pulmonary fibrosis treated with antifibrotics: a novel prognostic scoring system. SARCOIDOSIS, VASCULITIS, AND DIFFUSE LUNG DISEASES : OFFICIAL JOURNAL OF WASOG 2024; 41:e2024021. [PMID: 38940720 PMCID: PMC11275550 DOI: 10.36141/svdld.v41i2.13779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 01/25/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND AND AIM Any test that provides sufficient prognostic information to guide treatment decisions in idiopathic pulmonary fibrosis (IPF) is not available. The aim of our study was to determine the predictive factors of mortality in patients with IPF treated with antifibrotics. METHODS Patients with diagnosis of IPF who were treated with antifibrotics between 2016 - 2021 were included in the study. Demographic, clinical and laboratory characteristics of the patients was derived from hospital records retrospectively. Kaplan Meier and multivariate cox regression analysis were achieved for detection of mortality predictors. RESULTS Study population was composed of 119 IPF patients with a male predominance of 80.7% (n=96). Mean age of the patients was 67.9 ± 7.07 years. On univariate analysis, sex was not a significant predictor of mortality (HR 1.79; 95% CI: 0.87 - 3.69, p =0.11). BMI ≤ 26,6 m2/kg, DLCO ≤ 3.11 ml/mmHg/min, age over 62 years, 6DWT ≤ 382 meters, NLR ≤ 2.67 and PDW ≤ 16.7% were found to be significant for predicting mortality. On multivariate cox regression analysis four parameters remained significant for prediction of mortality: RDW > 14%, NLR ≤ 2.67, BMI ≤ 26,6 m2/kg and DLCO ≤ 3.11 ml/mmHg/min (respectively, HR: 2.0. 95% CI: 1.02 - 3.91, p=0.44; HR: 2.68. 95% CI: 1.48 - 4.85, p=0.001, HR: 2.07. 95% CI: 1.14 - 3.76, p=0.02, HR: 3.46. 95% CI: 1.85 - 6.47, p<0.001). A scoring system with these parameters discriminated patients with worse prognosis with a sensitivity of 89.1 % and a specificity of 65.8 % when total point was over 2 (AUC0.83, p<0.001). Conclusions In this study, DLCO, BMI, RDW and NLR levels significantly predicted mortality in IPF patients. Along with GAP index, scoring system with these simple parameters may give information about the prognosis of an IPF patient treated with antifibrotics.
Collapse
Affiliation(s)
- Gülru Polat
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| | - Özer Özdemir
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| | - Sinem Ermin
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| | - Damla Serçe Unat
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| | - Fatma Demirci Üçsular
- Department of Pulmonology, University of Health Sciences, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, Izmir, Turkey
| |
Collapse
|
7
|
Chang SE, Jia G, Gao X, Schiffman C, Gupta S, Wolters P, Neighbors M. Pursuing Clinical Predictors and Biomarkers for Progression in ILD: Analysis of the Pulmonary Fibrosis Foundation (PFF) Registry. Lung 2024; 202:269-273. [PMID: 38753183 PMCID: PMC11142961 DOI: 10.1007/s00408-024-00694-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/30/2024] [Indexed: 06/02/2024]
Abstract
INTRODUCTION Pulmonary fibrosis is a characteristic of various interstitial lung diseases (ILDs) with differing etiologies. Clinical trials in progressive pulmonary fibrosis (PPF) enroll patients based on previously described clinical criteria for past progression, which include a clinical practice guideline for PPF classification and inclusion criteria from the INBUILD trial. In this study, we compared the ability of past FVC (forced vital capacity) progression and baseline biomarker levels to predict future progression in a cohort of patients from the PFF Patient Registry. METHODS Biomarkers previously associated with pathobiology and/or progression in pulmonary fibrosis were selected to reflect cellular senescence (telomere length), pulmonary epithelium (SP-D, RAGE), myeloid activation (CXCL13, YKL40, CCL18, OPN) and fibroblast activation (POSTN, COMP, PROC3). RESULTS PFF or INBUILD-like clinical criteria was used to separate patients into past progressor and non-past progressor groups, and neither clinical criterion appeared to enrich for patients with greater future lung function decline. All baseline biomarkers measured were differentially expressed in patient groups compared to healthy controls. Baseline levels of SP-D and POSTN showed the highest correlations with FVC slope over one year, though correlations were low. CONCLUSIONS Our findings provide further evidence that prior decline in lung function may not predict future disease progression for ILD patients, and elevate the need for molecular definitions of a progressive phenotype. Across ILD subtypes, certain shared pathobiologies may be present based on the molecular profile of certain biomarker groups observed. In particular, SP-D may be a common marker of pulmonary injury and future lung function decline across ILDs.
Collapse
Affiliation(s)
| | - Guiquan Jia
- Genentech, Inc, South San Francisco, CA, USA
| | - Xia Gao
- Genentech, Inc, South San Francisco, CA, USA
| | | | | | - Paul Wolters
- Department of Medicine, University of California, San Francisco, CA, USA
| | | |
Collapse
|
8
|
Zanini U, Luppi F, Kaur K, Anzani N, Franco G, Ferrara G, Kalluri M, Mura M. Use of 6-minute walk distance to predict lung transplant-free survival in fibrosing non-IPF interstitial lung diseases. Respirology 2024; 29:387-395. [PMID: 38320863 DOI: 10.1111/resp.14669] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/23/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND AND OBJECTIVE The identification of progression in patients with fibrosing non-idiopathic pulmonary fibrosis (IPF) interstitial lung diseases (ILDs) represents an ongoing clinical challenge. Lung function decline alone may have significant limitations in the detection of clinically significant progression. We hypothesized that longitudinal changes of 6-min walk distance (6MWD) from baseline, simultaneously considered with measures of lung function, may independently predict survival and identifying clinically significant progression of disease. METHODS Forced vital capacity (FVC), diffusing lung capacity (DLCO) and 6MWD were considered both at baseline and at 1 year in a discovery cohort (n = 105) and in a validation cohort (n = 138) from different centres. The primary endpoint was lung transplant (LTx)-free survival. RESULTS Average follow-up was 3 years in both cohorts. Combined incidence of deaths and LTx was 29% and 21%, respectively. No collinearity and no strong correlations were observed among FVC, DLCO and 6MWD longitudinal changes. While age, gender and BMI were not significant, 6MWD decline ≥24 m predicted LTx-free-survival significantly and independently from FVC and DLCO declines, with high sensitivity and specificity, in both the discovery and the validation cohorts. Although FVC and DLCO declines remained significant predictors of LTx-free survival, 6MWD decline was more accurate than the proposed ATS/ERS/JRS/ALAT functional criteria. Results were confirmed after stratifying patients by baseline FVC. CONCLUSION Longitudinal declines of 6MWD are associated with poor survival in fibrosing ILDs across a wide range of baseline severity, with high accuracy. 6MWD longitudinal decline is largely independent from lung function decline and may be integrated into the routine assessment of progression.
Collapse
Affiliation(s)
- Umberto Zanini
- Department of Medicine and Surgery, University of Milan-Bicocca, SC Pneumologia, Fondazione IRCCS "San Gerardo dei Tintori", Monza, Italy
- Division of Pulmonary Medicine, University of Alberta, and Alberta Health Services, Edmonton, Alberta, Canada
| | - Fabrizio Luppi
- Department of Medicine and Surgery, University of Milan-Bicocca, SC Pneumologia, Fondazione IRCCS "San Gerardo dei Tintori", Monza, Italy
| | - Karina Kaur
- Division of Pulmonary Medicine, University of Alberta, and Alberta Health Services, Edmonton, Alberta, Canada
| | - Niccolò Anzani
- Department of Medicine and Surgery, University of Milan-Bicocca, SC Pneumologia, Fondazione IRCCS "San Gerardo dei Tintori", Monza, Italy
| | - Giovanni Franco
- Department of Medicine and Surgery, University of Milan-Bicocca, SC Pneumologia, Fondazione IRCCS "San Gerardo dei Tintori", Monza, Italy
| | - Giovanni Ferrara
- Division of Pulmonary Medicine, University of Alberta, and Alberta Health Services, Edmonton, Alberta, Canada
| | - Meena Kalluri
- Division of Pulmonary Medicine, University of Alberta, and Alberta Health Services, Edmonton, Alberta, Canada
| | - Marco Mura
- Division of Respirology, Western University, London, Ontario, Canada
| |
Collapse
|
9
|
Althobiani MA, Russell AM, Jacob J, Ranjan Y, Folarin AA, Hurst JR, Porter JC. Interstitial lung disease: a review of classification, etiology, epidemiology, clinical diagnosis, pharmacological and non-pharmacological treatment. Front Med (Lausanne) 2024; 11:1296890. [PMID: 38698783 PMCID: PMC11063378 DOI: 10.3389/fmed.2024.1296890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/26/2024] [Indexed: 05/05/2024] Open
Abstract
Interstitial lung diseases (ILDs) refer to a heterogeneous and complex group of conditions characterized by inflammation, fibrosis, or both, in the interstitium of the lungs. This results in impaired gas exchange, leading to a worsening of respiratory symptoms and a decline in lung function. While the etiology of some ILDs is unclear, most cases can be traced back to factors such as genetic predispositions, environmental exposures (including allergens, toxins, and air pollution), underlying autoimmune diseases, or the use of certain medications. There has been an increase in research and evidence aimed at identifying etiology, understanding epidemiology, improving clinical diagnosis, and developing both pharmacological and non-pharmacological treatments. This review provides a comprehensive overview of the current state of knowledge in the field of interstitial lung diseases.
Collapse
Affiliation(s)
- Malik A. Althobiani
- Royal Free Campus, UCL Respiratory, University College London, London, United Kingdom
- Department of Respiratory Therapy, Faculty of Medical Rehabilitation Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anne-Marie Russell
- School of Health and Care Professions, University of Exeter, Exeter, United Kingdom
- School of Medicine and Health, University of Birmingham, Birmingham, United Kingdom
| | - Joseph Jacob
- UCL Respiratory, University College London, London, United Kingdom
- Satsuma Lab, Centre for Medical Image Computing, University College London Respiratory, University College London, London, United Kingdom
| | - Yatharth Ranjan
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Amos A. Folarin
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, King's College London, London, United Kingdom
- Institute of Health Informatics, University College London, London, United Kingdom
- NIHR Biomedical Research Centre at University College London Hospitals, NHS Foundation Trust, London, United Kingdom
| | - John R. Hurst
- Royal Free Campus, UCL Respiratory, University College London, London, United Kingdom
| | - Joanna C. Porter
- UCL Respiratory, University College London, London, United Kingdom
| |
Collapse
|
10
|
Mueller AN, Miller HA, Taylor MJ, Suliman SA, Frieboes HB. Identification of Idiopathic Pulmonary Fibrosis and Prediction of Disease Severity via Machine Learning Analysis of Comprehensive Metabolic Panel and Complete Blood Count Data. Lung 2024; 202:139-150. [PMID: 38376581 DOI: 10.1007/s00408-024-00673-7] [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: 11/24/2023] [Accepted: 01/24/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Diagnosis of idiopathic pulmonary fibrosis (IPF) typically relies on high-resolution computed tomography imaging (HRCT) or histopathology, while monitoring disease severity is done via frequent pulmonary function testing (PFT). More reliable and convenient methods of diagnosing fibrotic interstitial lung disease (ILD) type and monitoring severity would allow for early identification and enhance current therapeutic interventions. This study tested the hypothesis that a machine learning (ML) ensemble analysis of comprehensive metabolic panel (CMP) and complete blood count (CBC) data can accurately distinguish IPF from connective tissue disease ILD (CTD-ILD) and predict disease severity as seen with PFT. METHODS Outpatient data with diagnosis of IPF or CTD-ILD (n = 103 visits by 53 patients) were analyzed via ML methodology to evaluate (1) IPF vs CTD-ILD diagnosis; (2) %predicted Diffusing Capacity of Lung for Carbon Monoxide (DLCO) moderate or mild vs severe; (3) %predicted Forced Vital Capacity (FVC) moderate or mild vs severe; and (4) %predicted FVC mild vs moderate or severe. RESULTS ML methodology identified IPF from CTD-ILD with AUCTEST = 0.893, while PFT was classified as DLCO moderate or mild vs severe with AUCTEST = 0.749, FVC moderate or mild vs severe with AUCTEST = 0.741, and FVC mild vs moderate or severe with AUCTEST = 0.739. Key features included albumin, alanine transaminase, %lymphocytes, hemoglobin, %eosinophils, white blood cell count, %monocytes, and %neutrophils. CONCLUSION Analysis of CMP and CBC data via proposed ML methodology offers the potential to distinguish IPF from CTD-ILD and predict severity on associated PFT with accuracy that meets or exceeds current clinical practice.
Collapse
Affiliation(s)
- Alex N Mueller
- School of Medicine, University of Louisville, Louisville, KY, USA
| | - Hunter A Miller
- Department of Bioengineering, University of Louisville, Lutz Hall 419, Louisville, KY, 40292, USA
| | - Matthew J Taylor
- Division of Pulmonary Medicine, University of Louisville, Louisville, KY, USA
| | - Sally A Suliman
- University of Arizona Medical Center Phoenix, 755 East McDowell Road, Phoenix, AZ, 85006, USA.
- Formerly at: Division of Pulmonary Medicine, University of Louisville, Louisville, KY, USA.
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Lutz Hall 419, Louisville, KY, 40292, USA.
- Department of Pharmacology/Toxicology, University of Louisville, Louisville, KY, USA.
