151
|
Cameli P, Carleo A, Bergantini L, Landi C, Prasse A, Bargagli E. Oxidant/Antioxidant Disequilibrium in Idiopathic Pulmonary Fibrosis Pathogenesis. Inflammation 2019; 43:1-7. [DOI: 10.1007/s10753-019-01059-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
152
|
Abstract
Objective: Provide information for pharmacists on idiopathic pulmonary fibrosis (IPF) and its treatment. Study Selection and Data Extraction: All articles with data from randomized controlled trials of nintedanib or pirfenidone were reviewed. Data Synthesis: IPF is a progressive and ultimately fatal interstitial lung disease characterized by decline in lung function and worsening dyspnea. It is uncommon and mainly occurs in individuals aged >60 years, particularly men with a history of smoking. Nintedanib and pirfenidone were approved in the United States for the treatment of IPF in 2014 and received conditional recommendations in the 2015 American Thoracic Society/European Respiratory Society/Japanese Respiratory Society/Latin American Thoracic Association treatment guidelines. These drugs slow the progression of IPF by reducing the rate of decline in lung function. Their adverse event profile is characterized mainly by gastrointestinal events, which can be managed through dose adjustment and symptom management. Management of IPF should also include smoking cessation, vaccinations, and supportive care such as patient education, pulmonary rehabilitation, and the use of supplemental oxygen as well as optimizing the management of comorbidities. Relevance to Patient Care and Clinical Practice: This review provides clinical pharmacists with information on the course of IPF, what can be expected of current treatments, and how to help patients manage their drug therapy. Conclusions: IPF is a progressive disease, but treatments are available that can slow the progression of the disease. Clinical pharmacists can play an important role in the care of patients with IPF through patient education, monitoring medication compliance and safety, ensuring drugs for comorbidities are optimized, and preventive strategies such as immunizations.
Collapse
Affiliation(s)
- Roy Pleasants
- The University of North Carolina at Chapel Hill, NC, USA.,Durham Veterans Administration Medical Center, Durham, NC, USA
| | | |
Collapse
|
153
|
Mathai SK, Schwartz DA. Translational research in pulmonary fibrosis. Transl Res 2019; 209:1-13. [PMID: 30768925 PMCID: PMC9977489 DOI: 10.1016/j.trsl.2019.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 11/26/2022]
Abstract
Pulmonary fibrosis refers to the development of diffuse parenchymal abnormalities in the lung that cause dyspnea, cough, hypoxemia, and impair gas exchange, ultimately leading to respiratory failure. Though pulmonary fibrosis can be caused by a variety of underlying etiologies, ranging from genetic defects to autoimmune diseases to environmental exposures, once fibrosis develops it is irreversible and most often progressive, such that fibrosis of the lung is one of the leading indications for lung transplantation. This review aims to provide a concise summary of the recent advances in our understanding of the genetics and genomics of pulmonary fibrosis, idiopathic pulmonary fibrosis in particular, and how these recent discoveries may be changing the clinical approach to diagnosing and treating patients with fibrotic interstitial lung disease.
Collapse
Affiliation(s)
- Susan K Mathai
- Interstitial Lung Disease Program, Center for Advanced Heart & Lung Disease, Department of Medicine, Baylor University Medical Center at Dallas, Dallas, Texas; Department of Internal Medicine, Texas A&M University College of Medicine.
| | - David A Schwartz
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
154
|
Pang LJ, Liu JP, Lv XD. Comparative effectiveness of 3 Traditional Chinese Medicine treatment methods for idiopathic pulmonary fibrosis: A systematic review and network meta-analysis protocol. Medicine (Baltimore) 2019; 98:e16325. [PMID: 31348231 PMCID: PMC6709242 DOI: 10.1097/md.0000000000016325] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION The morbidity of idiopathic pulmonary fibrosis (IPF) was found in an increasing trend, progressive worsening of symptoms and deterioration in lung function tend to trigger off a lower quality of life (QoL). Only pirfenidone and nintedanib have been recommended in the guidelines, which can modify the disease process. However, no evidence was verified to significantly alleviate the main clinical manifestations of IPF. At present, Chinese herbal formula (CHF) is widely prescribed as an adjunct to western medicine to treat the disease, and have shown promising benefits on clinical symptoms and QoL. There are mainly 3 Traditional Chinese Medicine (TCM) treatment methods guiding the composition of CHFs, which are devoting to comfort the common symptoms of IPF. Nevertheless, the paucity of direct comparative evidence of them posed a challenge for clinicians to determine the relative merits options. Therefore, we formulate this protocol, which is described for a systematic review to investigate relative advantages among different TCM treatment method and provide more reliable evidence for clinical decision-making. METHODS AND ANALYSIS A systematic literature search will be employed in 10 electronic databases. Inclusion criteria are randomized control trials of CHFs composed based on the 3 TCM treatment methods, which act as an adjuvant treatment with routine drugs, compared with routine drugs alone. The primary outcomes we focus on include St George's Hospital Respiratory Questionnaire (SGRQ) scores, TCM symptom (dyspnea, cough) scores. The research screening, data extraction, and methodological quality assessment will be conducted by 2 individuals separately, and dispute will be adjudicated by a third senior reviewer. We will employ network meta-analysis (NMA) in a Bayesian framework with vague priors and the surface under the cumulative ranking curve (SUCRA) to obtain the comprehensive rank for the 3 TCM treatment methods. RESULTS This systematic review will provide an evidence of CHFs composed under the guidance by 3 TCM treatment methods with routine drugs, compared with routine drugs alone for IPF, and will submit to a peer-reviewed journal for publication. CONCLUSION The conclusion of this systematic review will provide evidence for relative advantages among the 3 TCM treatment methods.
Collapse
Affiliation(s)
- Li-Jian Pang
- Beijing University of Chinese Medicine, Beijing
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang
| | - Jian-Ping Liu
- Beijing University of Chinese Medicine, Beijing
- Center for Evidence Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Dong Lv
- Liaoning University of Traditional Chinese Medicine
| |
Collapse
|
155
|
Cazzola M, Calzetta L, Page C, Rogliani P, Matera MG. Thiol-Based Drugs in Pulmonary Medicine: Much More than Mucolytics. Trends Pharmacol Sci 2019; 40:452-463. [DOI: 10.1016/j.tips.2019.04.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 04/03/2019] [Accepted: 04/30/2019] [Indexed: 02/02/2023]
|
156
|
Troy LK, Grainge C, Corte T, Williamson JP, Vallely MP, Cooper W, Mahar AM, Lai S, Mulyadi E, Torzillo PJ, Salamonsen M, Don G, Myers J, Raghu G, Lau EMT. Cryobiopsy versus open lung biopsy in the diagnosis of interstitial lung disease (COLDICE): protocol of a multicentre study. BMJ Open Respir Res 2019; 6:e000443. [PMID: 31321059 PMCID: PMC6606076 DOI: 10.1136/bmjresp-2019-000443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022] Open
Abstract
Introduction Transbronchial lung cryobiopsy (TBLC) is a novel, minimally invasive technique for obtaining lung tissue for histopathological assessment in interstitial lung disease (ILD). Despite its increasing popularity, the diagnostic accuracy of TBLC is not yet known. The COLDICE Study (Cryobiopsy versus Open Lung biopsy in the Diagnosis of Interstitial lung disease allianCE) aims to evaluate the agreement between TBLC and surgical lung biopsy sampled concurrently from the same patients, for both histopathological and multidisciplinary discussion (MDD) diagnoses. Methods and analysis This comparative, multicentre, prospective trial is enrolling patients with ILD requiring surgical lung biopsy to aid with their diagnosis. Participants are consented for both video-assisted thoracoscopic surgical (VATS) biopsy and TBLC within the same anaesthetic episode. Specimens will be blindly assessed by three expert pathologists both individually and by consensus. Each tissue sample will then be considered in conjunction with clinical and radiological data, within a centralised MDD. Each patient will be presented twice in random order, once with TBLC data and once with VATS data. Meeting participants will be blinded to the method of tissue sampling. The accuracy of TBLC will be assessed by agreement with VATS at (1) histopathological analysis and (2) MDD diagnosis. Data will be collected on interobserver agreement between pathologists, interobserver agreement between MDD participants, and detailed clinical and procedural characteristics. Ethics and dissemination The study is being conducted in accordance with the International Conference on Harmonisation Guideline for Good Clinical Practice and Australian legislation for the ethical conduct of research. Trial registration number ACTRN12615000718549.
