101
|
The Role of Microbiome and Virome in Idiopathic Pulmonary Fibrosis. Biomedicines 2021; 9:biomedicines9040442. [PMID: 33924195 PMCID: PMC8074588 DOI: 10.3390/biomedicines9040442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/23/2022] Open
Abstract
The interest in the lung microbiome and virome and their contribution to the pathogenesis, perpetuation and progression of idiopathic pulmonary fibrosis (IPF) has been increasing during the last decade. The utilization of high-throughput sequencing to detect microbial and/or viral genetic material in bronchoalveolar lavage fluid or lung tissue samples has amplified the ability to identify and quantify specific microbial and viral populations. In stable IPF, higher microbial burden is associated with worse prognosis but no specific microbe has been identified to contribute to this. Additionally, no causative relation has been established. Regarding viral infections, although in the past they have been associated with IPF, causation has not been proved. Although in the past the diagnosis of acute exacerbation of IPF (AE-IPF) was not considered in patients with overt infection, this was amended in the last few years and infection is considered a cause for exacerbation. Besides this, a higher microbial burden has been found in the lungs of patients with AE-IPF and an association with higher morbidity and mortality has been confirmed. In contrast, an association of AE-IPF with viral infection has not been established. Despite the progress during the last decade, a comprehensive knowledge of the microbiome and virome in IPF and their role in disease pathogenesis are yet elusive. Although association with disease severity, risk for progression and mortality has been established, causation has not been proven and the potential use as a biomarker or the benefits of antimicrobial therapeutic strategies are yet to be determined.
Collapse
|
102
|
Oxidative Stress and Endoplasmic Reticulum Stress in Rare Respiratory Diseases. J Clin Med 2021; 10:jcm10061268. [PMID: 33803835 PMCID: PMC8003245 DOI: 10.3390/jcm10061268] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Several studies have shown that some rare respiratory diseases, such as alpha-1 antitrypsin deficiency (AATD), idiopathic pulmonary fibrosis (IPF), cystic fibrosis (CF), and primary ciliary dyskinesia (PCD) present oxidative stress (OS) and endoplasmic reticulum (ER) stress. Their involvement in these pathologies and the use of antioxidants as therapeutic agents to minimize the effects of OS are discussed in this review.
Collapse
|
103
|
Calverley P, Rogliani P, Papi A. Safety of N-Acetylcysteine at High Doses in Chronic Respiratory Diseases: A Review. Drug Saf 2021; 44:273-290. [PMID: 33326056 PMCID: PMC7892733 DOI: 10.1007/s40264-020-01026-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2020] [Indexed: 12/14/2022]
Abstract
N-Acetylcysteine (NAC) is widely used in respiratory medicine, with a maximum licensed dose in chronic use of 600 mg/day; however, some clinical trials have studied the efficacy of NAC at higher doses. The aim of this review was to evaluate the adverse effects profile of NAC at higher than the standard dose in chronic respiratory diseases to establish a risk-benefit ratio in increasing the daily dose; therefore, studies using NAC at a dose of at least 600 mg/day were selected. Forty-one articles where NAC has been used at 600 mg and above, up to 3000 mg/day, and with a specific report on safety, were considered. Most of the studies used oral NAC and were conducted on patients with chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, bronchiectasis, chronic bronchitis and cystic fibrosis. In general, the safety profile was similar at both the high and standard doses with the oral formulation; gastrointestinal symptoms were reported but they were no more common than in the control group.