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
11
|
Schreiber J, Schütte W, Koerber W, Seese B, Koschel D, Neuland K, Grohé C. Clinical course of mild-to-moderate idiopathic pulmonary fibrosis during therapy with pirfenidone: Results of the non-interventional study AERplus. Pneumologie 2024; 78:236-243. [PMID: 38608658 PMCID: PMC11014748 DOI: 10.1055/a-2267-2074] [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: 11/08/2023] [Accepted: 01/11/2024] [Indexed: 04/14/2024]
Abstract
INTRODUCTION Pirfenidone was the first anti-fibrotic drug approved in Europe in 2011 for the treatment of mild-to-moderate idiopathic pulmonary fibrosis. OBJECTIVES To investigate the clinical course of mild-to-moderate idiopathic pulmonary fibrosis in pirfenidone-treated patients in a real-world setting. METHODS The non-interventional study was conducted at 18 sites in Germany from 6/2014-12/2016. Adult patients with mild-to-moderate idiopathic pulmonary fibrosis were treated with pirfenidone (escalated from 3×1 to 3×3 capsules of 267 mg/day within 3 weeks) for 12 months. The observation period comprised 4 follow-up visits at months 3, 6, 9 and 12. Disease progression was defined as decrease of ≥10% in vital capacity or ≥15% in diffusing capacity of the lung for carbon monoxide (DLCO) and/or ≥50m in 6-minute walking distance vs. baseline, or "lack of response/progression" as reason for therapy discontinuation. RESULTS A total of 51 patients (80.4% male, mean age 70.6 years) were included in the full analysis set. Disease progression at any visit was reported for 23 (67.6%) of 34 patients with available data. Over the course of the study, lung function parameters, physical resilience, impact of cough severity on quality of life, and the mean Gender, Age and Physiology Index (stage II) remained stable. In total, 29 patients (56.9%) experienced at least one adverse drug reaction (11 patients discontinued due to adverse drug reactions); serious adverse reactions were reported in 12 patients (23.5%). CONCLUSIONS The results of this study are in line with the established benefit-risk profile of pirfenidone. Therefore, pirfenidone can be considered a valuable treatment option to slow disease progression in mild-to-moderate idiopathic pulmonary fibrosis. NCT02622477.
Collapse
Affiliation(s)
- Jens Schreiber
- Pneumonology, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Wolfgang Schütte
- Klinik für Innere Medizin II, Krankenhaus Martha-Maria Halle-Dölau, Halle, Germany
| | - Wolfgang Koerber
- Abt. Pneumologie/Beatmungsmedizin und Schlaflabor, Evangelisches Krankenhaus Göttingen-Weende gGmbH, Bovenden-Lenglern, Germany
| | - Bernd Seese
- Abt. Pneumologie, Thoraxzentrum Bezirk Unterfranken, Münnerstadt, Germany
| | - Dirk Koschel
- Innere Medizin und Pneumologie, Fachkrankenhaus Coswig, Coswig, Germany
| | - Kathrin Neuland
- Global Scientific Communications, Roche Pharma AG, Grenzach-Wyhlen, Germany
| | - Christian Grohé
- Klinik für Pneumologie, Evangelische Lungenklinik Berlin, Berlin, Germany
| |
Collapse
|
12
|
Chen T, Zeng C. Compare three diagnostic criteria of progressive pulmonary fibrosis. J Thorac Dis 2024; 16:1034-1043. [PMID: 38505056 PMCID: PMC10944769 DOI: 10.21037/jtd-23-481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 12/08/2023] [Indexed: 03/21/2024]
Abstract
Background In patients with fibrotic interstitial lung disease (ILD), a progressive pulmonary fibrosis (PPF) typically demonstrates worsening respiratory symptoms, lung function decline and continuing fibrosis. The goal of this study was to compare the three different diagnostic criteria of PPF. Methods Except for idiopathic pulmonary fibrosis (IPF), all consecutive adult patients with fibrotic ILD were retrospectively examined for the three predefined diagnostic criteria of PPF. The three criteria assessed the disease progression in preceding 6 (0.5-year), 12 (1-year) and 24 (2-year) months respectively. The clinical characteristics, decline in predicted percent of forced vital capacity (FVC%) and survival of three groups were compared, followed by determination of risk factors for mortality. Results We identified 246 patients by 0.5-year standard, 154 patients by 1-year standard and 281 patients by 2-year standard. Among them, 95% of patients in 1-year group were also included in 2-year group. The average 1-year decline in FVC% was -1.0%, -2.7%, and -4.1% for 0.5-, 1-, and 2-year group respectively. The 4-year survival rate was 74% in 0.5-year group, 66% in 1-year group, and 62% in 2-year group. In multivariate Cox model, only baseline predicted percent diffusing capacity of the lungs for carbon monoxide (DLCO%) <50% was correlated with mortality, with a hazard ratio of 3.4 (95% CI: 1.1-10.6, P=0.03). Conclusions In the current situations, both the 1- and 2-year criterion are the reasonable choice to define PPF both in researches and clinical practice, and DLCO% is an independent predictor for mortality of PPF.
Collapse
Affiliation(s)
- Tao Chen
- Department of Respiratory and Critical Care Medicine, People's Hospital of Deyang City, Deyang, China
| | - Chunfang Zeng
- Department of Respiratory and Critical Care Medicine, People's Hospital of Deyang City, Deyang, China
| |
Collapse
|
13
|
Montesi SB, Gomez CR, Beers M, Brown R, Chattopadhyay I, Flaherty KR, Garcia CK, Gomperts B, Hariri LP, Hogaboam CM, Jenkins RG, Kaminski N, Kim GHJ, Königshoff M, Kolb M, Kotton DN, Kropski JA, Lasky J, Magin CM, Maher TM, McCormick M, Moore BB, Nickerson-Nutter C, Oldham J, Podolanczuk AJ, Raghu G, Rosas I, Rowe SM, Schmidt WT, Schwartz D, Shore JE, Spino C, Craig JM, Martinez FJ. Pulmonary Fibrosis Stakeholder Summit: A Joint NHLBI, Three Lakes Foundation, and Pulmonary Fibrosis Foundation Workshop Report. Am J Respir Crit Care Med 2024; 209:362-373. [PMID: 38113442 PMCID: PMC10878386 DOI: 10.1164/rccm.202307-1154ws] [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: 07/06/2023] [Accepted: 12/19/2023] [Indexed: 12/21/2023] Open
Abstract
Despite progress in elucidation of disease mechanisms, identification of risk factors, biomarker discovery, and the approval of two medications to slow lung function decline in idiopathic pulmonary fibrosis and one medication to slow lung function decline in progressive pulmonary fibrosis, pulmonary fibrosis remains a disease with a high morbidity and mortality. In recognition of the need to catalyze ongoing advances and collaboration in the field of pulmonary fibrosis, the NHLBI, the Three Lakes Foundation, and the Pulmonary Fibrosis Foundation hosted the Pulmonary Fibrosis Stakeholder Summit on November 8-9, 2022. This workshop was held virtually and was organized into three topic areas: 1) novel models and research tools to better study pulmonary fibrosis and uncover new therapies, 2) early disease risk factors and methods to improve diagnosis, and 3) innovative approaches toward clinical trial design for pulmonary fibrosis. In this workshop report, we summarize the content of the presentations and discussions, enumerating research opportunities for advancing our understanding of the pathogenesis, treatment, and outcomes of pulmonary fibrosis.
Collapse
Affiliation(s)
| | - Christian R. Gomez
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Michael Beers
- Pulmonary and Critical Care Division, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert Brown
- Program in Neurotherapeutics, University of Massachusetts Chan Medical School, Worchester, Massachusetts
| | | | | | - Christine Kim Garcia
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Irving Medical Center, New York, New York
| | | | - Lida P. Hariri
- Division of Pulmonary and Critical Care Medicine and
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Cory M. Hogaboam
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - R. Gisli Jenkins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Grace Hyun J. Kim
- Center for Computer Vision and Imaging Biomarkers, Department of Radiological Sciences, David Geffen School of Medicine, and
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| | - Melanie Königshoff
- Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Martin Kolb
- Division of Respirology, McMaster University, Hamilton, Ontario, Canada
| | - Darrell N. Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joseph Lasky
- Pulmonary Fibrosis Foundation, Chicago, Illinois
- Department of Medicine, Tulane University, New Orleans, Louisiana
| | - Chelsea M. Magin
- Department of Bioengineering
- Department of Pediatrics
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, and
| | - Toby M. Maher
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | | | | | | | - Anna J. Podolanczuk
- Division of Pulmonary and Critical Care, Weill Cornell Medical College, New York, New York
| | - Ganesh Raghu
- Division of Pulmonary, Sleep and Critical Care Medicine, University of Washington, Seattle, Washington
| | - Ivan Rosas
- Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, Texas; and
| | - Steven M. Rowe
- Department of Medicine and
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - David Schwartz
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Cathie Spino
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - J. Matthew Craig
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Fernando J. Martinez
- Division of Pulmonary and Critical Care, Weill Cornell Medical College, New York, New York
| |
Collapse
|
14
|
Walsh SLF, De Backer J, Prosch H, Langs G, Calandriello L, Cottin V, Brown KK, Inoue Y, Tzilas V, Estes E. Towards the adoption of quantitative computed tomography in the management of interstitial lung disease. Eur Respir Rev 2024; 33:230055. [PMID: 38537949 PMCID: PMC10966471 DOI: 10.1183/16000617.0055-2023] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 01/31/2024] [Indexed: 03/29/2025] Open
Abstract
The shortcomings of qualitative visual assessment have led to the development of computer-based tools to characterise and quantify disease on high-resolution computed tomography (HRCT) in patients with interstitial lung diseases (ILDs). Quantitative CT (QCT) software enables quantification of patterns on HRCT with results that are objective, reproducible, sensitive to change and predictive of disease progression. Applications developed to provide a diagnosis or pattern classification are mainly based on artificial intelligence. Deep learning, which identifies patterns in high-dimensional data and maps them to segmentations or outcomes, can be used to identify the imaging patterns that most accurately predict disease progression. Optimisation of QCT software will require the implementation of protocol standards to generate data of sufficient quality for use in computerised applications and the identification of diagnostic, imaging and physiological features that are robustly associated with mortality for use as anchors in the development of algorithms. Consortia such as the Open Source Imaging Consortium have a key role to play in the collation of imaging and clinical data that can be used to identify digital imaging biomarkers that inform diagnosis, prognosis and response to therapy.
Collapse
Affiliation(s)
- Simon L F Walsh
- National Heart and Lung Institute, Imperial College, London, UK
| | | | | | - Georg Langs
- Medical University of Vienna, Vienna, Austria
- contextflow GmbH, Vienna, Austria
| | | | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases, Louis Pradel Hospital, Hospices Civils de Lyon, Claude Bernard University Lyon 1, UMR 754, Lyon, France
| | - Kevin K Brown
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Yoshikazu Inoue
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai City, Japan
| | - Vasilios Tzilas
- 5th Respiratory Department, Chest Diseases Hospital Sotiria, Athens, Greece
| | | |
Collapse
|
15
|
Okamoto M, Fujimoto K, Johkoh T, Kawaguchi A, Mukae H, Sakamoto N, Ogura T, Ikeda S, Kondoh Y, Yamano Y, Komiya K, Umeki K, Nishikiori H, Tanino Y, Tsuda T, Arai N, Komatsu M, Sakamoto S, Yatera K, Inoue Y, Miyazaki Y, Hashimoto S, Shimizu Y, Hozumi H, Ohnishi H, Handa T, Hattori N, Kishaba T, Kato M, Inomata M, Ishii H, Hamada N, Konno S, Zaizen Y, Azuma A, Suda T, Izuhara K, Hoshino T. A prospective cohort study of periostin as a serum biomarker in patients with idiopathic pulmonary fibrosis treated with nintedanib. Sci Rep 2023; 13:22977. [PMID: 38151520 PMCID: PMC10752870 DOI: 10.1038/s41598-023-49180-4] [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: 04/27/2023] [Accepted: 12/05/2023] [Indexed: 12/29/2023] Open
Abstract
This study investigated the utility of periostin, a matricellular protein, as a prognostic biomarker in patients with idiopathic pulmonary fibrosis (IPF) who received nintedanib. Monomeric and total periostin levels were measured by enzyme-linked immunosorbent assay in 87 eligible patients who participated in a multicenter prospective study. Forty-three antifibrotic drug-naive patients with IPF described in previous studies were set as historical controls. Monomeric and total periostin levels were not significantly associated with the change in forced vital capacity (FVC) or diffusing capacity of the lungs for carbon monoxide (DLCO) during any follow-up period. Higher monomeric and total periostin levels were independent risk factors for overall survival in the Cox proportional hazard model. In the analysis of nintedanib effectiveness, higher binarized monomeric periostin levels were associated with more favorable suppressive effects on decreased vital capacity (VC) and DLCO in the treatment group compared with historical controls. Higher binarized levels of total periostin were associated with more favorable suppressive effects on decreased DLCO but not VC. In conclusion, higher periostin levels were independently associated with survival and better therapeutic effectiveness in patients with IPF treated with nintedanib. Periostin assessments may contribute to determining therapeutic strategies for patients with IPF.
Collapse
Affiliation(s)
- Masaki Okamoto
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan.