Collapse
Affiliation(s)
- Lauren K Troy
- Respiratory Medicine, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Christopher Grainge
- Faculty of Medicine, University of Newcastle, Newcastle, New South Wales, Australia.,Respiratory Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | - Tamera Corte
- Respiratory Medicine, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Jonathan P Williamson
- Respiratory Medicine, Liverpool Hospital, Liverpool, New South Wales, Australia.,Respiratory Medicine, Macquarie University Hospital, North Ryde BC, New South Wales, Australia
| | - Michael P Vallely
- Cardiothoracic Surgery, Macquarie University Hospital, North Ryde BC, New South Wales, Australia
| | - Wendy Cooper
- Sydney Medical School, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Annabelle M Mahar
- Sydney Medical School, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Simon Lai
- Radiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Ellie Mulyadi
- Radiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Paul J Torzillo
- Respiratory Medicine, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | - Matthew Salamonsen
- Respiratory Medicine, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Garrick Don
- Respiratory Medicine, Royal North Shore Hospital School, Saint Leonards, New South Wales, Australia
| | - Jeffrey Myers
- Pathology, University of Michigan Hospital, Ann Arbor, Michigan, USA
| | - Ganesh Raghu
- Division of Pulmonary, Sleep and Critical Care Medicine, University of Washington, Seattle, Washington, USA
| | - Edmund M T Lau
- Respiratory Medicine, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia.,Sydney Medical School, The University of Sydney Faculty of Medicine and Health, Sydney, New South Wales, Australia
| | | |
Collapse
|
157
|
Feng F, Zhang J, Wang Z, Wu Q, Zhou X. Efficacy and safety of N-acetylcysteine therapy for idiopathic pulmonary fibrosis: An updated systematic review and meta-analysis. Exp Ther Med 2019; 18:802-816. [PMID: 31258714 PMCID: PMC6566037 DOI: 10.3892/etm.2019.7579] [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: 11/26/2018] [Accepted: 04/12/2019] [Indexed: 12/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal lung disease with poor prognosis and limited treatment options. N-acetylcysteine (NAC), an anti-oxidant drug, has promising potential in the treatment of IPF. In the present systematic review and meta-analysis, the efficacy and safety of NAC for IPF were investigated. The following databases were comprehensively searched for relevant studies published until August 2018: Pubmed, Embase, Cochrane library, Chinese National Knowledge Infrastructure, Wangfang Database, VIP and the Chinese Biology Medical Database. A total of 21 controlled trials assessing the efficacy and safety of NAC therapy for IPF were identified and primary outcomes [forced vital capacity (FVC), adverse side effects] and secondary outcomes [diffusing capacity for carbon monoxide (DLCO) and its percentage predicted value (DLCO%), vital capacity (VC), partial arterial oxygen pressure (PaO2), 6-min walking distance test and mortality] were extracted for the meta-analysis. The risk ratio and mean difference or standardized mean difference with 95% confidence interval were calculated using RevMan 5.3 software. Analysis of the pooled data revealed that, compared with control treatments (routine treatment or drugs other than anti-oxidants), NAC therapy reduced the decline in lung function, as indicated by the FVC and DLCO, and slowed the progression of the disease, as indicated by the PaO2, while complications and mortality were similar. These results suggest good efficacy, tolerability and safety of the treatment. Furthermore, subgroup analysis revealed that combined therapy including NAC for IPF might be more effective than NAC monotherapy, while oral administration of NAC was safer than inhalation. In conclusion, the results of the present review and meta-analysis provide important information that may serve as a guide regarding NAC therapy for IPF in clinical practice.
Collapse
Affiliation(s)
- Fanchao Feng
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Jiarui Zhang
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Zhichao Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Qi Wu
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Xianmei Zhou
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China.,Department of Respiratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
158
|
Barros A, Oldham J, Noth I. Genetics of Idiopathic Pulmonary Fibrosis. Am J Med Sci 2019; 357:379-383. [PMID: 31010464 PMCID: PMC10538522 DOI: 10.1016/j.amjms.2019.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 01/28/2019] [Accepted: 02/10/2019] [Indexed: 01/02/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common form of interstitial lung disease. IPF is a complex disease, with environmental and genetic factors variably contributing to disease susceptibility and outcomes. A host of common gene variants with modest effect size impart disease risk in patients with sporadic IPF, while rare variants with large effect size influence disease risk in those with familial interstitial pneumonia. In this review, we highlight several common and rare variants underpinning IPF risk and call attention to recently published studies informing our understanding of this risk.
Collapse
Affiliation(s)
- Andrew Barros
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia.
| | - Justin Oldham
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The University of California at Davis, Sacramento, California.
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
159
|
Chen MJ, Yang GL, Ding YX, Tong ZQ. Efficacy of TCM therapy of tonifying lung-kidney's Qi-deficiency in a case of idiopathic pulmonary fibrosis: A case report. Medicine (Baltimore) 2019; 98:e15140. [PMID: 31045757 PMCID: PMC6504330 DOI: 10.1097/md.0000000000015140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
RATIONALE Idiopathic pulmonary fibrosis (IPF) is a special form of spontaneous, chronic, progressive interstitial fibrotic pneumonia of unknown cause, and treatments for IPF have shown a poor prognosis. This study reports a new treatment, traditional Chinese medicine (TCM) therapy, for tonifying Qi-deficiency of lung-kidney in a 64-year-old patient with IPF. PATIENT CONCERNS A 64-year-old man, who complained of cough and asthma, was diagnosed as IPF with mild impairment in lung function by thoracic high-resolution computed tomography and pulmonary function test. He received an 18-month N-acetylcysteine monotherapy but had no improvement in lung function. DIAGNOSES IPF with mild impairment in lung function was diagnosed. INTERVENTIONS The Chinese herbal medicine composition was decocted in 300 ml water for oral administration with 150 ml decoction twice daily in June 2017. OUTCOMES The pulmonary function test showed that diffusing capacity for carbon monoxide had increased to 81% of predicted back to normal after 2-month TCM monotherapy. And diffusing capacity for carbon monoxide had increased to 89% of predicted, and forced expiratory volume in 1 s/forced vital capacity ratio increased to 92% at 14-month follow-up. No adverse events occurred during the 14 months of therapy and observation. LESSONS The treatment by TCM therapy of tonifying lung-kidney's Qi-deficiency for IPF can improve the pulmonary function and reverse disease progression; it may be considered as a complementary treatment for IPF with mild-to-moderate impairment. However, the insights provided in this case report require further exploration and verification.
Collapse
Affiliation(s)
- Ming-Jun Chen
- Department of Traditional Chinese Medicine and Acupuncture, Chinese PLA General Hospital
| | - Ge-Lliang Yang
- Department of Traditional Chinese Medicine and Acupuncture, Chinese PLA General Hospital
| | | | - Zhan-Qi Tong
- Department of Traditional Chinese Medicine and Acupuncture, Chinese PLA General Hospital
| |
Collapse
|
160
|
Tsai MJ, Chang WA, Liao SH, Chang KF, Sheu CC, Kuo PL. The Effects of Epigallocatechin Gallate (EGCG) on Pulmonary Fibroblasts of Idiopathic Pulmonary Fibrosis (IPF)-A Next-Generation Sequencing and Bioinformatic Approach. Int J Mol Sci 2019; 20:E1958. [PMID: 31013581 PMCID: PMC6514693 DOI: 10.3390/ijms20081958] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 12/19/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disabling and lethal chronic progressive pulmonary disease. Epigallocatechin gallate (EGCG) is a polyphenol, which is the major biological component of green tea. The anti-oxidative, anti-inflammatory, and anti-fibrotic effects of EGCG have been shown in some studies, whereas its effects in altering gene expression in pulmonary fibroblasts have not been systematically investigated. This study aimed to explore the effect of EGCG on gene expression profiles in fibroblasts of IPF. The pulmonary fibroblasts from an IPF patient were treated with either EGCG or water, and the expression profiles of mRNAs and microRNAs were determined by next-generation sequencing (NGS) and analyzed with the bioinformatics approach. A total of 61 differentially expressed genes and 56 differentially expressed microRNAs were found in EGCG-treated IPF fibroblasts. Gene ontology analyses revealed that the differentially expressed genes were mainly involved in the biosynthetic and metabolic processes of cholesterol. In addition, five potential altered microRNA-mRNA interactions were found, including hsa-miR-939-5p-PLXNA4, hsa-miR-3918-CTIF, hsa-miR-4768-5p-PDE5A, hsa-miR-1273g-3p-VPS53, and hsa-miR-1972-PCSK9. In summary, differentially expressed genes and microRNAs in response to EGCG treatment in IPF fibroblasts were identified in the current study. Our findings provide a scientific basis to evaluate the potential benefits of EGCG in IPF treatment, and warrant future studies to understand the role of molecular pathways underlying cholesterol homeostasis in the pathogenesis of IPF.