Collapse
Affiliation(s)
- Peter Calverley
- School of Aging and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Paola Rogliani
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Alberto Papi
- Section of Cardiorespiratory and Internal Medicine, Department of Medical Sciences, Research Centre on Asthma and COPD, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
104
|
Bonella F, Campo I, Zorzetto M, Boerner E, Ohshimo S, Theegarten D, Taube C, Costabel U. Potential clinical utility of MUC5B und TOLLIP single nucleotide polymorphisms (SNPs) in the management of patients with IPF. Orphanet J Rare Dis 2021; 16:111. [PMID: 33639995 PMCID: PMC7913255 DOI: 10.1186/s13023-021-01750-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/16/2021] [Indexed: 11/26/2022] Open
Abstract
Background Genetic variants of TOLLIP and MUC5B, both on chromosome 11, have been reported to be associated with the development and/or prognosis of idiopathic pulmonary fibrosis (IPF). This retrospective study was conducted to investigate the association of MUC5B and TOLLIP SNPs with disease outcome in IPF. 62 IPF patients and 50 healthy controls (HC) from our Institution were genotyped for SNPs within MUC5B (rs35705950) and TOLLIP (rs3750920 and rs5743890). Correlation of SNPs genotypes with survival, acute exacerbation (AE) or disease progression (defined as a decline of ≥ 5% in FVC and or ≥ 10% in DLco in one year) was investigated. Results The MUC5B rs35705950 minor allele (T) was more frequent in IPF subjects than in HC (35% vs 9% p < 0.001). TOLLIP SNPs alleles and genotype distribution did not differ between IPF and HC and did not vary according to gender, age, BMI and lung functional impairment at baseline. The minor allele (C) in TOLLIP rs5743890 was associated with worse survival and with disease progression in all performed analyses. The MUC5B rs35705950 or the TOLLIP rs3750920 minor allele, were not associated with disease progression or AE.
Conclusion We confirm that the minor allele of MUC5B rs35705950 is associated with IPF. The minor allele of TOLLIP rs5743890 appears to be a predictor of worse survival and more rapid disease progression, therefore being of potential utility to stratify IPF patients at baseline.
Collapse
Affiliation(s)
- Francesco Bonella
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik University Hospital, University of Duisburg-Essen, 45239, Essen, Germany.
| | - Ilaria Campo
- SC Pneumologia - Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Michele Zorzetto
- Clinical Chemistry Laboratory, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Eda Boerner
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik University Hospital, University of Duisburg-Essen, 45239, Essen, Germany
| | - Shinichiro Ohshimo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Dirk Theegarten
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christian Taube
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik University Hospital, University of Duisburg-Essen, 45239, Essen, Germany
| | - Ulrich Costabel
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik University Hospital, University of Duisburg-Essen, 45239, Essen, Germany
| |
Collapse
|
105
|
Antoniou KM, Tsitoura E, Vasarmidi E, Symvoulakis EK, Aidinis V, Tzilas V, Tzouvelekis A, Bouros D. Precision medicine in idiopathic pulmonary fibrosis therapy: From translational research to patient-centered care. Curr Opin Pharmacol 2021; 57:71-80. [PMID: 33556824 DOI: 10.1016/j.coph.2020.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, irreversible fibrotic chronic lung disease affecting predominantly older adults, with a history of smoking. The current model of disease natural course is that recurrent injury of the alveolar epithelium in the context of advanced aging/cellular senescence is followed by defective re-epithelialization and scar tissue formation. Currently, two drugs, nintedanib and pirfenidone, that modify disease progression have been approved worldwide for the treatment of IPF. However, despite treatment, patients with IPF are not cured, and eventually, disease advances in most treated patients. Enhancing biogenomic and metabolic research output, its translation into clinical precision and optimal service delivery through patient-centeredness are key elements to support effective IPF care. In this review, we summarize therapeutic options currently investigated for IPF based on the major pathogenetic pathways and molecular targets that drive pulmonary fibrosis.
Collapse
Affiliation(s)
- Katerina M Antoniou
- Molecular & Cellular Pneumonology Laboratory, Department of Respiratory Medicine, Faculty of Medicine, University of Crete, Greece.