- Department of Respirology, NHO Kyushu Medical Center, 1-8-1 Jigyohama, Chuo-ku, Fukuoka, 810-0065, Japan.
| | - Kiminori Fujimoto
- Department of Radiology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Takeshi Johkoh
- Department of Radiology, Kansai Rosai Hospital, Inabasou 3-1-69, Amagasaki, Hyogo, 660-0064, Japan
| | - Atsushi Kawaguchi
- Education and Research Center for Community Medicine, Faculty of Medicine, Saga Medical School, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Noriho Sakamoto
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Takashi Ogura
- Division of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, 6-16-1 Tomiokahigashi, Yokohama, Kanagawa-ku, Kanagawa, 236-0051, Japan
| | - Satoshi Ikeda
- Division of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, 6-16-1 Tomiokahigashi, Yokohama, Kanagawa-ku, Kanagawa, 236-0051, Japan
| | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, 160 Nishioiwake, Seto, Aichi, 489-0065, Japan
| | - Yasuhiko Yamano
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, 160 Nishioiwake, Seto, Aichi, 489-0065, Japan
| | - Kosaku Komiya
- Respiratory Medicine and Infectious Diseases, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| | - Kenji Umeki
- Department of Respiratory Medicine, Tenshindo Hetsugi Hospital, 5956 Nakahetsugi, Oita, 879-7761, Japan
| | - Hirotaka Nishikiori
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, South-1-West-16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Yoshinori Tanino
- Department of Pulmonary Medicine, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima, 960-1295, Japan
| | - Toru Tsuda
- Kirigaoka Tsuda Hospital, 3-9-20 Kirigaoka, Kitakyushu, Fukuoka, 802-0052, Japan
| | - Naoki Arai
- Department of Respiratory Medicine, National Hospital Organization Ibarakihigashi National Hospital, 825 Terunuma, Tokai-mura, Ibaraki, 319-1113, Japan
| | - Masamichi Komatsu
- First Department of Internal Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Susumu Sakamoto
- Department of Respiratory Medicine, Toho University Omori Medical Center, 6-11-1 Omorinishi, Tokyo, 143-8541, Japan
| | - Kazuhiro Yatera
- Department of Respiratory Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yoshikazu Inoue
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, 1180 Nagasone-cho, Sakai, Osaka, 591-8555, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo, 113-8510, Japan
| | - Seishu Hashimoto
- Department of Respiratory Medicine, Tenri Hospital, 200 Mishima-cho, Tenri, Nara, 632-8552, Japan
| | - Yasuo Shimizu
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Shimotsuga, Tochigi, 321-0293, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Hiroshi Ohnishi
- Department of Respiratory Medicine and Allergology, Kochi Medical School, Kochi University, 185-1 Kohasu, Nankoku, Kochi, 783-8505, Japan
| | - Tomohiro Handa
- Department of Advanced Medicine for Respiratory Failure, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Tomoo Kishaba
- Department of Respiratory Medicine, Okinawa Chubu Hospital, 281 Miyazato, Uruma, Okinawa, 904-2293, Japan
| | - Motoyasu Kato
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Tokyo, 113-8421, Japan
| | - Minoru Inomata
- Department of Respiratory Medicine, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Tokyo, 150-8935, Japan
| | - Hiroshi Ishii
- Department of Respiratory Medicine, Fukuoka University Chikushi Hospital, 1-1-1 Zokumyouin, Chikushino, Fukuoka, 818-8502, Japan
| | - Naoki Hamada
- Department of Respiratory Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Fukuoka, 814-0180, Japan
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, N15W7 Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Yoshiaki Zaizen
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Arata Azuma
- Respirology and Clinical Research Center, Mihara General Hospital and Nippon Medical School, Tokorozawa, Saitama, 359-0045, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Sciences, Saga Medical School, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| |
Collapse
|
16
|
Yang R, Wang Y, Hao L, Zhao G, Liu X. Association of the 6-minute walking test ratio and difference with pulmonary function in patients with interstitial lung disease. Saudi Med J 2023; 44:1000-1005. [PMID: 37777275 PMCID: PMC10541989 DOI: 10.15537/smj.2023.44.10.20220940] [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: 06/24/2023] [Accepted: 08/31/2023] [Indexed: 10/02/2023] Open
Abstract
OBJECTIVES To examine the associations between 6-minute walk test (6MWT) and lung functions, blood gas analysis findings, fractional exhaled nitric oxide (FeNO), and hospital stay in interstitial lung disease (ILD). METHODS The present retrospective study included patients hospitalized in Beijing Hospital of Traditional Chinese Medicine, Capital Medical University between September 2018 and December 2019. The outcomes included the difference between the actual and predicted 6MWT values (6MWT difference) and the ratio of the actual to predicted 6MWT value (6MWT ratio). RESULTS This study included 137 patients. The predicted 6MWT value was 519±61 m and the actual 6MWT value was 449 (196.5,694)m. The 6MWT ratio was 84.7±177.6 and 6MWT difference was 73.9±95.1 m. Fractional exhaled nitric oxide (FeNO) (β= -2.157, standard error [SE]=0.836, p=0.014) and diffusing capacity of the lungs for carbon monoxide (DLCO) (β= -22.528, SE=7.48, p=0.004) had independent associations with 6MWT difference. The FeNO (β=0.403, SE=0.163, p=0.018) and DLCO (β=4.355, SE=1.458, p=0.005) had independent associations with 6MWT ratio. CONCLUSION In ILD, 6MWT difference and 6MWT ratio were associated with FeNO and DLCO. The 6MWT value was not associated with hospital stay. Therefore, the 6MWT might be a surrogate marker of pulmonary function in clinical ILD.
Collapse
Affiliation(s)
- Ru Yang
- From the Respiratory Department (Yang, Wang, Liu); and Nursing Department (Hao, Zhao), Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Yuguang Wang
- From the Respiratory Department (Yang, Wang, Liu); and Nursing Department (Hao, Zhao), Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Li Hao
- From the Respiratory Department (Yang, Wang, Liu); and Nursing Department (Hao, Zhao), Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Guomin Zhao
- From the Respiratory Department (Yang, Wang, Liu); and Nursing Department (Hao, Zhao), Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Xitong Liu
- From the Respiratory Department (Yang, Wang, Liu); and Nursing Department (Hao, Zhao), Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
17
|
Neely ML, Hellkamp AS, Bender S, Todd JL, Liesching T, Luckhardt TR, Oldham JM, Raj R, White ES, Palmer SM. Lung function trajectories in patients with idiopathic pulmonary fibrosis. Respir Res 2023; 24:209. [PMID: 37612608 PMCID: PMC10463468 DOI: 10.1186/s12931-023-02503-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 08/01/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing interstitial lung disease characterised by decline in lung function. We evaluated trajectories of forced vital capacity (FVC) and diffusing capacity (DLco) in a cohort of patients with IPF. METHODS Patients with IPF that was diagnosed or confirmed at the enrolling centre in the previous 6 months were enrolled into the IPF-PRO Registry between June 2014 and October 2018. Patients were followed prospectively, with lung function data collected as part of routine clinical care. Mean trajectories of FVC and DLco % predicted in all patients and in subgroups by characteristics assessed at enrolment were estimated using a joint model that accounted for factors such as disease severity and visit patterns. RESULTS Of 1002 patients in the registry, 941 had ≥ 1 FVC and/or DLco measurement after enrolment. The median (Q1, Q3) follow-up period was 35.1 (18.9, 47.2) months. Overall, mean estimated declines in FVC and DLco % predicted were 2.8% and 2.9% per year, respectively. There was no evidence that the mean trajectories of FVC or DLco had a non-linear relationship with time at the population level. Patients who were male, white, had a family history of ILD, were using oxygen, or had prior/current use of antifibrotic therapy at enrolment had greater rates of decline in FVC % predicted. Patients who were male or white had greater rates of decline in DLco % predicted. CONCLUSIONS Data from the IPF-PRO Registry suggest a constant rate of decline in lung function over a prolonged period, supporting the inexorably progressive nature of IPF. A graphical abstract summarising the data in this manuscript is available at: https://www.usscicomms.com/respiratory/IPF-PRORegistry_LungFunctionTrajectories . TRIAL REGISTRATION NCT01915511.
Collapse
Affiliation(s)
- Megan L Neely
- Duke Clinical Research Institute, Durham, NC, USA.
- Duke University Medical Center, Durham, NC, USA.
| | - Anne S Hellkamp
- Duke Clinical Research Institute, Durham, NC, USA
- Duke University Medical Center, Durham, NC, USA
| | - Shaun Bender
- Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Jamie L Todd
- Duke Clinical Research Institute, Durham, NC, USA
- Duke University Medical Center, Durham, NC, USA
| | | | - Tracy R Luckhardt
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Justin M Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rishi Raj
- Stanford University School of Medicine, Stanford, CA, USA
| | - Eric S White
- Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Scott M Palmer
- Duke Clinical Research Institute, Durham, NC, USA
- Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
18
|
Tomassetti S, Ravaglia C, Piciucchi S, Ryu J, Wells A, Donati L, Dubini A, Klersy C, Luzzi V, Gori L, Rosi E, Lavorini F, Poletti V. Historical eye on IPF: a cohort study redefining the mortality scenario. Front Med (Lausanne) 2023; 10:1151922. [PMID: 37332746 PMCID: PMC10273674 DOI: 10.3389/fmed.2023.1151922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/26/2023] [Indexed: 06/20/2023] Open
Abstract
Rationale Therapies that slow idiopathic pulmonary fibrosis (IPF) progression are now available and recent studies suggest that the use of antifibrotic therapy may reduce IPF mortality. Objectives The aim of the study was to evaluate whether, to what extent, and for which factors the survival of IPF in a real-life setting has changed in the last 15 years. Methods Historical eye is an observational study of a large cohort of consecutive IPF patients diagnosed and treated in a referral center for ILDs with prospective intention. We recruited all consecutive IPF patients seen at GB Morgagni Hospital, Forlì, Italy between January 2002 and December 2016 (15 years). We used survival analysis methods to describe and model the time to death or lung transplant and Cox regression to model prevalent and incident patient characteristics (time-dependent Cox models were fitted). Measurements and main results The study comprised 634 patients. The year 2012 identifies the time point of mortality shift (HR 0.58, CI 0.46-0.63, p < 0.001). In the more recent cohort, more patients had better preserved lung function, underwent cryobiopsy instead of surgery, and were treated with antifibrotics. Highly significant negative prognostic factors were lung cancer (HR 4.46, 95% CI 3.3-6, p < 0.001), hospitalizations (HR 8.37, 95% CI 6.5-10.7, p < 0.001), and acute exacerbations (HR 8.37, 95% CI 6.52-10.7, p < 0.001). The average antifibrotic treatment effect estimated using propensity score matching showed a significant effect in the reduction of all-cause mortality (ATE coeff -0.23, SE 0.04, p < 0.001), acute exacerbations (ATE coeff -0.15, SE 0.04, p < 0.001), and hospitalizations (ATE coeff -0.15, SE 0.04, p < 0.001) but no effect on lung cancer risk (ATE coeff -0.03, SE 0.03, p = 0.4). Conclusion Antifibrotic drugs significantly impact hospitalizations, acute exacerbations, and IPF survival. After the introduction of cryobiopsy and antifibrotic drugs, the prognosis of IPF patients has significantly improved together with our ability to detect IPF at an earlier stage.
Collapse
Affiliation(s)
- Sara Tomassetti
- Department of Clinical and Experimental Medicine, Interventional Pulmonology Unit, Careggi University Hospital, Florence, Italy
| | - Claudia Ravaglia
- Pulmonary Unit, Department of Diseases of the Thorax, GB Morgagni Hospital, Forlì, Italy
| | - Sara Piciucchi
- Radiology Department, Ospedale GB Morgagni, Forlì, Italy
| | - Jay Ryu
- Respiratory and Critical Care Medicine, Mayo Clinic, Rochester, MN, United States
| | - Athol Wells
- ILD Unit, Pulmonary Medicine, Royal Brompton Hospital, London, United Kingdom
| | - Luca Donati
- Pulmonary Unit, Department of Diseases of the Thorax, GB Morgagni Hospital, Forlì, Italy
| | | | - Catherine Klersy
- Servizio di Biometria ed Epidemiologia Clinica, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Valentina Luzzi
- Department of Clinical and Experimental Medicine, Interventional Pulmonology Unit, Careggi University Hospital, Florence, Italy
| | - Leonardo Gori
- Department of Clinical and Experimental Medicine, Interventional Pulmonology Unit, Careggi University Hospital, Florence, Italy
| | - Elisabetta Rosi
- Department of Clinical and Experimental Medicine, Interventional Pulmonology Unit, Careggi University Hospital, Florence, Italy
| | - Federico Lavorini
- Department of Clinical and Experimental Medicine, Interventional Pulmonology Unit, Careggi University Hospital, Florence, Italy
| | - Venerino Poletti
- Pulmonary Unit, Department of Diseases of the Thorax, GB Morgagni Hospital, Forlì, Italy
- Department of Respiratory Diseases and Allergology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
19
|
Wang BR, Oldham JM. Clinical Trial Enrichment in Systemic Sclerosis-Associated Interstitial Lung Disease: The Search Continues. Chest 2023; 163:475-476. [PMID: 36894257 DOI: 10.1016/j.chest.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 03/09/2023] Open
Affiliation(s)
- Bonnie R Wang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, University of Michigan, Ann Arbor, MI.