Collapse
Affiliation(s)
- Ming-Ju Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Respiratory Therapy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ssu-Hui Liao
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | | | - Chau-Chyun Sheu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Respiratory Therapy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
161
|
Courtwright AM, El-Chemaly S. Telomeres in Interstitial Lung Disease: The Short and the Long of It. Ann Am Thorac Soc 2019; 16:175-181. [PMID: 30540921 PMCID: PMC6376948 DOI: 10.1513/annalsats.201808-508cme] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/25/2018] [Indexed: 01/01/2023] Open
Abstract
Telomeres are repetitive nucleotide sequences that cap linear chromosomes, thereby limiting progressive chromosomal shortening during cell replication. In conjunction with environmental factors, common single-nucleotide polymorphisms and rare and ultra-rare telomere-related mutations are associated with accelerated telomere shortening resulting in organ dysfunction, including interstitial lung disease (ILD). The most common telomere-related mutation-associated ILD is idiopathic pulmonary fibrosis (IPF). Up to one-third of individuals with familial IPF have shortened telomeres and/or carry a telomere-related mutation, and 1 in 10 individuals with sporadic IPF have telomere-related mutations. Regardless of ILD phenotype, individuals with short telomeres and/or known telomere-related mutations have more rapid disease progression and shorter lung transplant-free survival. Management should include initiation of antifibrotic agents for those with an IPF phenotype and early referral to a transplant center. Patients with ILD being considered for transplant should be screened for short telomeres if there is a significant family history of pulmonary fibrosis or evidence of extrapulmonary organ dysfunction associated with a short telomere syndrome. Post-transplant management of recipients with telomere-related mutations should include careful adjustment of immunosuppression regimens on the basis of bone marrow reserve. Data on the impact of shortened telomeres on post-transplant outcomes, however, remain mixed.
Collapse
Affiliation(s)
- Andrew M. Courtwright
- Division of Pulmonary and Critical Care Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
162
|
Ballester B, Milara J, Cortijo J. Idiopathic Pulmonary Fibrosis and Lung Cancer: Mechanisms and Molecular Targets. Int J Mol Sci 2019; 20:ijms20030593. [PMID: 30704051 PMCID: PMC6387034 DOI: 10.3390/ijms20030593] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/18/2019] [Accepted: 01/28/2019] [Indexed: 12/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common idiopathic interstitial pulmonary disease with a median survival of 2–4 years after diagnosis. A significant number of IPF patients have risk factors, such as a history of smoking or concomitant emphysema, both of which can predispose the patient to lung cancer (LC) (mostly non-small cell lung cancer (NSCLC)). In fact, IPF itself increases the risk of LC development by 7% to 20%. In this regard, there are multiple common genetic, molecular, and cellular processes that connect lung fibrosis with LC, such as myofibroblast/mesenchymal transition, myofibroblast activation and uncontrolled proliferation, endoplasmic reticulum stress, alterations of growth factors expression, oxidative stress, and large genetic and epigenetic variations that can predispose the patient to develop IPF and LC. The current approved IPF therapies, pirfenidone and nintedanib, are also active in LC. In fact, nintedanib is approved as a second line treatment in NSCLC, and pirfenidone has shown anti-neoplastic effects in preclinical studies. In this review, we focus on the current knowledge on the mechanisms implicated in the development of LC in patients with IPF as well as in current IPF and LC-IPF candidate therapies based on novel molecular advances.
Collapse
Affiliation(s)
- Beatriz Ballester
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBERES, Health Institute Carlos III, 28029 Valencia, Spain.
| | - Javier Milara
- CIBERES, Health Institute Carlos III, 28029 Valencia, Spain.
- Pharmacy Unit, University Clinic Hospital of Valencia, 46010 Valencia, Spain.
- Institute of Health Research-INCLIVA, 46010 Valencia, Spain.
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBERES, Health Institute Carlos III, 28029 Valencia, Spain.
- Research and teaching Unit, University General Hospital Consortium, 46014 Valencia, Spain.
| |
Collapse
|
163
|
Abstract
This is a time of substantial progress in the evaluation and care of patients with idiopathic pulmonary fibrosis (IPF). In addition to the approval and widespread availability of the first IPF-specific therapies, there have been improvements in imaging interpretation and lung biopsy methods to enable more expeditious and more accurate diagnosis. Recent advances in identifying genetic factors that underlie susceptibility to IPF and affect prognosis have raised the possibility of personalized therapeutic approaches in the future. Further, evolving work is elucidating novel mechanisms influencing epithelial, mesenchymal, and inflammatory cell responses during the injury-repair process, thus advancing understanding of disease pathogenesis. As analytic approaches mature, the field is now poised to harness the power of rapidly advancing "omics" technologies to further accelerate progress.
Collapse
Affiliation(s)
- Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA; ,
- Department of Veterans Affairs Medical Center, Nashville, Tennessee 37212, USA
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA; ,
- Department of Veterans Affairs Medical Center, Nashville, Tennessee 37212, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37212, USA
| |
Collapse
|
164
|
Kolilekas L, Papiris S, Bouros D. Existing and emerging treatments for idiopathic pulmonary fibrosis. Expert Rev Respir Med 2019; 13:229-239. [DOI: 10.1080/17476348.2019.1568244] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Lykourgos Kolilekas
- 7th Department of Pneumonology, Hospital for Diseases of the Chest, “Sotiria”, Athens, Greece
| | - Spyridon Papiris
- 2nd Department of respiratory Medicine, National and Kapodistrian University of Athens, Attikon Hospital, Athens, Greece
| | - Demosthenes Bouros
- First Academic Department of Pneumonology, Interstitial Lung Diseases Unit, Hospital for Diseases of the Chest, “Sotiria”, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
165
|
Integrating Genomics Into Management of Fibrotic Interstitial Lung Disease. Chest 2019; 155:1026-1040. [PMID: 30660786 DOI: 10.1016/j.chest.2018.12.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/30/2018] [Accepted: 12/20/2018] [Indexed: 12/31/2022] Open
Abstract
Fibrotic interstitial lung diseases (ILDs) have a high mortality rate with an unpredictable disease course and clinical features that frequently overlap. Recent data indicate important roles for genomics in the mechanisms underlying susceptibility and progression of pulmonary fibrosis. The impact of these genomic markers on pharmacotherapy and their contribution to outcomes is increasingly recognized. Interstitial lung abnormalities, frequently considered representative of early ILD, have been consistently associated with the MUC5B promoter polymorphism, a common gene variant. Other rare gene variant mutations, including TERT, TERC, SFTPC, and DKC1, may be present in patients with familial interstitial pneumonia and are frequently associated with a usual interstitial pneumonia pattern of fibrosis. The minor allele of the MUC5B rs35705950 genotype is prevalent in several sporadic forms of ILD, including idiopathic pulmonary fibrosis and chronic hypersensitivity pneumonitis. Gene mutations that characterize familial pulmonary fibrosis may be present in patients with connective tissue disease-related ILD, such as rheumatoid arthritis-ILD. Additionally, shorter telomere lengths and mutations in telomere biology-related genes have been demonstrated in both familial and sporadic ILD, with significant implications for disease progression, lung function, and survival. An improved understanding of the impact of genetic and genomic risk factors on disease progression would better guide personalized therapeutic choices in persons with fibrotic ILD.
Collapse
|
166
|
Future Directions for IPF Research. Respir Med 2019. [DOI: 10.1007/978-3-319-99975-3_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
167
|
|
168
|
|
169
|
|
170
|
Inchingolo R, Varone F, Sgalla G, Richeldi L. Existing and emerging biomarkers for disease progression in idiopathic pulmonary fibrosis. Expert Rev Respir Med 2018; 13:39-51. [DOI: 10.1080/17476348.2019.1553620] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Riccardo Inchingolo
- Pulmonary Medicine Unit, Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Francesco Varone
- Pulmonary Medicine Unit, Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giacomo Sgalla
- Pulmonary Medicine Unit, Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Richeldi
- Pulmonary Medicine Unit, Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
171
|
Abstract
Lysyl oxidase (LOX) and lysyl oxidase-like proteins (LOXL), a family of extracellular matrix (ECM) crosslinking enzymes that have been recognised as playing an important role in fibrogenesis for more than 40 years, are logical targets for antifibrotic treatments. Pulmonary fibrosis, especially idiopathic pulmonary fibrosis (IPF), is a progressive and lethal disease characterised by excessive deposition of ECM in the lung parenchyma. In this review, we discuss the current clinical approaches for IPF and review members of LOX family-LOX, LOXL1, LOXL2, LOXL3 and LOXL4 in IPF patients and in animal models of bleomycin-induced pulmonary fibrosis. Although these findings are controversial and require further validation, LOX/LOXL1/LOXL2 as potential therapeutic targets for IPF deserve continued attention. So far to our knowledge, LOXL3 or LOXL4 has not clearly shown specific therapeutic potential.