| | - Eliza Tsitoura
- Molecular & Cellular Pneumonology Laboratory, Department of Respiratory Medicine, Faculty of Medicine, University of Crete, Greece
| | - Eirini Vasarmidi
- Molecular & Cellular Pneumonology Laboratory, Department of Respiratory Medicine, Faculty of Medicine, University of Crete, Greece
| | | | - Vassilis Aidinis
- Division of Immunology, Alexander Fleming Biomedical Sciences Research Center, Athens, Greece
| | - Vassilis Tzilas
- Center for Diseases of the Chest, Athens Medical Center, Athens, Greece
| | | | - Demosthenes Bouros
- Center for Diseases of the Chest, Athens Medical Center, Athens, Greece; Medical School, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
106
|
Sgalla G, Lerede M, Richeldi L. Phase three clinical trials in idiopathic pulmonary fibrosis. Expert Opin Orphan Drugs 2021. [DOI: 10.1080/21678707.2021.1882299] [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: 10/22/2022]
Affiliation(s)
- Giacomo Sgalla
- Dipartimento Scienze Gastroenterologiche, Endocrino-Metaboliche E Nefro-Urologiche, Unità Operativa Complessa Di Pneumologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - Marialessia Lerede
- Dipartimento Scienze Gastroenterologiche, Endocrino-Metaboliche E Nefro-Urologiche, Unità Operativa Complessa Di Pneumologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
| | - Luca Richeldi
- Dipartimento Scienze Gastroenterologiche, Endocrino-Metaboliche E Nefro-Urologiche, Unità Operativa Complessa Di Pneumologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome, Italy
- Istituto di Medicina Interna, Università Cattolica Del Sacro Cuore, Rome, Italy
| |
Collapse
|
107
|
Cutting CC, Bowman WS, Dao N, Pugashetti JV, Garcia CK, Oldham JM, Newton CA. Family History of Pulmonary Fibrosis Predicts Worse Survival in Patients With Interstitial Lung Disease. Chest 2021; 159:1913-1921. [PMID: 33484728 DOI: 10.1016/j.chest.2021.01.026] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND A number of genetic markers linked to familial pulmonary fibrosis predict differential survival in interstitial lung disease (ILD) patients. Although genetic testing is not performed routinely for ILD, family history commonly is obtained and may inform outcome risk. RESEARCH QUESTION Does survival vary between patients with and without self-reported familial pulmonary fibrosis? METHODS Family history was acquired systematically for consecutive ILD patients who consented to clinical registry enrollment at the University of Texas Southwestern and the University of California at Davis. Patients were stratified by idiopathic pulmonary fibrosis (IPF) and non-IPF ILD diagnosis and were substratified by presence or absence of familial pulmonary fibrosis, defined as one or more additional affected family members. Transplant-free survival was compared using multilevel, mixed-effects Cox proportional hazards regression. RESULTS Of the 1,262 patients included, 534 (42%) had IPF ILD and 728 (58%) had non-IPF ILD. Of those with non-IPF ILD, 18.5% had connective tissue disease, 15.6% had chronic hypersensitivity pneumonitis, and 23.5% had unclassifiable ILD. Familial pulmonary fibrosis was reported in 134 IPF ILD patients (25.1%) and 90 non-IPF ILD patients (12.4%). Those with familial IPF showed an 80% increased risk of death or transplantation compared with those with sporadic IPF (hazard ratio [HR], 1.8; 95% CI, 1.37-2.37; P < .001), whereas those with familial non-IPF ILD showed a twofold increased risk compared with their counterparts with sporadic disease (HR, 2.08; 95% CI, 1.46-2.96; P < .001). Outcome risk among those with familial non-IPF ILD was no different than for those with sporadic IPF ILD (HR, 1.27; 95% CI, 0.89-1.84; P = .19). INTERPRETATION Patient-reported familial pulmonary fibrosis is predictive of reduced transplant-free survival in IPF and non-IPF ILD patients. Because survival among patients with familial non-IPF ILD approximates that of sporadic IPF ILD, early intervention should be considered for such patients. Until clinical genetic testing is widely available and provides actionable results, family history should be ascertained and considered in risk stratification.