| | - Justin M Oldham
- Division of Pulmonary and Critical Care, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| |
Collapse
|
20
|
Rocha V, Paixão C, Marques A. Physical activity, exercise capacity and mortality risk in people with interstitial lung disease: A systematic review and meta-analysis. J Sci Med Sport 2022; 25:903-910. [DOI: 10.1016/j.jsams.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/22/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
|
21
|
Piotrowski WJ, Martusewicz-Boros MM, Białas AJ, Barczyk A, Batko B, Błasińska K, Boros PW, Górska K, Grzanka P, Jassem E, Jastrzębski D, Kaczyńska J, Kowal-Bielecka O, Kucharz E, Kuś J, Kuźnar-Kamińska B, Kwiatkowska B, Langfort R, Lewandowska K, Mackiewicz B, Majewski S, Makowska J, Miłkowska-Dymanowska J, Puścińska E, Siemińska A, Sobiecka M, Soroka-Dąda RA, Szołkowska M, Wiatr E, Ziora D, Śliwiński P. Guidelines of the Polish Respiratory Society on the Diagnosis and Treatment of Progressive Fibrosing Interstitial Lung Diseases Other than Idiopathic Pulmonary Fibrosis. Adv Respir Med 2022; 90:425-450. [PMID: 36285980 PMCID: PMC9717335 DOI: 10.3390/arm90050052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 08/24/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2023]
Abstract
The recommendations were developed as answers to previously formulated questions concerning everyday diagnostic and therapeutic challenges. They were developed based on a review of the current literature using the GRADE methodology. The experts suggest that PF-ILD be diagnosed based on a combination of different criteria, such as the aggravation of symptoms, progression of radiological lesions, and worsening of lung function test parameters. The experts recommend a precise diagnosis of an underlying disease, with serological testing for an autoimmune disease always being included. The final diagnosis should be worked out by a multidisciplinary team (MDT). Patients with an interstitial lung disease other than IPF who do not meet the criteria for the progressive fibrosis phenotype should be monitored for progression, and those with systemic autoimmune diseases should be regularly monitored for signs of interstitial lung disease. In managing patients with interstitial lung disease associated with autoimmune diseases, an opinion of an MDT should be considered. Nintedanib rather than pirfenidon should be introduced in the event of the ineffectiveness of the therapy recommended for the treatment of the underlying disease, but in some instances, it is possible to start antifibrotic treatment without earlier immunomodulatory therapy. It is also admissible to use immunomodulatory and antifibrotic drugs simultaneously. No recommendations were made for or against termination of anti-fibrotic therapy in the case of noted progression during treatment of a PF-ILD other than IPF. The experts recommend that the same principles of non-pharmacological and palliative treatment and eligibility for lung transplantation should be applied to patients with an interstitial lung disease other than IPF with progressive fibrosis as in patients with IPF.
Collapse
Affiliation(s)
| | - Magdalena M. Martusewicz-Boros
- 3rd Lung Diseases and Oncology Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Adam J. Białas
- Department of Pathobiology of Respiratory Diseases, Medical University of Lodz, 90-153 Lodz, Poland
| | - Adam Barczyk
- Department of Pneumonology, School of Medicine in Katowice, Medical University of Silesia, 40-635 Katowice, Poland
| | - Bogdan Batko
- Department of Rheumatology and Immunology, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University, 30-705 Krakow, Poland
| | - Katarzyna Błasińska
- Department of Radiology, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Piotr W. Boros
- Lung Pathophysiology Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Katarzyna Górska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Piotr Grzanka
- Department of Radiology, Voivodeship Hospital in Opole, 45-061 Opole, Poland
| | - Ewa Jassem
- Department of Allergology and Pneumonology, Medical University of Gdansk, 80-214 Gdańsk, Poland
| | - Dariusz Jastrzębski
- Department of Lung Diseases and Tuberculosis, Medical University of Silesia, 41-803 Zabrze, Poland
| | | | - Otylia Kowal-Bielecka
- Department of Rheumatology and Internal Medicine, Medical University of Białystok, 15-276 Białystok, Poland
| | - Eugeniusz Kucharz
- Department of Internal Medicine, Rheumatology and Clinical Immunology, Medical University of Silesia, 40-635 Katowice, Poland
| | - Jan Kuś
- 1st Lung Diseases Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Barbara Kuźnar-Kamińska
- Department of Pulmonology, Allergology and Respiratory Oncology, University of Medical Sciences in Poznan, 61-701 Poznan, Poland
| | - Brygida Kwiatkowska
- Department of Rheumatology, Eleonora Reicher Rheumatology Institute, 02-637 Warszawa, Poland
| | - Renata Langfort
- Department of Pathology, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warszawa, Poland
| | - Katarzyna Lewandowska
- 1st Lung Diseases Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Barbara Mackiewicz
- Department of Pneumonology, Oncology and Allergology, Medical University, Lublin, 20-090 Lublin, Poland
| | - Sebastian Majewski
- Department of Pneumology, Medical University of Lodz, 90-153 Lodz, Poland
| | - Joanna Makowska
- Department of Rheumatology, Medical University of Lodz, 92-213 Lodz, Poland
| | | | - Elżbieta Puścińska
- 2nd Department of Respiratory Medicine, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Alicja Siemińska
- Department of Allergology, Medical University of Gdańsk, 80-214 Gdansk, Poland
| | - Małgorzata Sobiecka
- 1st Lung Diseases Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | | | - Małgorzata Szołkowska
- Department of Pathology, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warszawa, Poland
| | - Elżbieta Wiatr
- 3rd Lung Diseases and Oncology Department, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| | - Dariusz Ziora
- Department of Lung Diseases and Tuberculosis, Medical University of Silesia, 41-803 Zabrze, Poland
| | - Paweł Śliwiński
- 2nd Department of Respiratory Medicine, National Tuberculosis and Lung Diseases Research Institute in Warsaw, 01-138 Warsaw, Poland
| |
Collapse
|
22
|
Khan FA, Stewart I, Moss S, Fabbri L, Robinson KA, Johnson SR, Jenkins RG. Three-Month FVC Change: A Trial Endpoint for Idiopathic Pulmonary Fibrosis Based on Individual Participant Data Meta-analysis. Am J Respir Crit Care Med 2022; 205:936-948. [PMID: 35020580 DOI: 10.1164/rccm.202109-2091oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/11/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Novel therapies for idiopathic pulmonary fibrosis (IPF) are in development, but there remains uncertainty about the optimal trial endpoint. An earlier endpoint would enable assessment of a greater number of therapies in adaptive trial designs. Objectives: To determine whether short-term changes in FVC, DlCO, and six-minute-walk distance could act as surrogate endpoints to accelerate early-phase trials in IPF. Methods: Individual participant data (IPD) from IPF clinical trials were included in a two-step random-effects meta-analysis to determine whether baseline or 3-month changes in FVC, DlCO, and 6-minute-walk distance were associated with mortality or disease progression in placebo arms. Three-month and 12-month FVC decline endpoints were compared with treatment arm data from antifibrotic studies by meta-regression. Measurements and Main Results: IPD were available from 12 placebo cohorts totaling 1,819 participants, with baseline and 3-month changes in all physiological variables independently associated with poorer outcomes. Treatment data were available from six cohorts with 1,684 participants. For each 2.5% relative decline in FVC over 3 months, there was an associated 15% (adjusted hazard ratio, 1.15; 95% confidence interval [CI], 1.06-1.24; I2 = 59.4%) and 20% (adjusted hazard ratio, 1.20; 95% CI, 1.12-1.28; I2 = 18.0%) increased risk for mortality in untreated and treated individuals, respectively. An FVC change treatment effect was observed between treatment and placebo arms at 3 months (difference in FVC change of 42.9 ml; 95% CI, 24.0-61.8 ml; P < 0.001). Conclusions: IPD meta-analysis demonstrated that 3-month changes in physiological variables, particularly FVC, were associated with mortality among individuals with IPF. FVC change over 3 months may hold potential as a surrogate endpoint in IPF adaptive trials.
Collapse
Affiliation(s)
- Fasihul A Khan
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nottingham Biomedical Research Centre, National Institute for Health Research, Nottingham, United Kingdom
| | - Iain Stewart
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nottingham Biomedical Research Centre, National Institute for Health Research, Nottingham, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Samuel Moss
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nottingham Biomedical Research Centre, National Institute for Health Research, Nottingham, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Laura Fabbri
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nottingham Biomedical Research Centre, National Institute for Health Research, Nottingham, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | | | - Simon R Johnson
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nottingham Biomedical Research Centre, National Institute for Health Research, Nottingham, United Kingdom
| | - R Gisli Jenkins
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Nottingham Biomedical Research Centre, National Institute for Health Research, Nottingham, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| |
Collapse
|
23
|
Silva H, Mantoani LC, Zamboti CL, Aguiar WF, Ries AL, Gonçalves AFL, da Silva TG, Ribeiro M, Pitta F, Camillo CA. Validation of the Brazilian Portuguese version of the University of California San Diego Shortness of Breath Questionnaire in patients with interstitial lung disease. J Bras Pneumol 2021; 47:e20210172. [PMID: 34932719 PMCID: PMC8836616 DOI: 10.36416/1806-3756/e20210172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To investigate the reliability, internal consistency and validity of the Brazilian Portuguese version of the University of California San Diego Shortness of Breath Questionnaire (UCSD SOBQ) in patients with interstitial lung disease (ILD). METHODS Patients with ILD completed the questionnaire at three different time points, one week apart, with the assistance of two independent assessors. Intra- and inter-rater reliability were analysed via the intraclass correlation coefficient (ICC). Internal consistency was assessed with the Cronbach's alpha coefficient. For the validity analysis, associations between variables were assessed with Spearman's or Pearson's correlation coefficient. RESULTS Thirty patients with ILD (idiopathic pulmonary fibrosis, connective tissue disease-associated pulmonary fibrosis, sarcoidosis, asbestosis or non-specific interstitial pneumonia) were included (15 men; mean age, 59 ± 10 years; DLCO: 46 [33-64] % predicted). UCSD SOBQ scores showed excellent agreement and internal consistency in the intra-rater analysis (ICC: 0.93 [0.85-0.97]; Cronbach alpha: 0.95) and in the inter-rater analysis (ICC: 0.95 [0.89-0.97]; Cronbach alpha: 0.95), as well as correlating significantly with dyspnoea (as assessed by the Medical Research Council scale; r = 0.56); Medical Outcomes Study 36-item Short-Form Health Survey domains bodily pain, general health, vitality and physical functioning (-0.40 ≤ r ≤ -0.74); six-minute walk distance (r = -0.38); and quadriceps muscle strength (r = -0.41). CONCLUSIONS The Brazilian Portuguese version of the UCSD SOBQ is valid, is reliable and has internal consistency in patients with ILD in Brazil.
Collapse
Affiliation(s)
- Humberto Silva
- . Laboratório de Pesquisa em Fisioterapia Pulmonar - LFIP - Departamento de Fisioterapia, Universidade Estadual de Londrina - UEL - Londrina (PR) Brasil
| | - Leandro Cruz Mantoani
- . Laboratório de Pesquisa em Fisioterapia Pulmonar - LFIP - Departamento de Fisioterapia, Universidade Estadual de Londrina - UEL - Londrina (PR) Brasil
| | - Camile Ludovico Zamboti
- . Laboratório de Pesquisa em Fisioterapia Pulmonar - LFIP - Departamento de Fisioterapia, Universidade Estadual de Londrina - UEL - Londrina (PR) Brasil
| | - Wagner Florentin Aguiar
- . Laboratório de Pesquisa em Fisioterapia Pulmonar - LFIP - Departamento de Fisioterapia, Universidade Estadual de Londrina - UEL - Londrina (PR) Brasil
| | - Andrew L. Ries
- . School of Medicine, University of California San Diego, San Diego (CA) USA
| | - Aline Ferreira Lima Gonçalves
- . Laboratório de Pesquisa em Fisioterapia Pulmonar - LFIP - Departamento de Fisioterapia, Universidade Estadual de Londrina - UEL - Londrina (PR) Brasil
| | - Thatielle Garcia da Silva
- . Laboratório de Pesquisa em Fisioterapia Pulmonar - LFIP - Departamento de Fisioterapia, Universidade Estadual de Londrina - UEL - Londrina (PR) Brasil
| | - Marcos Ribeiro
- . Departamento de Pneumologia, Universidade Estadual de Londrina - UEL - Londrina (PR) Brasil
| | - Fabio Pitta
- . Laboratório de Pesquisa em Fisioterapia Pulmonar - LFIP - Departamento de Fisioterapia, Universidade Estadual de Londrina - UEL - Londrina (PR) Brasil
| | - Carlos Augusto Camillo
- . Laboratório de Pesquisa em Fisioterapia Pulmonar - LFIP - Departamento de Fisioterapia, Universidade Estadual de Londrina - UEL - Londrina (PR) Brasil
- . Departamento de Ciências da Reabilitação, Universidade Pitágoras-Universidade Norte do Paraná - UNOPAR - Londrina (PR) Brasil
| |
Collapse
|
24
|
Jessen H, Hoyer N, Prior TS, Frederiksen P, Rønnow SR, Karsdal MA, Leeming DJ, Bendstrup E, Sand JMB, Shaker SB. Longitudinal serological assessment of type VI collagen turnover is related to progression in a real-world cohort of idiopathic pulmonary fibrosis. BMC Pulm Med 2021; 21:382. [PMID: 34814865 PMCID: PMC8609852 DOI: 10.1186/s12890-021-01684-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/29/2021] [Indexed: 12/02/2022] Open
Abstract
Background Remodeling of the extracellular matrix (ECM) is a central mechanism in the progression of idiopathic pulmonary fibrosis (IPF), and remodeling of type VI collagen has been suggested to be associated with disease progression. Biomarkers that reflect and predict the progression of IPF would provide valuable information for clinicians when treating IPF patients. Methods Two serological biomarkers reflecting formation (PRO-C6) and degradation (C6M) of type VI collagen were evaluated in a real-world cohort of 178 newly diagnoses IPF patients. All patients were treatment naïve at the baseline visit. Blood samples and clinical data were collected from baseline, six months, and 12 months visit. The biomarkers were measured by competitive ELISA using monoclonal antibodies. Results Patients with progressive disease had higher (P = 0.0099) serum levels of PRO-C6 compared to those with stable disease over 12 months with an average difference across all timepoints of 12% (95% CI 3–22), whereas C6M levels tended (P = 0.061) to be higher in patients with progressive disease compared with stable patients over 12 months with an average difference across all timepoints of 12% (95% CI − 0.005–27). Patients who did not receive antifibrotic medicine had a greater increase of C6M (P = 0.043) compared to treated patients from baseline over 12 months with an average difference across all timepoints of 12% (95% CI − 0.07–47). There were no differences in biomarker levels between patients receiving pirfenidone or nintedanib. Conclusions Type VI collagen formation was related to progressive disease in patients with IPF in a real-world cohort and antifibrotic therapy seemed to affect the degradation of type VI collagen. Type VI collagen formation and degradation products might be potential biomarkers for disease progression in IPF. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-021-01684-3.