Collapse
Affiliation(s)
- Lijun Chen
- a Department of Pharmacology , Zhongshan Medical School, Sun Yat-sen University , Guangzhou , China
| | - Shifeng Li
- a Department of Pharmacology , Zhongshan Medical School, Sun Yat-sen University , Guangzhou , China
| | - Wande Li
- b Department of Biochemistry , Boston University School of Medicine , Boston , MA , USA
| |
Collapse
|
172
|
Saito A, Horie M, Micke P, Nagase T. The Role of TGF-β Signaling in Lung Cancer Associated with Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2018; 19:ijms19113611. [PMID: 30445777 PMCID: PMC6275044 DOI: 10.3390/ijms19113611] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease of unknown etiology and dismal prognosis. IPF patients are known to have an increased risk of lung cancer and careful decision-making is required for the treatment of lung cancer associated with IPF. Transforming growth factor (TGF)-β signaling plays a central role in tissue fibrosis and tumorigenesis. TGF-β-mediated pathological changes that occur in IPF lung tissue may promote the process of field cancerization and provide the microenvironment favorable to cancer initiation and progression. This review summarizes the current knowledge related to IPF pathogenesis and explores the molecular mechanisms that underlie the occurrence of lung cancer in the background of IPF, with an emphasis on the multifaceted effects of TGF-β signaling.
Collapse
Affiliation(s)
- Akira Saito
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
- Division for Health Service Promotion, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Masafumi Horie
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden.
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
173
|
Saito S, Lasky JA, Hagiwara K, Kondoh Y. Ethnic differences in idiopathic pulmonary fibrosis: The Japanese perspective. Respir Investig 2018; 56:375-383. [PMID: 30061050 DOI: 10.1016/j.resinv.2018.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 05/03/2018] [Accepted: 06/12/2018] [Indexed: 01/20/2023]
Abstract
Epidemiologic data suggest that there are ethnic differences between Japanese and other populations with regard to the important clinical aspects of interstitial lung disease (ILD), such as the cause of death and prognostic factors in patients with idiopathic pulmonary fibrosis (IPF). Acute exacerbation (AE) of IPF may be more common in Japan than in the rest of the world, although this suggestion remains controversial. Moreover, AE of ILD induced by gefitinib may also be more common in Japan, indicating that Japanese patients have a genetic vulnerability or susceptibility to AE. Recent large-scale studies are starting to reveal ethnic differences in the genetics of ILD, including the prevalence of the genetic polymorphisms associated with the clinical course of ILD. We anticipate that ongoing and upcoming research regarding ethnic differences will continue to provide valuable insights into the pathogenesis and management of ILD.
Collapse
Affiliation(s)
- Shigeki Saito
- Department of Medicine, Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University Health Science Center, New Orleans, LA, USA.
| | - Joseph A Lasky
- Department of Medicine, Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University Health Science Center, New Orleans, LA, USA.
| | - Koichi Hagiwara
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
| | - Yasuhiro Kondoh
- Nagoya University School of Medicine, Department of Respiratory Medicine and Allergy, Tosei General Hospital, 160 Nishioiwake-cho, Seto, Aichi 489-8642, Japan.
| |
Collapse
|
174
|
Thannickal VJ, Antony VB. Is personalized medicine a realistic goal in idiopathic pulmonary fibrosis? Expert Rev Respir Med 2018; 12:441-443. [PMID: 29671654 PMCID: PMC6157605 DOI: 10.1080/17476348.2018.1464913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 04/11/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Victor J Thannickal
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine , University of Alabama , Birmingham , AL , USA
| | - Veena B Antony
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine , University of Alabama , Birmingham , AL , USA
| |
Collapse
|
175
|
Wells AU, Brown KK, Flaherty KR, Kolb M, Thannickal V. What's in a name? That which we call IPF, by any other name would act the same. Eur Respir J 2018; 51:13993003.00692-2018. [DOI: 10.1183/13993003.00692-2018] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/01/2018] [Indexed: 12/11/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) remains a truly idiopathic fibrotic disease, with a modest genetic predilection and candidate triggers but no overall explanation for the development of disease in non-familial cases. Agreement on terminology has contributed to major clinical and translational advances since the millennium. It is likely that the entity currently captured by the term “IPF” will be radically reclassified over the next decade, either through “splitting” (into IPF subgroups responding selectively to individual disease-modifying agents) or through “lumping” of IPF with other forms of progressive fibrotic lung disease (with shared pathogenetic mechanisms and IPF-like disease behaviour). In this perspective, we summarise the clinical and pathogenetic justification for a focus on “the progressive fibrotic phenotype” in future clinical and translational research. By this means, we can hope to address the needs of non-IPF patients with inexorably progressive fibrotic disease, currently disenfranchised by lack of access to agents that are efficacious in IPF. In this regard, ongoing trials of anti-fibrotic therapies in non-IPF patients with progressive fibrosis may be highly influential. Future revision of IPF nomenclature may be warranted if there are major conceptual changes but without compelling justification, the benefits of renaming IPF are likely to be outweighed by the resulting confusion.
Collapse
|
176
|
Affiliation(s)
- David J Lederer
- From the Departments of Medicine and Epidemiology, Columbia University Irving Medical Center (D.J.L.), and the Department of Medicine, Weill Cornell Medical Center (F.J.M.) - both in New York
| | - Fernando J Martinez
- From the Departments of Medicine and Epidemiology, Columbia University Irving Medical Center (D.J.L.), and the Department of Medicine, Weill Cornell Medical Center (F.J.M.) - both in New York
| |
Collapse
|
177
|
Raghu G, Noth I, Martinez F. N-acetylcysteine for idiopathic pulmonary fibrosis: the door is still open. THE LANCET RESPIRATORY MEDICINE 2018; 5:e1-e2. [PMID: 28000596 DOI: 10.1016/s2213-2600(16)30327-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/22/2016] [Indexed: 10/20/2022]
Affiliation(s)
- Ganesh Raghu
- Center for Interstitial Lung Disease, University of Washington, Seattle, WA 98195, USA.
| | - Imre Noth
- University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
178
|
Newton CA, Molyneaux PL, Oldham JM. Clinical Genetics in Interstitial Lung Disease. Front Med (Lausanne) 2018; 5:116. [PMID: 29755982 PMCID: PMC5932165 DOI: 10.3389/fmed.2018.00116] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 12/14/2022] Open
Abstract
Interstitial lung disease (ILD) comprises a heterogeneous group of diffuse parenchymal lung processes with overlapping clinical, radiographic, and histopathologic features. Among the most common and deadly ILDs are idiopathic pulmonary fibrosis (IPF) and chronic hypersensitivity pneumonitis (CHP). As the name implies, the cause of IPF remains elusive, but a variety of genetic and infectious risk factors have been identified. CHP results from chronic inhalation of an organic antigen, usually of avian or mold origin, and may occur in patients with a genetic predisposition. While IPF is treated with anti-fibrotic compounds, CHP is generally treated by suppression of the immune system and elimination of the causative antigen. Despite advances in our understanding of IPF and CHP, there exists substantial variability in the diagnosis and treatment of these disease processes. Furthermore, IPF and CHP natural history and treatment response remain far from uniform, leaving it unclear which patients derive the most benefit from disease-specific therapy. While clinical prediction models have improved our understanding of outcome risk in patients with various forms of ILD, recent advances in genomic technology provides a valuable opportunity to begin understanding the basis for outcome variability. Such advances will ultimately allow for the incorporation of genomic markers into risk stratification and clinical decision-making. In this piece, we highlight recent advances in our understanding of the genomic factors that influence susceptibility and outcome risk among patients with IPF and CHP. Genomic modalities used to identify these genomic markers include genome-wide association studies, analyses of gene expression, drug–gene interaction testing, telomere length determination, telomerase mutation analysis, and studies of the lung microbiome. We then identify gaps in knowledge that should be addressed to help facilitate the incorporation of these genomic technologies into ILD clinical practice.