Collapse
Affiliation(s)
- Claire C Cutting
- Department of Internal Medicine, University of California at Davis, Sacramento, CA.
| | - Willis S Bowman
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Nam Dao
- Department of Internal Medicine, University of California at Davis, Sacramento, CA
| | - Janelle Vu Pugashetti
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Christine Kim Garcia
- Medicine within the Department of Medicine at Columbia University, College of Physicians and Surgeons, New York, NY
| | - Justin M Oldham
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California at Davis, Sacramento, CA
| | - Chad A Newton
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
108
|
Podolanczuk AJ, Noth I, Raghu G. Idiopathic pulmonary fibrosis: prime time for a precision-based approach to treatment with N-acetylcysteine. Eur Respir J 2021; 57:57/1/2003551. [PMID: 33402374 DOI: 10.1183/13993003.03551-2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 11/05/2022]
Affiliation(s)
| | - Imre Noth
- Dept of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Ganesh Raghu
- Dept of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
109
|
Sakamoto S, Kataoka K, Kondoh Y, Kato M, Okamoto M, Mukae H, Bando M, Suda T, Yatera K, Tanino Y, Kishaba T, Hattori N, Taguchi Y, Saito T, Nishioka Y, Kuwano K, Kishi K, Inase N, Sasaki S, Takizawa H, Johkoh T, Sakai F, Homma S. Pirfenidone plus inhaled N-acetylcysteine for idiopathic pulmonary fibrosis: a randomised trial. Eur Respir J 2021; 57:13993003.00348-2020. [PMID: 32703779 DOI: 10.1183/13993003.00348-2020] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/16/2020] [Indexed: 11/05/2022]
Abstract
BACKGROUND A randomised controlled trial in Japan showed that inhaled N-acetylcysteine monotherapy stabilised serial decline in forced vital capacity (FVC) in some patients with early idiopathic pulmonary fibrosis (IPF). However, the efficacy and tolerability of combination therapy with an antifibrotic agent and inhaled N-acetylcysteine are unknown. METHODS This 48-week, randomised, open-label, multicentre phase 3 trial compared the efficacy and tolerability of combination therapy with pirfenidone plus inhaled N-acetylcysteine 352.4 mg twice daily with the results for pirfenidone alone in patients with IPF. The primary end-point was annual rate of decline in FVC. Exploratory efficacy measurements included serial change in diffusing capacity of the lung for carbon monoxide (D LCO) and 6-min walk distance (6MWD), progression-free survival (PFS), incidence of acute exacerbation, and tolerability. RESULTS 81 patients were randomly assigned in a 1:1 ratio to receive pirfenidone plus inhaled N-acetylcysteine (n=41) or pirfenidone (n=40). The 48-week rate of change in FVC was -300 mL and -123 mL, respectively (difference -178 mL, 95% CI -324--31 mL; p=0.018). Serial change in D LCO, 6MWD, PFS and incidence of acute exacerbation did not significantly differ between the two groups. The incidence of adverse events (n=19 (55.9%) for pirfenidone plus N-acetylcysteine; n=18 (50%) for pirfenidone alone) was similar between groups. CONCLUSIONS Combination treatment with inhaled N-acetylcysteine and pirfenidone is likely to result in worse outcomes for IPF.
Collapse
Affiliation(s)
- Susumu Sakamoto
- Dept of Respiratory Medicine, Toho University Omori Medical Center, Tokyo, Japan
| | - Kensuke Kataoka
- Tosei General Hospital, Dept of Respiratory Medicine and Allergy, Seto, Japan
| | - Yasuhiro Kondoh
- Tosei General Hospital, Dept of Respiratory Medicine and Allergy, Seto, Japan
| | - Motoyasu Kato
- Dept of Respiratory Medicine, Juntendo University, Graduate School of Medicine, Tokyo, Japan
| | - Masaki Okamoto
- Dept of Internal Medicine, Division of Respirology, Neurology, and Rheumatology, Kurume University School of Medicine, Fukuoka, Japan
| | - Hiroshi Mukae
- Second Dept of Internal Medicine, Nagasaki University, Graduate School of Biomedical Sciences, Fukuoka, Japan
| | - Masashi Bando
- Division of Pulmonary Medicine, Dept of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takafumi Suda
- Internal Medicine 2, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhiro Yatera
- Dept of Respiratory Medicine, University of Occupational and Environmental Health, Japan, Fukuoka, Japan
| | - Yoshinori Tanino
- Dept of Pulmonary Medicine, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Tomoo Kishaba
- Dept of Respiratory Medicine, Okinawa Chubu Hospital, Okinawa, Japan
| | - Noboru Hattori
- Dept of Molecular and Internal Medicine, Hiroshima University, Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yoshio Taguchi