Collapse
Affiliation(s)
- Henrik Jessen
- Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark. .,Department of Respiratory Medicine, Herlev and Gentofte University Hospital, Copenhagen, Denmark.
| | - Nils Hoyer
- Department of Respiratory Medicine, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Thomas S Prior
- Department of Respiratory Disease and Allergy, Center for Rare Lung Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Peder Frederiksen
- Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Sarah R Rønnow
- Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Morten A Karsdal
- Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Diana J Leeming
- Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Elisabeth Bendstrup
- Department of Respiratory Disease and Allergy, Center for Rare Lung Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Jannie M B Sand
- Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Saher B Shaker
- Department of Respiratory Medicine, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| |
Collapse
|
25
|
Caminati A, Madotto F, Conti S, Cesana G, Mantovani L, Harari S. The natural history of idiopathic pulmonary fibrosis in a large European population: the role of age, sex and comorbidities. Intern Emerg Med 2021; 16:1793-1802. [PMID: 33586036 DOI: 10.1007/s11739-021-02651-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/21/2021] [Indexed: 02/08/2023]
Abstract
Placebo arms of clinical trials provide an opportunity to investigate the natural history of idiopathic pulmonary fibrosis (IPF) but these patients are not representative of the real life IPF population. Objective of this article is to evaluate patients' characteristics of incident IPF cases and their impact on mortality and hospitalizations risk. We conducted a retrospective cohort study using data from administrative databases from 2000 to 2010. Based on different algorithms reported in literature, incident IPF cases were identified. We applied Cox proportional hazards models to assess relationship between patients' characteristics, mortality and hospitalization. According to three case definitions, we identified 2338, 460 and 1704 incident IPF cases. Mean age at diagnosis was about 72 years, the proportion of male varied between 59 and 62% and patients with at least one chronic disease were between 70 and 74%. Age, male sex and comorbidities were associated to worse outcomes. Congestive heart failure (CHF), diabetes and cancer were conditions associated to mortality, while those associated to hospitalization were CHF and chronic obstructive pulmonary disease. Our data source provided one of the largest samples of unselected patients with a long follow-up period. Using different algorithms proposed and validated in literature, we observed that mortality and hospitalization rate are high in patients with IPF and age, sex and comorbidities significantly affect clinical outcomes. Females show a significant survival advantage over males, even after adjusting for age and comorbidities. Patients with pre-existing diseases, especially those with pulmonary and cardiovascular diseases are at higher risk.
Collapse
Affiliation(s)
- Antonella Caminati
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare. Ospedale San Giuseppe, MultiMedica IRCCS, Via San Vittore 12, 20123, Milan, MI, Italy.
| | - Fabiana Madotto
- Value-based Healthcare Unit, IRCCS MultiMedica, Via Milanese 300, Sesto San Giovanni, 20099, Milan, Italy
- Research Centre on Public Health, Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900, Monza, Monza e Brianza, Italy
| | - Sara Conti
- Research Centre on Public Health, Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900, Monza, Monza e Brianza, Italy
| | - Giancarlo Cesana
- Research Centre on Public Health, Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900, Monza, Monza e Brianza, Italy
| | - Lorenzo Mantovani
- Value-based Healthcare Unit, IRCCS MultiMedica, Via Milanese 300, Sesto San Giovanni, 20099, Milan, Italy
- Research Centre on Public Health, Department of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900, Monza, Monza e Brianza, Italy
| | - Sergio Harari
- U.O. di Pneumologia e Terapia Semi-Intensiva Respiratoria, Servizio di Fisiopatologia Respiratoria ed Emodinamica Polmonare. Ospedale San Giuseppe, MultiMedica IRCCS, Via San Vittore 12, 20123, Milan, MI, Italy
- Clinica Medica, Ospedale San Giuseppe MultiMedica IRCCS, Via San Vittore 12, 20123, Milan, Italy
- Dipartimento di Scienze Cliniche e di Comunità, Università di Milano, Milan, Italy
| |
Collapse
|
26
|
Wells AU, Kouranos V. An IPF-like disease course in disorders other than IPF: how can this be anticipated, recognized, and managed? Expert Rev Clin Immunol 2021; 17:1091-1101. [PMID: 34467827 DOI: 10.1080/1744666x.2021.1968832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF)-like chronic disease progression despite treatment cannot be predicted with confidence in interstitial lung diseases (ILDs) other than IPF at the time of diagnosis. AREAS COVERED We review key determinants of a progressive fibrotic phenotype, at initial diagnosis of an ILD other than IPF. Medline literature searches (2000 to 2020) were undertaken with regard to the issues discussed in this review. EXPERT OPINION The definition of the progressive fibrotic phenotype in non-IPF patients should remain real world, with a conclusion reached by an experienced clinician that progression has occurred despite the use of appropriate historical therapies, on a case by case basis. There is an urgent need for pathogenetic studies to identify pathways and genetic predilections that are common to chronic progressive fibrosis across different diseases. Efforts should also be focused on the identification of the progressive fibrotic phenotype at first presentation, potentially through a combination of CT and biopsy evaluation and the definition of a biomarker profile associated with subsequent disease progression. Recent anti-fibrotic trials of non-IPF disorders should lead to trials of combination regimens of anti-fibrotic agents and immunomodulatory or other therapies specific to individual diseases.
Collapse
Affiliation(s)
- Athol U Wells
- Interstitial Lung Disease Unit, Royal Brompton Hospital, London, UK
| | | |
Collapse
|
27
|
Novel Artificial Intelligence-based Technology for Chest Computed Tomography Analysis of Idiopathic Pulmonary Fibrosis. Ann Am Thorac Soc 2021; 19:399-406. [PMID: 34410886 DOI: 10.1513/annalsats.202101-044oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RATIONALE There is a growing need to accurately estimate the prognosis of idiopathic pulmonary fibrosis (IPF) in clinical practice, given the development of effective drugs for treating IPF. OBJECTIVE To develop artificial intelligence-based image analysis software to detect parenchymal and airway abnormalities on chest computed tomography (CT) and to explore their prognostic importance in patients with IPF. METHODS A novel artificial intelligence-based quantitative CT image analysis software (AIQCT) was developed by applying 304 HRCT scans from patients with diffuse lung diseases as the training set. AIQCT automatically categorized and quantified ten types of parenchymal patterns as well as airways, expressing the volumes as percentages of the total lung volume. To validate the software, the area percentages of each lesion quantified by AIQCT were compared with those of the visual scores using 30 plain HRCT images with lung diseases. In addition, three-dimensional analysis for similarity with ground truth was performed using HRCT images from 10 patients with IPF. AIQCT was then applied to 120 patients with IPF who underwent chest HRCT scanning at our institute. Associations between the measured volumes and survival were analyzed. RESULTS The correlations between AIQCT and the visual scores were moderate to strong (correlation coefficient 0.44 to 0.95) depending on the parenchymal pattern. The Dice indexes for similarity between AIQCT data and ground truth were 0.67, 0.76, and 0.64 for reticulation, honeycomb, and bronchi, respectively. During a median follow-up period of 2,184 days, 66 patients died, and 1 underwent lung transplantation. In multivariable Cox regression analysis, bronchial volumes [adjusted hazard ratio (HR), 1.33; 95% confidence interval (CI), 1.16 to 1.53] and normal lung volumes (adjusted HR, 0.97; 95% CI, 0.94 to 0.99) were independently associated with survival after adjusting for the GAP stage of IPF. CONCLUSIONS Our newly developed artificial intelligence-based image analysis software successfully quantified parenchymal lesions and airway volumes. Bronchial and normal lung volumes on chest HRCT may provide additional prognostic information on the GAP stage of IPF.
Collapse
|
28
|
Huang Y, Oldham JM, Ma SF, Unterman A, Liao SY, Barros AJ, Bonham CA, Kim JS, Vij R, Adegunsoye A, Strek ME, Molyneaux PL, Maher TM, Herazo-Maya JD, Kaminski N, Moore BB, Martinez FJ, Noth I. Blood Transcriptomics Predicts Progression of Pulmonary Fibrosis and Associated Natural Killer Cells. Am J Respir Crit Care Med 2021; 204:197-208. [PMID: 33689671 PMCID: PMC8650792 DOI: 10.1164/rccm.202008-3093oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 03/08/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: Disease activity in idiopathic pulmonary fibrosis (IPF) remains highly variable, poorly understood, and difficult to predict. Objectives: To identify a predictor using short-term longitudinal changes in gene expression that forecasts future FVC decline and to characterize involved pathways and cell types. Methods: Seventy-four patients from COMET (Correlating Outcomes with Biochemical Markers to Estimate Time-Progression in IPF) cohort were dichotomized as progressors (≥10% FVC decline) or stable. Blood gene-expression changes within individuals were calculated between baseline and 4 months and regressed with future FVC status, allowing determination of expression variations, sample size, and statistical power. Pathway analyses were conducted to predict downstream effects and identify new targets. An FVC predictor for progression was constructed in COMET and validated using independent cohorts. Peripheral blood mononuclear single-cell RNA-sequencing data from healthy control subjects were used as references to characterize cell type compositions from bulk peripheral blood mononuclear RNA-sequencing data that were associated with FVC decline. Measurements and Main Results: The longitudinal model reduced gene-expression variations within stable and progressor groups, resulting in increased statistical power when compared with a cross-sectional model. The FVC predictor for progression anticipated patients with future FVC decline with 78% sensitivity and 86% specificity across independent IPF cohorts. Pattern recognition receptor pathways and mTOR pathways were downregulated and upregulated, respectively. Cellular deconvolution using single-cell RNA-sequencing data identified natural killer cells as significantly correlated with progression. Conclusions: Serial transcriptomic change predicts future FVC decline. An analysis of cell types involved in the progressor signature supports the novel involvement of natural killer cells in IPF progression.
Collapse
Affiliation(s)
- Yong Huang
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - Justin M. Oldham
- Division of Pulmonary, Critical Care, and Sleep Medicine, The University of California at Davis, Sacramento, California
| | - Shwu-Fan Ma
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - Avraham Unterman
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Shu-Yi Liao
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Andrew J. Barros
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - Catherine A. Bonham
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - John S. Kim
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| | - Rekha Vij
- Section of Pulmonary and Critical Care Medicine and
| | - Ayodeji Adegunsoye
- Section of Pulmonary and Critical Care Medicine and
- Department of Human Genetics, Genetics, Genomic and Systems Biology, University of Chicago, Chicago, Illinois
| | | | - Philip L. Molyneaux
- National Heart and Lung Institute, Imperial College, London, United Kingdom
- Royal Brompton Hospital, London, United Kingdom
| | - Toby M. Maher
- National Heart and Lung Institute, Imperial College, London, United Kingdom
- Royal Brompton Hospital, London, United Kingdom
- Division of Pulmonary, Critical Care and Sleep Medicine, Hastings Center for Pulmonary Research, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jose D. Herazo-Maya
- Division of Pulmonary, Critical Care, and Sleep Medicine, Tampa General Hospital, University of South Florida, Tampa, Florida
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan; and
| | - Fernando J. Martinez
- Internal Medicine, Weill Cornell Medical College, Cornell University, New York, New York
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, The University of Virginia, Charlottesville, Virginia
| |
Collapse
|
29
|
Turnover of type I and III collagen predicts progression of idiopathic pulmonary fibrosis. Respir Res 2021; 22:205. [PMID: 34261485 PMCID: PMC8281632 DOI: 10.1186/s12931-021-01801-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/11/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is characterized by the accumulation of fibrillar collagens in the alveolar space resulting in reduced pulmonary function and a high mortality rate. Biomarkers measuring the turnover of type I and III collagen could provide valuable information for prognosis and treatment decisions in IPF. METHODS Serological biomarkers reflecting the formation of type III collagen (PRO-C3) and degradation of type I (C1M) and III collagen (C3M) were evaluated in a real-world cohort of 178 newly diagnosed IPF patients. Blood samples and clinical data were collected at baseline, six, and 12 months. Baseline and longitudinal biomarker levels were related to disease progression of IPF (defined as ≥ 5% decline in forced vital capacity (FVC) and/or ≥ 10% decline in diffusing capacity for carbon monoxide (DLco) and/or all-cause mortality at 12 months). Furthermore, we analysed differences in percentage change of biomarker levels from baseline between patients receiving antifibrotic treatment or not. RESULTS Increased baseline levels of type I and III collagen turnover biomarkers were associated with a greater risk of disease progression within 12 months compared to patients with a low baseline type I and III collagen turnover. Patients with progressive disease had higher serum levels of C1M (P = 0.038) and PRO-C3 (P = 0.0022) compared to those with stable disease over one year. There were no differences in biomarker levels between patients receiving pirfenidone, nintedanib, or no antifibrotics. CONCLUSION Baseline levels of type I and III collagen turnover were associated with disease progression within 12 months in a real-world cohort of IPF patients. Longitudinal biomarker levels of type I and III collagen turnover were related to progressive disease. Moreover, antifibrotic therapy did not affect type I and III collagen turnover biomarkers in these patients. PRO-C3 and C1M may be potential biomarkers for a progressive disease behavior in IPF.