Collapse
Affiliation(s)
- Chad A Newton
- Eugene McDermott Centre for Human Growth and Development, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Philip L Molyneaux
- Fibrosis Research Group, National Heart and Lung Institute, Imperial College, London, United Kingdom.,National Institute for Health Research Respiratory Biomedical Research Unit, Royal Brompton Hospital, London, United Kingdom
| | - Justin M Oldham
- Division of Pulmonary, Department of Medicine, Critical Care and Sleep Medicine, University of California at Davis, Davis, CA, United States
| |
Collapse
|
179
|
Kokosi MA, Margaritopoulos GA, Wells AU. Personalised medicine in interstitial lung diseases. Eur Respir Rev 2018; 27:27/148/170117. [DOI: 10.1183/16000617.0117-2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 03/05/2018] [Indexed: 12/14/2022] Open
Abstract
Interstitial lung diseases in general, and idiopathic pulmonary fibrosis in particular, are complex disorders with multiple pathogenetic pathways, various disease behaviour profiles and different responses to treatment, all facets that make personalised medicine a highly attractive concept. Personalised medicine is aimed at describing distinct disease subsets taking into account individual lifestyle, environmental exposures, genetic profiles and molecular pathways. The cornerstone of personalised medicine is the identification of biomarkers that can be used to inform diagnosis, prognosis and treatment stratification. At present, no data exist validating a personalised approach in individual diseases. However, the importance of the goal amply justifies the characterisation of genotype and pathway signatures with a view to refining prognostic evaluation and trial design, with the ultimate aim of selecting treatments according to profiles in individual patients.
Collapse
|
180
|
Abstract
PURPOSE OF REVIEW A number of recent studies have explored the possibility to apply personalized medicine to interstitial lung diseases (ILDs), particularly idiopathic pulmonary fibrosis (IPF), the most common and deadly of the idiopathic interstitial pneumonias. In our review, we summarize and discuss the most recent literature on personalized medicine in IPF as well as hypersensitivity pneumonitis and sarcoidosis, with emphasis on patient subgroups for which a personalized approach to disease prognostication and management may become a reality in the near future. RECENT FINDINGS Most of the studies that have explored the applicability of personalized medicine to ILDs have been conducted in patients with IPF. Such studies have suggested the existence of several distinct disease subgroups defined by similar genetic profiles, molecular pathways, exposures and individual lifestyles. Personalized medicine in hypersensitivity pneumonitis is in its infancy. The development and applicability of personalized medicine to sarcoidosis, on the other hand, remains problematic for several reasons, including the lack of a diagnostic gold standard, the highly variable and unpredictable disease course, particularly across patients of different ethnicities, the poor correlation between disease activity and disease severity and the lack of a validated management algorithm. SUMMARY A number of distinct patient subgroups have been identified in ILDs. Although available data need to be validated longitudinally, the possibility to study homogeneous groups of patients may allow prediction of disease behavior and response to treatment with dramatic clinical implications.
Collapse
|
181
|
Fois AG, Paliogiannis P, Sotgia S, Mangoni AA, Zinellu E, Pirina P, Carru C, Zinellu A. Evaluation of oxidative stress biomarkers in idiopathic pulmonary fibrosis and therapeutic applications: a systematic review. Respir Res 2018; 19:51. [PMID: 29587761 PMCID: PMC5872514 DOI: 10.1186/s12931-018-0754-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/19/2018] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF), a fatal lung disease of unknown origin, is characterized by chronic and progressive fibrosing interstitial pneumonia which progressively impairs lung function. Oxidative stress is one of the main pathogenic pathways in IPF. The aim of this systematic review was to describe the type of markers of oxidative stress identified in different biological specimens and the effects of antioxidant therapies in patients with IPF. METHODS We conducted a systematic search of publications listed in electronic databases (Pubmed, Web of Science, Scopus and Google Scholar) from inception to October 2017. Two investigators independently reviewed all identified articles to determine eligibility. RESULTS After a substantial proportion of the initially identified articles (n = 554) was excluded because they were duplicates, abstracts, irrelevant, or did not meet the selection criteria, we identified 30 studies. In each study, we critically appraised the type, site (systemic vs. local, e.g. breath, sputum, expired breath condensate, epithelial lining fluid, bronchoalveolar lavage, and lung tissue specimens), and method used for measuring the identified oxidative stress biomarkers. Furthermore, the current knowledge on antioxidant therapies in IPF was summarized. CONCLUSIONS A number of markers of oxidative stress, with individual advantages and limitations, have been described in patients with IPF. Nevertheless, trials of antioxidant treatments have been unable to demonstrate consistent benefits, barring recent pharmacogenomics data suggesting different results in specific genotype subgroups of patients with IPF.
Collapse
Affiliation(s)
- Alessandro G Fois
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy. .,Department of Respiratory Diseases, University Hospital Sassari (AOU), Sassari, Italy.
| | | | - Salvatore Sotgia
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Elisabetta Zinellu
- Department of Respiratory Diseases, University Hospital Sassari (AOU), Sassari, Italy
| | - Pietro Pirina
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy.,Department of Respiratory Diseases, University Hospital Sassari (AOU), Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
182
|
Fibrosis: Lessons from OMICS analyses of the human lung. Matrix Biol 2018; 68-69:422-434. [PMID: 29567123 DOI: 10.1016/j.matbio.2018.03.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/16/2018] [Accepted: 03/16/2018] [Indexed: 12/30/2022]
Abstract
In recent decades there has been a significant shift in our understanding of idiopathic pulmonary fibrosis (IPF), a progressive and lethal disorder. While initially much of the mechanistic understanding was derived from hypotheses generated from animal models of disease, in recent decades new insights derived from humans with IPF have taken precedence. This is mainly because of the establishment of large collections of IPF lung tissues and patient cohorts, and the emergence of high throughput profiling technologies collectively termed 'omics' technologies based on their shared suffix. In this review we describe impacts of 'omics' analyses of human IPF samples on our understanding of the disease. In particular, we discuss the results of genomics and transcriptomics studies, as well as proteomics, epigenomics and metabolomics. We then describe how these findings can be integrated in a modified paradigm of human idiopathic pulmonary fibrosis, that introduces the 'hallmarks of aging' as a central theme in the IPF lung. This allows resolution of all the disparate cellular and molecular features in IPF, from the central role of epithelial cells, through the dramatic phenotypic alterations observed in fibroblasts and the numerous aberrations that inflammatory cells exhibit. We end with reiterating a call for renewed efforts to collect and analyze carefully characterized human tissues, in ways that would facilitate implementation of novel technologies for high resolution single cell omics profiling.
Collapse
|
183
|
Mercer PF, Chambers RC. Innate immune signaling and stem cell renewal in idiopathic pulmonary fibrosis. Nat Med 2018; 22:1210-1212. [PMID: 27824820 DOI: 10.1038/nm.4230] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Paul F Mercer
- Centre for Inflammation and Tissue Repair, UCL Respiratory, UCL Division of Medicine, University College London, London, United Kingdom
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, UCL Respiratory, UCL Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
184
|
Oldham JM, Witt LJ, Adegunsoye A, Chung JH, Lee C, Hsu S, Chen LW, Husain A, Montner S, Vij R, Strek ME, Noth I. N-acetylcysteine exposure is associated with improved survival in anti-nuclear antibody seropositive patients with usual interstitial pneumonia. BMC Pulm Med 2018; 18:30. [PMID: 29422061 PMCID: PMC5806226 DOI: 10.1186/s12890-018-0599-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
Background Mortality is similarly high among individuals with usual interstitial pneumonia (UIP) due to idiopathic pulmonary fibrosis (IPF) and interstitial pneumonia with autoimmune features (IPAF). Circulating anti-nuclear antibodies (ANA) are commonly found in this patient population, suggesting possible aberrant immune activation. Because an environment of oxidative stress can result from immunologic activation, we hypothesized that ANA positive patients with UIP would have improved outcome when exposed to the antioxidant N-acetylcysteine (NAC) compared to ANA negative patients. Methods A single center, retrospective cohort analysis was performed. Patients with UIP due to IPF and IPAF were stratified according to ANA status to and NAC exposure. Transplant-free survival (TFS) was assessed using the Kaplan-Meier estimator and multivariable Cox regression adjusted for diagnosis, gender/age/physiology score, immunosuppressant exposure and anti-fibrotic exposure. Results Of 293 individuals with UIP due to IPF (74%) or IPAF (26%), NAC exposure was documented in 58 (19.8%). Among NAC exposed individuals, 33 (56.9%) were ANA seropositive and 25 (43.1%) were seronegative. NAC exposure was associated with improved TFS survival among ANA seropositive individuals in unadjusted analysis (plogrank = 0.02) and after multi-variable adjustment (HR 0.51, 95% CI 0.30–0.87; p = 0.01). There was no association between NAC exposure and TFS in ANA seronegative individuals (HR 1.26, 95% CI 0.69–2.32; p = 0.45). Formal interaction testing confirmed NAC*ANA interaction (p = 0.04) and sensitivity analysis demonstrated an increasing effect size associated with NAC therapy as ANA titer increased. Among patients with available genetic data, a marginally higher proportion of ANA positive patients (p = 0.08) carried the rs3750920 (TOLLIP) genotype previously shown to predict favorable outcome in NAC exposed patients. Conclusion NAC exposure is associated with improved transplant-free survival ANA positive patients with UIP. These findings support the prospective collection of ANA data in in future NAC clinical trials performed in patients with UIP. Electronic supplementary material The online version of this article (10.1186/s12890-018-0599-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Justin M Oldham
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, The University of California at Davis, Sacramento, CA, USA.