- Dept of Respiratory Medicine, Tenri Yorozu Hospital, Nara, Japan
| | - Takefumi Saito
- Dept of Respiratory Medicine, Ibarakihigashi National Hospital, Ibaragi, Japan
| | - Yasuhiko Nishioka
- Dept of Respiratory Medicine and Rheumatology, Tokushima University Graduate School of Medical Sciences, Tokushima, Japan
| | - Kazuyoshi Kuwano
- Division of Respiratory Diseases, Dept of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Kazuma Kishi
- Dept of Respiratory Medicine, Toho University Omori Medical Center, Tokyo, Japan.,Dept of Respiratory Medicine, Toranomon Hospital, Tokyo, Japan
| | - Naohiko Inase
- Dept of Respiratory Medicine, Tokyo Medical and Dental University, Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Shinichi Sasaki
- Dept of Respiratory Medicine, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Hajime Takizawa
- Dept of Respiratory Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | | | - Fumikazu Sakai
- Dept of Diagnostic Radiology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Sakae Homma
- Dept of Respiratory Medicine, Toho University Omori Medical Center, Tokyo, Japan.,Dept of Advanced and Integrated Interstitial Lung Diseases Research, School of Medicine, Toho University, Tokyo, Japan
| | | |
Collapse
|
110
|
Hamanaka RB, Mutlu GM. Metabolic requirements of pulmonary fibrosis: role of fibroblast metabolism. FEBS J 2021; 288:6331-6352. [PMID: 33393204 DOI: 10.1111/febs.15693] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/22/2020] [Accepted: 12/31/2020] [Indexed: 12/27/2022]
Abstract
Fibrosis is a pathologic condition characterized by excessive deposition of extracellular matrix and chronic scaring that can affect every organ system. Organ fibrosis is associated with significant morbidity and mortality, contributing to as many as 45% of all deaths in the developed world. In the lung, many chronic lung diseases may lead to fibrosis, the most devastating being idiopathic pulmonary fibrosis (IPF), which affects approximately 3 million people worldwide and has a median survival of 3.8 years. Currently approved therapies for IPF do not significantly extend lifespan, and thus, there is pressing need for novel therapeutic strategies to treat IPF and other fibrotic diseases. At the heart of pulmonary fibrosis are myofibroblasts, contractile cells with characteristics of both fibroblasts and smooth muscle cells, which are the primary cell type responsible for matrix deposition in fibrotic diseases. Much work has centered around targeting the extracellular growth factors and intracellular signaling regulators of myofibroblast differentiation. Recently, metabolic changes associated with myofibroblast differentiation have come to the fore as targetable mechanisms required for myofibroblast function. In this review, we will discuss the metabolic changes associated with myofibroblast differentiation, as well as the mechanisms by which these changes promote myofibroblast function. We will then discuss the potential for this new knowledge to lead to the development of novel therapies for IPF and other fibrotic diseases.
Collapse
Affiliation(s)
- Robert B Hamanaka
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, IL, USA
| | - Gökhan M Mutlu
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, IL, USA
| |
Collapse
|
111
|
Macrophage metabolic reprogramming during chronic lung disease. Mucosal Immunol 2021; 14:282-295. [PMID: 33184475 PMCID: PMC7658438 DOI: 10.1038/s41385-020-00356-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/13/2020] [Accepted: 10/24/2020] [Indexed: 02/04/2023]
Abstract
Airway macrophages (AMs) play key roles in the maintenance of lung immune tolerance. Tissue tailored, highly specialised and strategically positioned, AMs are critical sentinels of lung homoeostasis. In the last decade, there has been a revolution in our understanding of how metabolism underlies key macrophage functions. While these initial observations were made during steady state or using in vitro polarised macrophages, recent studies have indicated that during many chronic lung diseases (CLDs), AMs adapt their metabolic profile to fit their local niche. By generating reactive oxygen species (ROS) for pathogen defence, utilising aerobic glycolysis to rapidly generate cytokines, and employing mitochondrial respiration to fuel inflammatory responses, AMs utilise metabolic reprogramming for host defence, although these changes may also support chronic pathology. This review focuses on how metabolic alterations underlie AM phenotype and function during CLDs. Particular emphasis is given to how our new understanding of AM metabolic plasticity may be exploited to develop AM-focused therapies.