Collapse
|
30
|
Maher TM, Brown KK, Kreuter M, Devaraj A, Walsh SLF, Lancaster LH, Belloli EA, Padilla M, Behr J, Goeldner RG, Tetzlaff K, Schlenker-Herceg R, Flaherty KR. Effects of nintedanib by inclusion criteria for progression of interstitial lung disease. Eur Respir J 2021; 59:13993003.04587-2020. [PMID: 34210788 PMCID: PMC8812469 DOI: 10.1183/13993003.04587-2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/04/2021] [Indexed: 11/19/2022]
Abstract
Background The INBUILD trial investigated nintedanib versus placebo in patients with progressive fibrosing interstitial lung diseases (ILDs). We investigated the decline in forced vital capacity (FVC) in subgroups based on the inclusion criteria for ILD progression. Methods Subjects had a fibrosing ILD other than idiopathic pulmonary fibrosis and met the following criteria for ILD progression within the 24 months before screening despite management deemed appropriate in clinical practice: Group A, relative decline in FVC ≥10% predicted; Group B, relative decline in FVC ≥5–<10% predicted with worsened respiratory symptoms and/or increased extent of fibrosis on high-resolution computed tomography (HRCT); Group C, worsened respiratory symptoms and increased extent of fibrosis on HRCT only. Results In the placebo group, the rates of FVC decline over 52 weeks in Groups A, B and C, respectively, were −241.9, −133.1 and −115.3 mL per year in the overall population (p=0.0002 for subgroup-by-time interaction) and −288.9, −156.2 and −100.1 mL per year among subjects with a usual interstitial pneumonia (UIP)-like fibrotic pattern on HRCT (p=0.0005 for subgroup-by-time interaction). Nintedanib had a greater absolute effect on reducing the rate of FVC decline in Group A than in Group B or C. However, the relative effect of nintedanib versus placebo was consistent across the subgroups (p>0.05 for heterogeneity). Conclusions The inclusion criteria used in the INBUILD trial, based on FVC decline or worsening of symptoms and extent of fibrosis on HRCT, were effective at identifying patients with progressive fibrosing ILDs. Nintedanib reduced the rate of decline in FVC across the subgroups based on the inclusion criteria related to ILD progression. In the INBUILD trial in patients with fibrosing ILDs, the relative effect of nintedanib versus placebo on reducing the rate of FVC decline was consistent across subgroups based on the criteria regarding ILD progression that patients fulfilled on trial entryhttps://bit.ly/35jpOiE
Collapse
Affiliation(s)
- Toby M Maher
- National Heart and Lung Institute, Imperial College London, London, UK, National Institute for Health Research Clinical Research Facility, Royal Brompton Hospital, London, UK and Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Kevin K Brown
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Michael Kreuter
- Center for Interstitial and Rare Lung Diseases, Pneumology and Respiratory Care Medicine, Thoraxklinik, University of Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany
| | - Anand Devaraj
- Department of Radiology, Royal Brompton Hospital, London; National Heart and Lung Institute, Imperial College, London, UK
| | - Simon L F Walsh
- National Heart and Lung Institute, Imperial College, London, UK
| | | | - Elizabeth A Belloli
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Maria Padilla
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Juergen Behr
- Department of Medicine V, University Hospital, LMU Munich and Asklepios Klinik München-Gauting, Member of the German Centre for Lung Research, Germany
| | | | - Kay Tetzlaff
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany.,Department of Sports Medicine, University of Tübingen, Tübingen, Germany
| | | | - Kevin R Flaherty
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
31
|
Niwamoto T, Handa T, Murase Y, Nakatsuka Y, Tanizawa K, Taguchi Y, Tomioka H, Tomii K, Kita H, Uyama M, Tsuchiya M, Emura M, Kawamura T, Arai N, Arita M, Uno K, Yoshizawa A, Uozumi R, Yamaguchi I, Matsuda F, Chin K, Hirai T. Cutaneous T-cell-attracting chemokine as a novel biomarker for predicting prognosis of idiopathic pulmonary fibrosis: a prospective observational study. Respir Res 2021; 22:181. [PMID: 34158044 PMCID: PMC8218397 DOI: 10.1186/s12931-021-01779-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/13/2021] [Indexed: 12/31/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive fibrotic lung disease that leads to respiratory failure and death. Although there is a greater understanding of the etiology of this disease, accurately predicting the disease course in individual patients is still not possible. This study aimed to evaluate serum cytokines/chemokines as potential biomarkers that can predict outcomes in IPF patients. Methods A multi-institutional prospective two-stage discovery and validation design using two independent cohorts was adopted. For the discovery analysis, serum samples from 100 IPF patients and 32 healthy controls were examined using an unbiased, multiplex immunoassay of 48 cytokines/chemokines. The serum cytokine/chemokine values were compared between IPF patients and controls; the association between multiplex measurements and survival time was evaluated in IPF patients. In the validation analysis, the cytokines/chemokines identified in the discovery analysis were examined in serum samples from another 81 IPF patients to verify the ability of these cytokines/chemokines to predict survival. Immunohistochemical assessment of IPF-derived lung samples was also performed to determine where this novel biomarker is expressed. Results In the discovery cohort, 18 cytokines/chemokines were significantly elevated in sera from IPF patients compared with those from controls. Interleukin-1 receptor alpha (IL-1Rα), interleukin-8 (IL-8), macrophage inflammatory protein 1 alpha (MIP-1α), and cutaneous T-cell-attracting chemokine (CTACK) were associated with survival: IL-1Rα, hazard ratio (HR) = 1.04 per 10 units, 95% confidence interval (95% CI) 1.01–1.07; IL-8, HR = 1.04, 95% CI 1.01–1.08; MIP-1α, HR = 1.19, 95% CI 1.00–1.36; and CTACK, HR = 1.12 per 100 units, 95% CI 1.02–1.21. A replication analysis was performed only for CTACK because others were previously reported to be potential biomarkers of interstitial lung diseases. In the validation cohort, CTACK was associated with survival: HR = 1.14 per 100 units, 95% CI 1.01–1.28. Immunohistochemistry revealed the expression of CTACK and CC chemokine receptor 10 (a ligand of CTACK) in airway and type II alveolar epithelial cells of IPF patients but not in those of controls. Conclusions CTACK is a novel prognostic biomarker of IPF. Trial registration None (because of no healthcare intervention) Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01779-9.
Collapse
Affiliation(s)
- Takafumi Niwamoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tomohiro Handa
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Advanced Medicine for Respiratory Failure, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Yuko Murase
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshinari Nakatsuka
- Department of Respiratory Care and Sleep Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kiminobu Tanizawa
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshio Taguchi
- Department of Respiratory Medicine, Tenri Hospital, 200 Mishima-cho, Nara, 632-0015, Japan
| | - Hiromi Tomioka
- Department of Respiratory Medicine, Kobe City Medical Center West Hospital, 2-4 Ichiban-cho, Nagata-ku, Hyogo, 653-0013, Japan
| | - Keisuke Tomii
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojimaminami-machi, Chuou-ku, Hyogo, 650-0047, Japan
| | - Hideo Kita
- Department of Respiratory Medicine, Takatsuki Red Cross Hospital, 1-1-1 abuno, Osaka, 569-1045, Japan
| | - Michihiro Uyama
- Respiratory Disease Center, Kitano Hospital, Tazuke Kofukai Medical, Research Institute, 2-4-0 Ohgimachi, Kita-ku, Osaka, 530-8480, Japan
| | - Michiko Tsuchiya
- Department of Respiratory Medicine, Otowa Hospital, 2 Otowachinji-cho, Yamashina-ku, Kyoto, 607-8062, Japan
| | - Masahito Emura
- Department of Respiratory Medicine, Kyoto City Hospital, 1-2 Mibuhigasitakada-cho, nakagyo-ku, Kyoto, 604-8845, Japan
| | - Tetsuji Kawamura
- Department of Respiratory Medicine, Himeji Medical Center, 68 Hon-machi, Hyogo, 670-8520, Japan
| | - Naoki Arai
- National Hospital Organization Ibaraki Higashi National Hospital, Terunuma 825, Tokai, Ibaraki, 319-1113, Japan
| | - Machiko Arita
- Department of Respiratory Medicine, Ohara Healthcare Foundation, Kurashiki Central Hospital, 1-1-1 Miwa, Kurashiki, Okayama, 710-8602, Japan
| | - Kazuko Uno
- Louis Pasteur Center for Medical Research, 103-5 Tanakamonzen-cho, Sakyo-ku, Kyoto, 606-8225, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryuji Uozumi
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Izumi Yamaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kazuo Chin
- Department of Respiratory Care and Sleep Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
32
|
Sakamoto K, Furukawa T, Yamano Y, Kataoka K, Teramachi R, Walia A, Suzuki A, Inoue M, Nakahara Y, Ryu C, Hashimoto N, Kondoh Y. Serum mitochondrial DNA predicts the risk of acute exacerbation and progression of idiopathic pulmonary fibrosis. Eur Respir J 2021; 57:13993003.01346-2020. [PMID: 32855220 DOI: 10.1183/13993003.01346-2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/29/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Koji Sakamoto
- Dept of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan .,These authors contributed equally to this work
| | - Taiki Furukawa
- Dept of Medical IT Center, Nagoya University Hospital, Nagoya, Japan.,These authors contributed equally to this work
| | - Yasuhiko Yamano
- Dept of Respiratory and Allergy, Tosei General Hospital, Seto, Japan
| | - Kensuke Kataoka
- Dept of Respiratory and Allergy, Tosei General Hospital, Seto, Japan
| | - Ryo Teramachi
- Dept of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Anjali Walia
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Atsushi Suzuki
- Dept of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahide Inoue
- Dept of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshio Nakahara
- Dept of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Changwan Ryu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Naozumi Hashimoto
- Dept of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kondoh
- Dept of Respiratory and Allergy, Tosei General Hospital, Seto, Japan
| |
Collapse
|
33
|
Adegunsoye A, Alqalyoobi S, Linderholm A, Bowman WS, Lee CT, Pugashetti JV, Sarma N, Ma SF, Haczku A, Sperling A, Strek ME, Noth I, Oldham JM. Circulating Plasma Biomarkers of Survival in Antifibrotic-Treated Patients With Idiopathic Pulmonary Fibrosis. Chest 2020; 158:1526-1534. [PMID: 32450241 PMCID: PMC7545483 DOI: 10.1016/j.chest.2020.04.066] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/30/2020] [Accepted: 04/08/2020] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND A number of circulating plasma biomarkers have been shown to predict survival in patients with idiopathic pulmonary fibrosis (IPF), but most were identified before the use of antifibrotic (AF) therapy in this population. Because pirfenidone and nintedanib have been shown to slow IPF progression and may prolong survival, the role of such biomarkers in AF-treated patients is unclear. RESEARCH QUESTION To determine whether plasma concentration of cancer antigen 125 (CA-125), C-X-C motif chemokine 13 (CXCL13), matrix metalloproteinase 7 (MMP7), surfactant protein D (SP-D), chitinase-3-like protein-1 (YKL-40), vascular cell adhesion protein-1 (VCAM-1), and osteopontin (OPN) is associated with differential transplant-free survival (TFS) in AF-exposed and nonexposed patients with IPF. STUDY DESIGN AND METHODS A pooled, multicenter, propensity-matched analysis of IPF patients with and without AF exposure was performed. Optimal thresholds for biomarker dichotomization were identified in each group using iterative Cox regression. Longitudinal biomarker change was assessed in a subset of patients using linear mixed regression modeling. A clinical-molecular signature of IPF TFS was then derived and validated in an independent IPF cohort. RESULTS Three hundred twenty-five patients were assessed, of which 68 AF-exposed and 172 nonexposed patients were included after propensity matching. CA-125, CXCL13, MMP7, YKL-40, and OPN predicted differential TFS in AF-exposed patients but at higher thresholds than in AF-nonexposed individuals. Plasma biomarker level generally increased over time in nonexposed patients but remained unchanged in AF-exposed patients. A clinical-molecular signature predicted decreased TFS in AF-exposed patients (hazard ratio [HR], 5.91; 95% CI, 2.25-15.5; P < .001) and maintained this association in an independent AF-exposed cohort (HR, 3.97; 95% CI, 1.62-9.72; P = .003). INTERPRETATION Most plasma biomarkers assessed predicted differential TFS in AF-exposed patients with IPF, but at higher thresholds than in nonexposed patients. A clinical-molecular signature of IPF TFS may provide a reliable predictor of outcome risk in AF-treated patients but requires additional research for optimization and validation.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL
| | - Shehabaldin Alqalyoobi
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, East Carolina University, Greenville, NC
| | - Angela Linderholm
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Willis S Bowman
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Cathryn T Lee
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL
| | - Janelle Vu Pugashetti
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Nandini Sarma
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Shwu-Fan Ma
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlotteville, VA
| | - Angela Haczku
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Anne Sperling
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL
| | - Mary E Strek
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, IL
| | - Imre Noth
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlotteville, VA
| | - Justin M Oldham
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA.