| | - Leah J Witt
- Department of Medicine; Division of Geriatrics, University of California at San Francisco, San Francisco, USA
| | - Ayodeji Adegunsoye
- Department of Medicine; Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, USA
| | - Jonathan H Chung
- Department of Radiology, The University of Chicago, Chicago, USA
| | - Cathryn Lee
- Department of Medicine; Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, USA
| | - Scully Hsu
- Department of Medicine; Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, USA
| | - Lena W Chen
- Department of Medicine; Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, USA
| | - Aliya Husain
- Department of Pathology, The University of Chicago, Chicago, USA
| | - Steven Montner
- Department of Radiology, The University of Chicago, Chicago, USA
| | - Rekha Vij
- Department of Medicine; Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, USA
| | - Mary E Strek
- Department of Medicine; Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, USA
| | - Imre Noth
- Department of Medicine; Section of Pulmonary and Critical Care Medicine, The University of Chicago, Chicago, USA
| |
Collapse
|
185
|
Drakopanagiotakis F, Wujak L, Wygrecka M, Markart P. Biomarkers in idiopathic pulmonary fibrosis. Matrix Biol 2018; 68-69:404-421. [PMID: 29408012 DOI: 10.1016/j.matbio.2018.01.023] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/28/2018] [Accepted: 01/29/2018] [Indexed: 12/15/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, debilitating, fibrotic lung disease leading to respiratory failure and ultimately to death. Being the prototype of interstitial lung diseases, IPF is characterized by marked heterogeneity regarding its clinical course. Despite significant progress in the understanding of its pathogenesis, we still cannot reliably predict the course of the disease and the response to treatment of an individual patient. Non-invasive biomarkers, in particular serum biomarkers, for the (early) diagnosis, differential diagnosis, prognosis and prediction of therapeutic response are urgently needed. Numerous molecules involved in alveolar epithelial cell injury, fibroproliferation and matrix remodeling as well as immune regulation have been proposed as potential biomarkers. Furthermore, genetic variants of TOLLIP, MUC5B, and other genes are associated with a differential response to treatment and with the development and/or the prognosis of IPF. Additionally, the bacterial signature in IPF lungs, as shown from microbiome analyses, as well as mitochondrial DNA seem to have promising roles as biomarkers. Moreover, combination of multiple biomarkers may identify comprehensive biomarker signatures in IPF patients. However, there is still a long way until these potential biomarkers complete or substitute for the clinical and functional parameters currently available for IPF.
Collapse
Affiliation(s)
- F Drakopanagiotakis
- Department of Pulmonary Medicine (Medical Clinic V), Fulda Hospital, University Medicine Marburg, Campus Fulda, Pacelliallee 4, 36043 Fulda, Germany
| | - Lukasz Wujak
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - P Markart
- Department of Pulmonary Medicine (Medical Clinic V), Fulda Hospital, University Medicine Marburg, Campus Fulda, Pacelliallee 4, 36043 Fulda, Germany; Department of Internal Medicine, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany.
| |
Collapse
|
186
|
Moor CC, Heukels P, Kool M, Wijsenbeek MS. Integrating Patient Perspectives into Personalized Medicine in Idiopathic Pulmonary Fibrosis. Front Med (Lausanne) 2017; 4:226. [PMID: 29326935 PMCID: PMC5742327 DOI: 10.3389/fmed.2017.00226] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/27/2017] [Indexed: 12/29/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal disease which has a major impact on patients' quality of life (QOL). Except for lung transplantation, there is no curative treatment option. Fortunately, two disease-modifying drugs that slow down disease decline were recently approved. Though this is a major step forward, these drugs do not halt or reverse the disease, nor convincingly improve health-related QOL. In daily practice, disease behavior and response to therapy greatly vary among patients. It is assumed that this is related to the multiple biological pathways and complex interactions between genetic, molecular, and environmental factors that are involved in the pathogenesis of IPF. Recently, research in IPF has therefore started to focus on developing targeted therapy through identifying genetic risk factors and biomarkers. In this rapidly evolving field of personalized medicine, patient factors such as lifestyle, comorbidities, preferences, and experiences with medication should not be overlooked. This review describes recent insights and methods on how to integrate patient perspectives into personalized medicine. Furthermore, it provides an overview of the most used patient-reported outcome measures in IPF, to facilitate choices for both researchers and clinicians when incorporating the patient voice in their research and care. To enhance truly personalized treatment in IPF, biology should be combined with patient perspectives.
Collapse
Affiliation(s)
- Catharina C. Moor
- Department of Respiratory Medicine, Erasmus Medical Center, University Hospital Rotterdam, Rotterdam, Netherlands
| | - Peter Heukels
- Department of Respiratory Medicine, Erasmus Medical Center, University Hospital Rotterdam, Rotterdam, Netherlands
| | - Mirjam Kool
- Department of Respiratory Medicine, Erasmus Medical Center, University Hospital Rotterdam, Rotterdam, Netherlands
| | - Marlies S. Wijsenbeek
- Department of Respiratory Medicine, Erasmus Medical Center, University Hospital Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
187
|
Kaner RJ, Brown KK, Martinez FJ. AJRCCM: 100-Year Anniversary. Progress in Interstitial Lung Disease. Am J Respir Crit Care Med 2017; 195:1104-1107. [PMID: 28459330 DOI: 10.1164/rccm.201703-0584ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Robert J Kaner
- 1 Joan and Sanford I. Weill Department of Medicine Weill Cornell Medicine New York, New York.,2 Department of Genetic Medicine Weill Cornell Medicine New York, New York and
| | - Kevin K Brown
- 3 Department of Medicine National Jewish Health Denver, Colordao
| | - Fernando J Martinez
- 1 Joan and Sanford I. Weill Department of Medicine Weill Cornell Medicine New York, New York
| |
Collapse
|
188
|
Kropski JA, Young LR, Cogan JD, Mitchell DB, Lancaster LH, Worrell JA, Markin C, Liu N, Mason WR, Fingerlin TE, Schwartz DA, Lawson WE, Blackwell TS, Phillips JA, Loyd JE. Genetic Evaluation and Testing of Patients and Families with Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2017; 195:1423-1428. [PMID: 27786550 DOI: 10.1164/rccm.201609-1820pp] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Jonathan A Kropski
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Lisa R Young
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine.,2 Division of Pulmonary Medicine and
| | - Joy D Cogan
- 3 Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, and
| | - Daphne B Mitchell
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Lisa H Lancaster
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - John A Worrell
- 4 Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cheryl Markin
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Na Liu
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Wendi R Mason
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Tasha E Fingerlin
- 5 Department of Biomedical Research, National Jewish Health, Denver, Colorado
| | - David A Schwartz
- 6 Department of Medicine, University of Colorado, Denver, Colorado
| | - William E Lawson
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine.,7 Department of Veterans Affairs Medical Center, Nashville, Tennessee; and
| | - Timothy S Blackwell
- 7 Department of Veterans Affairs Medical Center, Nashville, Tennessee; and.,8 Department of Cancer Biology.,9 Department of Cell and Developmental Biology, and
| | - John A Phillips
- 3 Division of Medical Genetics and Genomic Medicine, Department of Pediatrics, and.,10 Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James E Loyd
- 1 Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| |
Collapse
|
189
|
Ryu C, Homer RJ, Herzog EL. The Airway in Idiopathic Pulmonary Fibrosis: Protecting the Lung or Promoting Disease? Am J Respir Crit Care Med 2017; 193:1081-2. [PMID: 27174477 DOI: 10.1164/rccm.201601-0055ed] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Changwan Ryu
- 1 Department of Internal Medicine Yale University School of Medicine New Haven, Connecticut and
| | - Robert J Homer
- 2 Department of Pathology Yale University School of Medicine New Haven, Connecticut
| | - Erica L Herzog
- 1 Department of Internal Medicine Yale University School of Medicine New Haven, Connecticut and
| |
Collapse
|
190
|
Thomson NC. Targeting oxidant-dependent mechanisms for the treatment of respiratory diseases and their comorbidities. Curr Opin Pharmacol 2017; 40:1-8. [PMID: 29223018 DOI: 10.1016/j.coph.2017.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 11/24/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
Abstract
Oxidative stress is implicated in the pathogenesis of respiratory diseases, such as COPD and its comorbidities, asthma, idiopathic pulmonary fibrosis and radiation pneumonitis. Antioxidants drugs, such as small molecule thiols, nuclear erythroid-2 related factor 2 activators and catalytic enzyme mimetics have been developed to target oxidant-dependent mechanisms. The therapeutic effects of antioxidants have been generally disappointing. A small number of antioxidants are approved for clinical use, such as the small molecule thiol N-acetyl-l-cysteine for chronic obstructive pulmonary disease, and in the United States, the superoxide dismutase mimetic AEOL 10150 for severe radiation pneumonitis. The future use of antioxidants for the treatment of chronic respiratory diseases may require a precision medicine approach to identify responsive patients.