| |
Collapse
|
34
|
Gupta S, Batt J, Bourbeau J, Chapman KR, Gershon A, Granton J, Hambly N, Hernandez P, Kolb M, Mehta S, Mielniczuk L, Provencher S, Stephenson AL, Swiston J, Tullis DE, Vozoris NT, Wald J, Weatherald J, Bhutani M. Triaging Access to Critical Care Resources in Patients With Chronic Respiratory Diseases in the Event of a Major COVID-19 Surge: Key Highlights From the Canadian Thoracic Society (CTS) Position Statement. Chest 2020; 158:2270-2274. [PMID: 32693101 PMCID: PMC7368654 DOI: 10.1016/j.chest.2020.07.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 01/23/2023] Open
Affiliation(s)
- Samir Gupta
- St Michael's Hospital Unity Health Toronto, Li Ka Shing Knowledge Institute, Department of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Jane Batt
- Keenan Research Center for Biomedical Science, St Michael's Hospital Unity Health Toronto, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jean Bourbeau
- Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Kenneth R Chapman
- Toronto General Hospital Research Institute, University of Toronto, Toronto, ON, Canada
| | - Andrea Gershon
- Sunnybrook Health Sciences Centre, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - John Granton
- Division of Respirology, Department of Medicine, University Health Network, Sinai Health System, University of Toronto, Toronto, ON, Canada
| | - Nathan Hambly
- Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Paul Hernandez
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Martin Kolb
- Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sanjay Mehta
- Division of Respirology, Department of Medicine, London Health Sciences Centre, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Lisa Mielniczuk
- Department of Medicine, University of Ottawa, Division of Cardiology, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Institut universitaire de cardiologie et de pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Anne L Stephenson
- Adult Cystic Fibrosis Program, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - John Swiston
- Division of Respirology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - D Elizabeth Tullis
- St Michael's Hospital Unity Health Toronto, Li Ka Shing Knowledge Institute, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nicholas T Vozoris
- St Michael's Hospital Unity Health Toronto, Li Ka Shing Knowledge Institute, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Joshua Wald
- Department of Medicine, Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jason Weatherald
- Department of Medicine, Division of Respirology, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Mohit Bhutani
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
35
|
Sarkar P, Avram C, Chaudhuri N. The extended utility of antifibrotic therapy in progressive fibrosing interstitial lung disease. Expert Rev Respir Med 2020; 14:1001-1008. [PMID: 32567402 DOI: 10.1080/17476348.2020.1784730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION The approval of two antifibrotic treatment agents for delaying disease progression in idiopathic pulmonary fibrosis (IPF), has prompted researchers to look at expanding the role of antifibrotic therapy to other fibrosing interstitial lung disease (ILD). Similarities in the pathological mechanisms that lead to the development of IPF have been implicated in other progressive fibrosing ILD (PF-ILD) such as chronic hypersensitivity pneumonitis, connective tissues disease associated ILD, sarcoidosis, occupational ILD and idiopathic non-specific interstitial pneumonia (iNSIP). This has prompted the rationale to use antifibrotic therapy to target similar molecular pathways in these diseases. AREAS COVERED This review will summarise the available evidence from randomised controlled trials that have evaluated the use of antifibrotic therapy in PF-ILD outside the realm of IPF. EXPERT OPINION There is promising data for antifibrotic therapy as a therapeutic option for non IPF PF-ILD. The new therapy option does provide some challenges that need to be addressed such as timing of initiation of therapy, clarifying the strategy for overlap or combination with existing immunosuppressive therapies and potential drug interactions. There is an unmet need to determine accurate predictors of disease progression to allow early intervention for the preservation of lung function and mortality reduction.
Collapse
Affiliation(s)
- Paroma Sarkar
- Department of Thoracic Medicine, The Royal Adelaide Hospital , Adelaide, Australia
| | - Cristina Avram
- Department of Respiratory Medicine, Manchester University NHS Foundation Trust , Manchester, UK
| | - Nazia Chaudhuri
- Department of Respiratory Medicine, Manchester University NHS Foundation Trust , Manchester, UK
| |
Collapse
|
36
|
Kärkkäinen M, Kettunen HP, Nurmi H, Selander T, Purokivi M, Kaarteenaho R. Comparison of disease progression subgroups in idiopathic pulmonary fibrosis. BMC Pulm Med 2019; 19:228. [PMID: 31783748 PMCID: PMC6883511 DOI: 10.1186/s12890-019-0996-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/14/2019] [Indexed: 01/08/2023] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial pneumonia with an unpredictable course. The aims of this study were to retrospectively re-evaluate a cohort of patients with IPF according to the 2011 international IPF guidelines and 1) to characterize the subgroups of patients when classified according to their observed survival times and 2) to evaluate whether Composite Physiologic Index (CPI), Gender-Age-Physiology (GAP) Index or clinical variables could predict mortality. Methods Retrospective data was collected and patients were classified into subgroups according to their observed lifespans. Differences in clinical variables, CPI and GAP stages as well as in comorbidities were investigated between the subgroups. Predictors of mortality were identified by COX proportional hazard analyses. Results A total of 132 patients were included in this study. The disease course was rapid (≤ 2 years) in 30.0%, moderate (2–5 years) in 28.0% and slow (≥ 5 years) in 29.0% of the patients. Pulmonary function tests (PFT) and CPI at baseline differentiated significantly between the rapid disease course group and those patients with longer survival times. However, the predictive accuracy of the investigated clinical variables was mainly less than 0.80. The proportions of patients with comorbidities did not differ between the subgroups, but more patients with a rapid disease course were diagnosed with heart failure after the diagnosis of IPF. Most patients with a rapid disease course were categorized in GAP stages I and II, but all patients in GAP stage III had a rapid disease course. The best predictive multivariable model included age, gender and CPI. GAP staging had slightly better accuracy (0.67) than CPI (0.64) in predicting 2-year mortality. Conclusions Although the patients with a rapid disease course could be differentiated at baseline in terms of PFT and CPI, the predictive accuracy of any single clinical variable as well as CPI and GAP remained low. GAP staging was unable to identify the majority of patients with a rapid disease progression. It is challenging to predict disease progression and mortality in IPF even with risk prediction models.
Collapse
Affiliation(s)
- Miia Kärkkäinen
- Division of Respiratory Medicine, Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland. .,Kuopio City Home Care, Rehabilitation and Medical Services for Elderly, Tulliportinkatu 37E, 70100, Kuopio, Finland. .,Kuopio University Hospital, P.O. Box 100, Puijonlaaksontie 2, 70210, Kuopio, Finland.
| | - Hannu-Pekka Kettunen
- Department of Clinical Radiology, Kuopio University Hospital, P.O. Box 100, 70029 KYS, Kuopio, Finland
| | - Hanna Nurmi
- Division of Respiratory Medicine, Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.,Center of Medicine and Clinical Research, Division of Respiratory Medicine, Kuopio University Hospital, P.O. Box 100, 70029 KYS, Kuopio, Finland
| | - Tuomas Selander
- Science Services Center, Kuopio University Hospital, P.O. Box 100, 70029 KYS, Kuopio, Finland
| | - Minna Purokivi
- Center of Medicine and Clinical Research, Division of Respiratory Medicine, Kuopio University Hospital, P.O. Box 100, 70029 KYS, Kuopio, Finland
| | - Riitta Kaarteenaho
- Respiratory Medicine, Research Unit of Internal Medicine, University of Oulu and Medical Research Center Oulu, Oulu University Hospital, P.O. Box 20, 90029, Oulu, Finland
| |
Collapse
|
37
|
Santermans E, Ford P, Kreuter M, Verbruggen N, Meyvisch P, Wuyts WA, Brown KK, Lederer DJ, Byrne AJ, Molyneaux PL, Sivananthan A, Moor CC, Maher TM, Wijsenbeek M. Modelling Forced Vital Capacity in Idiopathic Pulmonary Fibrosis: Optimising Trial Design. Adv Ther 2019; 36:3059-3070. [PMID: 31565781 PMCID: PMC6822798 DOI: 10.1007/s12325-019-01093-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Indexed: 11/05/2022]
Abstract
Introduction Forced vital capacity is the only registrational endpoint in idiopathic pulmonary fibrosis clinical trials. As most new treatments will be administered on top of standard of care, estimating treatment response will become more challenging. We developed a simulation model to quantify variability associated with forced vital capacity decline. Methods The model is based on publicly available clinical trial summary and home spirometry data. A single, illustrative trial setting is reported. Model assumptions are 400 subjects randomised 1:1 to investigational drug or placebo over 52 weeks, 50% of each group receiving standard of care (all-comer population), and a 90-mL treatment difference in annual forced vital capacity decline. Longitudinal profiles were simulated and the impact of varying clinical scenarios evaluated. Results Power to detect a significant treatment difference was 87–97%, depending on the analysis method. Repeated measures analysis generally outperformed analysis of covariance and mixed linear models, particularly with missing data (as simulated data were non-linear). A 15% yearly random dropout rate led to 0.6–5% power loss. Forced vital capacity decline-related dropout introduced greater power loss (up to 12%), as did subjects starting/stopping standard of care or investigational drug. Power was substantially lower for a 26-week trial due to the smaller assumed treatment effect at week 26 (sample size would need doubling to reach a power similar to that of a 52-week trial). Conclusions Our model quantifies forced vital capacity decline and associated variability, with all the caveats of background therapy, permitting robust power calculations to inform future idiopathic pulmonary fibrosis clinical trial design. Funding Galapagos NV (Mechelen, Belgium). Electronic supplementary material The online version of this article (10.1007/s12325-019-01093-3) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Sullivan DI, Kass DJ. Signals and signposts: Biomarkers in IPF and PAH at the crossroads of clinical relevance. Respirology 2019; 24:1044-1045. [PMID: 31486582 PMCID: PMC8491577 DOI: 10.1111/resp.13694] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 01/10/2023]
Abstract
See related Article and Article
Collapse
Affiliation(s)
- Daniel I Sullivan
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel J Kass
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
39
|
Raghu G, Ley B, Brown KK, Cottin V, Gibson KF, Kaner RJ, Lederer DJ, Noble PW, Song JW, Wells AU, Whelan TP, Lynch DA, Humphries SM, Moreau E, Goodman K, Patterson SD, Smith V, Gong Q, Sundy JS, O'Riordan TG, Martinez FJ. Risk factors for disease progression in idiopathic pulmonary fibrosis. Thorax 2019; 75:78-80. [PMID: 31611341 DOI: 10.1136/thoraxjnl-2019-213620] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 11/04/2022]
Abstract
In this retrospective study of a randomised trial of simtuzumab in idiopathic pulmonary fibrosis (IPF), prodromal decline in forced vital capacity (FVC) was significantly associated with increased risk of mortality, respiratory and all-cause hospitalisations, and categorical disease progression. Predictive modelling of progression-free survival event risk was used to assess the effect of population enrichment for patients at risk of rapid progression of IPF; C-index values were 0.64 (death), 0.69 (disease progression), and 0.72 (adjudicated respiratory hospitalisation) and 0.76 (all-cause hospitalisation). Predictive modelling may be a useful tool for improving efficiency of clinical trials with categorical end points.
Collapse
Affiliation(s)
- Ganesh Raghu
- Center for Interstitial Lung Diseases, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Brett Ley
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, California, USA
| | - Kevin K Brown
- Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, Colorado, USA
| | - Vincent Cottin
- Center for Rare Pulmonary Diseases, Hospices Civils de Lyon, University of Lyon, UMR754, Lyon, France
| | - Kevin F Gibson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert J Kaner
- Department of Clinical Medicine and Genetic Medicine, Weill Cornell Medicine, New York, New York, USA
| | - David J Lederer
- Division of Pulmonary, Allergy, and Critical Care, Columbia University Medical Center, New York, New York, USA
| | - Paul W Noble
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Jin Woo Song
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Athol U Wells
- Department of Medicine, National Heart & Lung Institute, Royal Brompton Hospital, Imperial College, London, UK
| | - Timothy P Whelan
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - David A Lynch
- Department of Radiology, National Jewish Health, Denver, Colorado, USA
| | | | | | - Krista Goodman
- Clinical Research, Gilead Sciences, Inc, Seattle, Washington, USA
| | | | - Victoria Smith
- Clinical Research, Gilead Sciences, Inc, Seattle, Washington, USA
| | - Qi Gong
- Biostatistics, Gilead Sciences, Inc, Foster City, California, USA
| | - John S Sundy
- Clinical Research, Gilead Sciences, Inc, Seattle, Washington, USA
| | | | | |
Collapse
|
40
|
Abstract
Lung transplantation is a valuable therapeutic option for many patients with severe lung disease who have exhausted other medical or surgical therapies. However, since lungs are not a manufacturable organ like artificial heart valves or left ventricular assist devices, and since they are a limited resource compared to number of patients requiring the organs, the Department of Health and Human Services set the Final Rule of organ allocation in 1998. This led to development and implementation of Lung Allocation Score (LAS) in 2005. The score broadly divides lung diseases into 4 diagnostic criteria with a coefficient factor given to each category. The score is based on the prognostic factors of each patient to determine the risk of mortality without a transplant combined with the probability of patient survival post-transplant. Most of the guidelines for "Indications for referral and listing in lung transplant" is based on consensus opinion as there is limited amount of robust data and trials about this topic. The International Society for Heart and Lung Transplant (ISHLT) has published three editions for candidate selection and listing. In this article, we have attempted to highlight the guidelines and incorporated other disease specific prognostic factors that are not captured in the LAS. Ultimately, there are other factors like geographic location, height, blood group, preformed antibodies, transplant center experience, past wait times and transplant rate, availability of organs, etc., which also play a role especially when considering listing a patient for lung transplant. We also highlighted a representative disease in each category and most criteria for that disease will apply to other diseases in that category. Finally, this article does not delve into the history and reasoning behind each guideline but is meant to provide a general overview of indications and contraindications applicable in the field of adult lung transplantation.