Collapse
Affiliation(s)
- Neil C Thomson
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK.
| |
Collapse
|
191
|
Abstract
PURPOSE OF REVIEW Interstitial lung disease (ILD) is comprised of a heterogeneous group of disorders with highly variable natural histories and response to therapies. Pharmacogenetics focuses on the variability in drug response because of the presence of genetic factors that influence drug metabolism or disease activity. In this article, we review relevant drug-specific and disease-specific polymorphisms that may influence therapeutic response, and then highlight a recently identified drug-gene interaction in patients with idiopathic pulmonary fibrosis (IPF). RECENT FINDINGS The emergence of high-throughput genomic technology has allowed for identification of gene polymorphisms associated with susceptibility to specific disease states, including IPF and several connective tissue diseases known to cause ILD. IPF risk loci span a diverse group of genes, while most associated with connective tissue disease are critical to immune signaling. A recent pharmacogenetic analysis of patients enrolled in an IPF clinical trial identified a variant within TOLLIP to be associated with differential response to N-acetylcysteine therapy. SUMMARY Though few pharmacogenetic investigations have been conducted in patients with ILD to date, ample opportunities for pharmacogenetic exploration exist in this patient population. Such exploration will advance our understanding of specific ILDs and help usher in an era of personalized medicine.
Collapse
|
192
|
Mora AL, Rojas M, Pardo A, Selman M. Emerging therapies for idiopathic pulmonary fibrosis, a progressive age-related disease. Nat Rev Drug Discov 2017; 16:810. [PMID: 29081515 DOI: 10.1038/nrd.2017.225] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
193
|
Raghu G. Idiopathic pulmonary fibrosis: lessons from clinical trials over the past 25 years. Eur Respir J 2017; 50:50/4/1701209. [PMID: 29074545 DOI: 10.1183/13993003.01209-2017] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/23/2017] [Indexed: 01/24/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal disease. A major breakthrough in treatment came when, after decades of clinical trials which failed to identify an efficacious treatment regimen, two therapies were successful in Phase-III trials. The advent of these therapies, nintedanib and pirfenidone, meant that for the first time IPF patients had two treatment options that could reduce disease progression. This review summarises the key lessons to be obtained from the clinical trials that led to the current international clinical practice guidelines for the treatment of IPF and provides insights for the design of future clinical trials that are needed if we are to improve outcomes that are clinically meaningful to IPF patients.
Collapse
Affiliation(s)
- Ganesh Raghu
- Center for Interstitial Lung Diseases, University of Washington, Seattle, WA, USA
| |
Collapse
|
194
|
Mora AL, Rojas M, Pardo A, Selman M. Emerging therapies for idiopathic pulmonary fibrosis, a progressive age-related disease. Nat Rev Drug Discov 2017; 16:755-772. [DOI: 10.1038/nrd.2017.170] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
195
|
Cottin V, Crestani B, Cadranel J, Cordier JF, Marchand-Adam S, Prévot G, Wallaert B, Bergot E, Camus P, Dalphin JC, Dromer C, Gomez E, Israel-Biet D, Jouneau S, Kessler R, Marquette CH, Reynaud-Gaubert M, Aguilaniu B, Bonnet D, Carré P, Danel C, Faivre JB, Ferretti G, Just N, Lebargy F, Philippe B, Terrioux P, Thivolet-Béjui F, Trumbic B, Valeyre D. French practical guidelines for the diagnosis and management of idiopathic pulmonary fibrosis – 2017 update. Full-length version. Rev Mal Respir 2017; 34:900-968. [DOI: 10.1016/j.rmr.2017.07.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
196
|
Adegunsoye A, Oldham JM, Chung JH, Montner SM, Lee C, Witt LJ, Stahlbaum D, Bermea RS, Chen LW, Hsu S, Husain AN, Noth I, Vij R, Strek ME, Churpek M. Phenotypic Clusters Predict Outcomes in a Longitudinal Interstitial Lung Disease Cohort. Chest 2017; 153:349-360. [PMID: 28964798 DOI: 10.1016/j.chest.2017.09.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/06/2017] [Accepted: 09/11/2017] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The current interstitial lung disease (ILD) classification has overlapping clinical presentations and outcomes. Cluster analysis modeling is a valuable tool in identifying distinct clinical phenotypes in heterogeneous diseases. However, this approach has yet to be implemented in ILD. METHODS Using cluster analysis, novel ILD phenotypes were identified among subjects from a longitudinal ILD cohort, and outcomes were stratified according to phenotypic clusters compared with subgroups according to current American Thoracic Society/European Respiratory Society ILD classification criteria. RESULTS Among subjects with complete data for baseline variables (N = 770), four clusters were identified. Cluster 1 (ie, younger white obese female subjects) had the highest baseline FVC and diffusion capacity of the lung for carbon monoxide (Dlco). Cluster 2 (ie, younger African-American female subjects with elevated antinuclear antibody titers) had the lowest baseline FVC. Cluster 3 (ie, elderly white male smokers with coexistent emphysema) had intermediate FVC and Dlco. Cluster 4 (ie, elderly white male smokers with severe honeycombing) had the lowest baseline Dlco. Compared with classification according to ILD subgroup, stratification according to phenotypic clusters was associated with significant differences in monthly FVC decline (Cluster 4, -0.30% vs Cluster 2, 0.01%; P < .0001). Stratification by using clusters also independently predicted progression-free survival (P < .001) and transplant-free survival (P < .001). CONCLUSIONS Among adults with diverse chronic ILDs, cluster analysis using baseline characteristics identified four distinct clinical phenotypes that might better predict meaningful clinical outcomes than current ILD diagnostic criteria.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL.
| | - Justin M Oldham
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of California at Davis, Davis, CA
| | | | | | - Cathryn Lee
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL
| | - Leah J Witt
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL
| | | | - Rene S Bermea
- Department of Medicine, University of Chicago, Chicago, IL
| | - Lena W Chen
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL
| | - Scully Hsu
- Department of Medicine, University of Chicago, Chicago, IL
| | - Aliya N Husain
- Department of Pathology, University of Chicago, Chicago, IL
| | - Imre Noth
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL
| | - Rekha Vij
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL
| | - Mary E Strek
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL
| | - Matthew Churpek
- Section of Pulmonary & Critical Care, Department of Medicine, University of Chicago, Chicago, IL
| |
Collapse
|
197
|
Greiffo FR, Eickelberg O, Fernandez IE. Systems medicine advances in interstitial lung disease. Eur Respir Rev 2017; 26:26/145/170021. [PMID: 28954764 DOI: 10.1183/16000617.0021-2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/15/2017] [Indexed: 01/17/2023] Open
Abstract
Fibrotic lung diseases involve subject-environment interactions, together with dysregulated homeostatic processes, impaired DNA repair and distorted immune functions. Systems medicine-based approaches are used to analyse diseases in a holistic manner, by integrating systems biology platforms along with clinical parameters, for the purpose of understanding disease origin, progression, exacerbation and remission.Interstitial lung diseases (ILDs) refer to a heterogeneous group of complex fibrotic diseases. The increase of systems medicine-based approaches in the understanding of ILDs provides exceptional advantages by improving diagnostics, unravelling phenotypical differences, and stratifying patient populations by predictable outcomes and personalised treatments. This review discusses the state-of-the-art contributions of systems medicine-based approaches in ILDs over the past 5 years.
Collapse
Affiliation(s)
- Flavia R Greiffo
- Comprehensive Pneumology Center, Ludwig-Maximilians-Universität, University Hospital Grosshadern and Helmholtz Zentrum München and Member of the German Center for Lung Research, Munich, Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, Ludwig-Maximilians-Universität, University Hospital Grosshadern and Helmholtz Zentrum München and Member of the German Center for Lung Research, Munich, Germany.,Division of Respiratory Sciences and Critical Care Medicine, Dept of Medicine, University of Colorado, Denver, CO, USA
| | - Isis E Fernandez
- Comprehensive Pneumology Center, Ludwig-Maximilians-Universität, University Hospital Grosshadern and Helmholtz Zentrum München and Member of the German Center for Lung Research, Munich, Germany
| |
Collapse
|
198
|
Raghu G. Pharmacotherapy for idiopathic pulmonary fibrosis: current landscape and future potential. Eur Respir Rev 2017; 26:26/145/170071. [PMID: 28954769 PMCID: PMC9488742 DOI: 10.1183/16000617.0071-2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/12/2017] [Indexed: 11/30/2022] Open
Abstract
Over the past two and a half decades, many clinical trials have been designed to determine the safety and efficacy of pharmacotherapy for patients with idiopathic pulmonary fibrosis (IPF). However, so far, only two drugs (pirfenidone and nintedanib) have been found to have an impact on disease progression as defined by reducing the rate of decline in forced vital capacity over a year among IPF patients with mild to moderate impairment in lung function. These two drugs have been approved for treatment of IPF by regulatory agencies and are currently in clinical use worldwide. This article summarises the current landscape of pharmacotherapy for IPF and highlights the prospects and potential of new therapies that are currently being pursued in clinical trials. The landscape of treatment for IPF is on new turf with nintedanib, pirfenidone and new clinical trialshttp://ow.ly/bav230eQPgl
Collapse
Affiliation(s)
- Ganesh Raghu
- Center for Interstitial Lung Diseases, University of Washington Medical Center, Division of Pulmonary, Critical Care and Sleep Medicine, Seattle, WA, USA
| |
Collapse
|
199
|
Kaur A, Mathai SK, Schwartz DA. Genetics in Idiopathic Pulmonary Fibrosis Pathogenesis, Prognosis, and Treatment. Front Med (Lausanne) 2017; 4:154. [PMID: 28993806 PMCID: PMC5622313 DOI: 10.3389/fmed.2017.00154] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), the most common form of idiopathic interstitial pneumonia (IIP), is characterized by irreversible scarring of the lung parenchyma and progressive decline in lung function leading to eventual respiratory failure. The prognosis of IPF is poor with a median survival of 3–5 years after diagnosis and no curative medical therapies. Although the pathogenesis of IPF is not well understood, there is a growing body of evidence that genetic factors contribute to disease risk. Recent studies have identified common and rare genetic variants associated with both sporadic and familial forms of pulmonary fibrosis, with at least one-third of the risk for developing fibrotic IIP explained by common genetic variants. The IPF-associated genetic loci discovered to date are implicated in diverse biological processes, including alveolar stability, host defense, cell–cell barrier function, and cell senescence. In addition, some common variants have also been associated with distinct clinical phenotypes. Better understanding of how genetic variation plays a role in disease risk and phenotype could identify potential therapeutic targets and inform clinical decision-making. In addition, clinical studies should be designed controlling for the genetic backgrounds of subjects, since clinical outcomes and therapeutic responses may differ by genotype. Further understanding of these differences will allow the development of personalized approaches to the IPF management.
Collapse
Affiliation(s)
- Amarpreet Kaur
- Department of Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States
| | - Susan K Mathai
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States
| | - David A Schwartz
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver School of Medicine, Aurora, CO, United States
| |
Collapse
|
200
|
Cottin V, Crestani B, Cadranel J, Cordier JF, Marchand-Adam S, Prévot G, Wallaert B, Bergot E, Camus P, Dalphin JC, Dromer C, Gomez E, Israel-Biet D, Jouneau S, Kessler R, Marquette CH, Reynaud-Gaubert M, Aguilaniu B, Bonnet D, Carré P, Danel C, Faivre JB, Ferretti G, Just N, Lebargy F, Philippe B, Terrioux P, Thivolet-Béjui F, Trumbic B, Valeyre D. [French practical guidelines for the diagnosis and management of idiopathic pulmonary fibrosis. 2017 update. Full-length update]. Rev Mal Respir 2017:S0761-8425(17)30209-7. [PMID: 28943227 DOI: 10.1016/j.rmr.2017.07.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- V Cottin
- Centre national de référence des maladies pulmonaires rares, pneumologie, hôpital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard-Lyon 1, Lyon, France.
| | - B Crestani
- Service de pneumologie A, centre de compétences pour les maladies pulmonaires rares, CHU Bichat, université Paris Diderot, Paris, France
| | - J Cadranel
- Service de pneumologie et oncologie thoracique, centre de compétences pour les maladies pulmonaires rares, hôpital Tenon, université Pierre-et-Marie-Curie, Paris 6, GH-HUEP, Assistance publique-Hôpitaux de Paris, Paris, France
| | - J-F Cordier
- Centre national de référence des maladies pulmonaires rares, pneumologie, hôpital Louis-Pradel, hospices civils de Lyon, université Claude-Bernard-Lyon 1, Lyon, France
| | - S Marchand-Adam
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU de Tours, Tours, France
| | - G Prévot
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU Larrey, Toulouse, France
| | - B Wallaert
- Service de pneumologie et immuno-allergologie, centre de compétences pour les maladies pulmonaires rares, hôpital Calmette, CHRU de Lille, Lille, France
| | - E Bergot
- Service de pneumologie et oncologie thoracique, centre de compétences pour les maladies pulmonaires rares, CHU de Caen, Caen, France
| | - P Camus
- Service de pneumologie et oncologie thoracique, centre de compétences pour les maladies pulmonaires rares, CHU Dijon-Bourgogne, Dijon, France
| | - J-C Dalphin
- Service de pneumologie, allergologie et oncologie thoracique, centre de compétences pour les maladies pulmonaires rares, hôpital Jean-Minjoz, CHRU de Besançon, Besançon, France
| | - C Dromer
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, hôpital Haut-Lévèque, CHU de Bordeaux, Bordeaux, France
| | - E Gomez
- Département de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU de Nancy, Vandœuvre-lès-Nancy, France
| | - D Israel-Biet
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, hôpital européen Georges-Pompidou, université Paris-Descartes, Paris, France
| | - S Jouneau
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU de Rennes, IRSET UMR 1085, université de Rennes 1, Rennes, France
| | - R Kessler
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, hôpital civil, CHU de Strasbourg, Strasbourg, France
| | - C-H Marquette
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, CHU de Nice, FHU Oncoage, université Côte d'Azur, France
| | - M Reynaud-Gaubert
- Service de pneumologie, centre de compétence des maladies pulmonaires rares, CHU Nord, Marseille, France
| | | | - D Bonnet
- Service de pneumologie, centre hospitalier de la Côte-Basque, Bayonne, France
| | - P Carré
- Service de pneumologie, centre hospitalier, Carcassonne, France
| | - C Danel
- Département de pathologie, hôpital Bichat-Claude-Bernard, université Paris Diderot, Assistance publique-Hôpitaux de Paris, Paris 7, Paris, France
| | - J-B Faivre
- Service d'imagerie thoracique, hôpital Calmette, CHRU de Lille, Lille, France
| | - G Ferretti
- Clinique universitaire de radiologie et imagerie médicale, CHU Grenoble-Alpes, Grenoble, France
| | - N Just
- Service de pneumologie, centre hospitalier Victor-Provo, Roubaix, France
| | - F Lebargy
- Service des maladies respiratoires, CHU Maison-Blanche, Reims, France
| | - B Philippe
- Service de pneumologie, centre hospitalier René-Dubos, Pontoise, France
| | - P Terrioux
- Service de pneumologie, centre hospitalier de Meaux, Meaux, France
| | - F Thivolet-Béjui
- Service d'anatomie et cytologie pathologiques, hôpital Louis-Pradel, Lyon, France
| | | | - D Valeyre
- Service de pneumologie, centre de compétences pour les maladies pulmonaires rares, hôpital Avicenne, CHU Paris-Seine-Saint-Denis, Bobigny, France
| |
Collapse
|