Collapse
Affiliation(s)
- Omar Shweish
- Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Goutham Dronavalli
- Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
41
|
Richeldi L, Crestani B, Azuma A, Kolb M, Selman M, Stansen W, Quaresma M, Stowasser S, Cottin V. Outcomes following decline in forced vital capacity in patients with idiopathic pulmonary fibrosis: Results from the INPULSIS and INPULSIS-ON trials of nintedanib. Respir Med 2019; 156:20-25. [DOI: 10.1016/j.rmed.2019.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/12/2019] [Accepted: 08/05/2019] [Indexed: 10/26/2022]
|
42
|
Schuster Bruce C, Brhlikova P, Heath J, McGettigan P. The use of validated and nonvalidated surrogate endpoints in two European Medicines Agency expedited approval pathways: A cross-sectional study of products authorised 2011-2018. PLoS Med 2019; 16:e1002873. [PMID: 31504034 PMCID: PMC6736244 DOI: 10.1371/journal.pmed.1002873] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/07/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND In situations of unmet medical need or in the interests of public health, expedited approval pathways, including conditional marketing authorisation (CMA) and accelerated assessment (AA), speed up European Medicines Agency (EMA) marketing authorisation recommendations for medicinal products. CMAs are based on incomplete benefit-risk assessment data and authorisation remains conditional until regulator-imposed confirmatory postmarketing measures are fulfilled. For products undergoing AA, complete safety and efficacy data should be available, and postauthorisation measures may include only standard requirements of risk management and pharmacovigilance plans. In the pivotal trials supporting products assessed by expedited pathways, surrogate endpoints reduce drug development time compared with waiting for the intended clinical outcomes. Whether surrogate endpoints supporting products authorised through CMA and AA pathways reliably predict clinical benefits of therapy has not been studied systematically. Our objectives were to determine the extent to which surrogate endpoints are used and to assess whether their validity had been confirmed according to published hierarchies. METHODS AND FINDINGS We used European Public Assessment Reports (EPARs) to identify the primary endpoints in the pivotal trials supporting products authorised through CMA or AA pathways during January 1, 2011 to December 31, 2018. We excluded products that were vaccines, topical, reversal, or bleeding prophylactic agents or withdrawn within the study time frame. Where pivotal trials reported surrogate endpoints, we conducted PubMed searches for evidence of validity for predicting clinical outcomes. We used 2 published hierarchies to assess validity level. Surrogates with randomised controlled trials supporting the surrogate-clinical outcome relationship were rated as 'validated'. Fifty-one products met the inclusion criteria; 26 underwent CMAs, and 25 underwent AAs. Overall, 26 products were for oncology indications, 10 for infections, 8 for genetic disorders, and 7 for other systems disorders. Five products (10%), all AAs, were authorised based on pivotal trials reporting clinical outcomes, and 46 (90%) were authorised based on surrogate endpoints. No studies were identified that validated the surrogate endpoints. Among a total of 49 products with surrogate endpoints reported, most were rated according to the published hierarchies as being 'reasonably likely' (n = 30; 61%) or of having 'biological plausibility' (n = 46; 94%) to predict clinical outcomes. EPARs did not consistently explain the nature of the pivotal trial endpoints supporting authorisations, whether surrogate endpoints were validated or not, or describe the endpoints to be reported in the confirmatory postmarketing studies. Our study has limitations: we may have overlooked relevant validation studies; the findings apply to 2 expedited pathways and may not be generalisable to products authorised through the standard assessment pathway. CONCLUSIONS The pivotal trial evidence supporting marketing authorisations for products granted CMA or AA was based dominantly on nonvalidated surrogate endpoints. EPARs and summary product characteristic documents, including patient information leaflets, need to state consistently the nature and limitations of endpoints in pivotal trials supporting expedited authorisations so that prescribers and patients appreciate shortcomings in the evidence about actual clinical benefit. For products supported by nonvalidated surrogate endpoints, postauthorisation measures to confirm clinical benefit need to be imposed by the regulator on the marketing authorisation holders.
Collapse
Affiliation(s)
- Catherine Schuster Bruce
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Petra Brhlikova
- The Institute of Health and Society, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Joseph Heath
- Greater Manchester Mental Health NHS Foundation Trust, Prestwich, Manchester, United Kingdom
| | - Patricia McGettigan
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| |
Collapse
|
43
|
Maher TM, Stowasser S, Nishioka Y, White ES, Cottin V, Noth I, Selman M, Rohr KB, Michael A, Ittrich C, Diefenbach C, Jenkins RG. Biomarkers of extracellular matrix turnover in patients with idiopathic pulmonary fibrosis given nintedanib (INMARK study): a randomised, placebo-controlled study. THE LANCET RESPIRATORY MEDICINE 2019; 7:771-779. [PMID: 31326319 DOI: 10.1016/s2213-2600(19)30255-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND A hallmark of idiopathic pulmonary fibrosis is the excess accumulation of extracellular matrix in the lungs. Degradation of extracellular matrix generates free-circulating protein fragments called neoepitopes. The aim of the INMARK trial was to investigate changes in neoepitopes as predictors of disease progression in patients with idiopathic pulmonary fibrosis and the effect of nintedanib on these biomarkers. METHODS In this randomised, double-blind, placebo-controlled trial, patients with a diagnosis of idiopathic pulmonary fibrosis within the past 3 years and forced vital capacity (FVC) of 80% predicted or higher were eligible to participate. Patients were recruited from hospitals, private practices, clinical research units, and academic medical centres. Patients were randomly assigned (1:2) with the use of a pseudo-random number generator to receive oral nintedanib 150 mg twice a day or placebo for 12 weeks in a double-blind fashion, followed by open-label nintedanib for 40 weeks. The primary endpoint was the rate of change in C-reactive protein (CRP) degraded by matrix metalloproteinases 1 and 8 (CRPM) from baseline to week 12 in the intention-to-treat population. The trial has been completed and is registered with ClinicalTrials.gov, number NCT02788474, and with the European Clinical Trials Database, number 2015-003148-38. FINDINGS Between June 27, 2016, and May 15, 2017, 347 patients were randomly assigned to the nintedanib group (n=116) or to the placebo group (n=231). One patient from the placebo group was not treated owing to a randomisation error. At baseline, mean FVC was 97·5% (SD 13·5) predicted. In the double-blind period, 116 patients received nintedanib and 230 patients received placebo. The rate of change in CRPM from baseline to week 12 was -2·57 × 10-3 ng/mL/month in the nintedanib group and -1·90 × 10-3 ng/mL/month in the placebo group (between-group difference -0·66 × 10-3 ng/mL/month [95% CI -6·21 × 10-3 to 4·88 × 10-3]; p=0·8146). The adjusted rate of change in FVC over 12 weeks was 5·9 mL in the nintedanib group and -70·2 mL in the placebo group (difference 76·1 mL/12 weeks [31·7 to 120·4]). In patients who received placebo for 12 weeks followed by open-label nintedanib, rising concentrations of CRPM over 12 weeks were associated with disease progression (absolute decline in FVC ≥10% predicted or death) over 52 weeks. In the double-blind period, serious adverse events were reported in eight (7%) patients given nintedanib and 18 (8%) patients given placebo. Grade 3 diarrhoea was reported in two (2%) patients in the nintedanib group and two (1%) patients in the placebo group. No patients had grade 4 diarrhoea. INTERPRETATION In patients with idiopathic pulmonary fibrosis and preserved lung function, treatment with nintedanib versus placebo for 12 weeks did not affect the rate of change in CRPM but was associated with a reduced rate of decline in FVC. These results suggest that change in CRPM is not a marker of response to nintedanib in patients with idiopathic pulmonary fibrosis. FUNDING Boehringer Ingelheim.
Collapse
Affiliation(s)
- Toby M Maher
- National Heart and Lung Institute, Imperial College London, London, UK; National Institute for Health Research Clinical Research Facility, Royal Brompton Hospital, London, UK.
| | - Susanne Stowasser
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Rhein, Germany
| | - Yasuhiko Nishioka
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Eric S White
- University of Michigan, Division of Pulmonary and Critical Care Medicine, Ann Arbor, MI, USA
| | - Vincent Cottin
- National Reference Centre for Rare Pulmonary Diseases, Louis Pradel Hospital, Hospices Civils de Lyon, Claude Bernard University Lyon 1, Lyon, France
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VI, USA
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Klaus B Rohr
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Rhein, Germany
| | | | - Carina Ittrich
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | | | - R Gisli Jenkins
- National Institute for Health Research Respiratory Biomedical Research Centre, City Campus, Nottingham University Hospital, Nottingham, UK
| | | |
Collapse
|
44
|
O’Donnell DE, Milne KM, Vincent SG, Neder JA. Unraveling the Causes of Unexplained Dyspnea. Clin Chest Med 2019; 40:471-499. [DOI: 10.1016/j.ccm.2019.02.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
45
|
Cottin V, Annesi-Maesano I, Günther A, Galvin L, Kreuter M, Powell P, Prasse A, Reynolds G, Richeldi L, Spagnolo P, Valenzuela C, Wijsenbeek M, Wuyts WA, Crestani B. The Ariane-IPF ERS Clinical Research Collaboration: seeking collaboration through launch of a federation of European registries on idiopathic pulmonary fibrosis. Eur Respir J 2019; 53:53/5/1900539. [DOI: 10.1183/13993003.00539-2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 04/24/2019] [Indexed: 12/31/2022]
|
46
|
|
47
|
Ryerson CJ, Kolb M, Richeldi L, Lee J, Wachtlin D, Stowasser S, Poletti V. Effects of nintedanib in patients with idiopathic pulmonary fibrosis by GAP stage. ERJ Open Res 2019; 5:00127-2018. [PMID: 31044139 PMCID: PMC6487272 DOI: 10.1183/23120541.00127-2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 02/08/2019] [Indexed: 11/26/2022] Open
Abstract
We conducted a post hoc analysis to assess the potential impact of GAP (gender, age, physiology) stage on the treatment effect of nintedanib in patients with idiopathic pulmonary fibrosis. Outcomes were compared in patients at GAP stage I versus II/III at baseline in the INPULSIS® trials. At baseline, 500 patients were at GAP stage I (nintedanib 304, placebo 196), 489 were at GAP stage II (nintedanib 296, placebo 193) and 71 were at GAP stage III (nintedanib 38, placebo 33). In nintedanib-treated patients, the annual rate of decline in forced vital capacity (FVC) was similar in patients at GAP stage I and GAP stage II/III at baseline (−110.1 and −116.6 mL·year−1, respectively), and in both subgroups was lower than in placebo-treated patients (−218.5 and −227.6 mL·year−1, respectively) (treatment-by-time-by-subgroup interaction p=0.92). In the nintedanib group, the number of deaths was 43.8% of those predicted based on GAP stage (35 versus 79.9). In the placebo group, the number of deaths was 59.8% of those predicted based on GAP stage (33 versus 55.2). In conclusion, data from the INPULSIS® trials suggest that nintedanib has a similar beneficial effect on the rate of FVC decline in patients at GAP stage I versus II/III at baseline. Nintedanib provides a similar benefit versus placebo on the rate of decline in forced vital capacity in patients with idiopathic pulmonary fibrosis irrespective of GAP stage at baselinehttp://ow.ly/HfJ730nNkRT
Collapse
Affiliation(s)
- Christopher J Ryerson
- Dept of Medicine and Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| | - Martin Kolb
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Luca Richeldi
- Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Joyce Lee
- Dept of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Daniel Wachtlin
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Susanne Stowasser
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | | |
Collapse
|
48
|
High-Resolution Computed Tomography (HRCT) Reflects Disease Progression in Patients with Idiopathic Pulmonary Fibrosis (IPF): Relationship with Lung Pathology. J Clin Med 2019; 8:jcm8030399. [PMID: 30909411 PMCID: PMC6463252 DOI: 10.3390/jcm8030399] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 01/16/2023] Open
Abstract
High-Resolution Computed Tomography (HRCT) plays a central role in diagnosing Idiopathic Pulmonary Fibrosis (IPF) while its role in monitoring disease progression is not clearly defined. Given the variable clinical course of the disease, we evaluated whether HRCT abnormalities predict disease behavior and correlate with functional decline in untreated IPF patients. Forty-nine patients (with HRCT1) were functionally categorized as rapid or slow progressors. Twenty-one had a second HRCT2. Thirteen patients underwent lung transplantation and pathology was quantified. HRCT Alveolar (AS) and Interstitial Scores (IS) were assessed and correlated with Forced Vital Capacity (FVC) decline between HRCT1 and HRCT2. At baseline, AS was greater in rapids than in slows, while IS was similar in the two groups. In the 21 subjects with HRCT2, IS increased over time in both slows and rapids, while AS increased only in rapids. The IS change from HRCT1 to HRCT2 normalized per month correlated with FVC decline/month in the whole population, but the change in AS did not. In the 13 patients with pathology, the number of total lymphocytes was higher in rapids than in slows and correlated with AS. Quantitative estimation of HRCTs AS and IS reflects the distinct clinical and pathological behavior of slow and rapid decliners. Furthermore, AS, which reflects the immune/inflammatory infiltrate in lung tissue, could be a useful tool to differentiate rapid from slow progressors at presentation.
Collapse
|
49
|
|
50
|
|