Collapse
|
112
|
Raghu G, Berk M, Campochiaro PA, Jaeschke H, Marenzi G, Richeldi L, Wen FQ, Nicoletti F, Calverley PMA. The Multifaceted Therapeutic Role of N-Acetylcysteine (NAC) in Disorders Characterized by Oxidative Stress. Curr Neuropharmacol 2021; 19:1202-1224. [PMID: 33380301 PMCID: PMC8719286 DOI: 10.2174/1570159x19666201230144109] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/27/2020] [Accepted: 12/13/2020] [Indexed: 02/08/2023] Open
Abstract
Oxidative stress, which results in the damage of diverse biological molecules, is a ubiquitous cellular process implicated in the etiology of many illnesses. The sulfhydryl-containing tripeptide glutathione (GSH), which is synthesized and maintained at high concentrations in all cells, is one of the mechanisms by which cells protect themselves from oxidative stress. N-acetylcysteine (NAC), a synthetic derivative of the endogenous amino acid L-cysteine and a precursor of GSH, has been used for several decades as a mucolytic and as an antidote to acetaminophen (paracetamol) poisoning. As a mucolytic, NAC breaks the disulfide bonds of heavily cross-linked mucins, thereby reducing mucus viscosity. In vitro, NAC has antifibrotic effects on lung fibroblasts. As an antidote to acetaminophen poisoning, NAC restores the hepatic GSH pool depleted in the drug detoxification process. More recently, improved knowledge of the mechanisms by which NAC acts has expanded its clinical applications. In particular, the discovery that NAC can modulate the homeostasis of glutamate has prompted studies of NAC in neuropsychiatric diseases characterized by impaired glutamate homeostasis. This narrative review provides an overview of the most relevant and recent evidence on the clinical application of NAC, with a focus on respiratory diseases, acetaminophen poisoning, disorders of the central nervous system (chronic neuropathic pain, depression, schizophrenia, bipolar disorder, and addiction), cardiovascular disease, contrast-induced nephropathy, and ophthalmology (retinitis pigmentosa).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Peter M. A. Calverley
- Address correspondence to this author at Clinical Science Centre, University Hospital Aintree, Longmoor Lane, Liverpool UK L9 7AL; Tel: +44 151 529 5886, Fax: +44 151 529 5888; E-mail:
| |
Collapse
|
113
|
p53: A Key Protein That Regulates Pulmonary Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6635794. [PMID: 33312337 PMCID: PMC7721501 DOI: 10.1155/2020/6635794] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/05/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Pulmonary fibrosis is a progressively aggravating lethal disease that is a serious public health concern. Although the incidence of this disease is increasing, there is a lack of effective therapies. In recent years, the pathogenesis of pulmonary fibrosis has become a research hotspot. p53 is a tumor suppressor gene with crucial roles in cell cycle, apoptosis, tumorigenesis, and malignant transformation. Previous studies on p53 have predominantly focused on its role in neoplastic disease. Following in-depth investigation, several studies have linked it to pulmonary fibrosis. This review covers the association between p53 and pulmonary fibrosis, with the aim of providing novel ideas to improve the clinical diagnosis, treatment, and prognosis of pulmonary fibrosis.
Collapse
|
114
|
Rackow AR, Nagel DJ, McCarthy C, Judge J, Lacy S, Freeberg MAT, Thatcher TH, Kottmann RM, Sime PJ. The self-fulfilling prophecy of pulmonary fibrosis: a selective inspection of pathological signalling loops. Eur Respir J 2020; 56:13993003.00075-2020. [PMID: 32943406 PMCID: PMC7931159 DOI: 10.1183/13993003.00075-2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/01/2020] [Indexed: 12/28/2022]
Abstract
Pulmonary fibrosis is a devastating, progressive disease and carries a prognosis worse than most cancers. Despite ongoing research, the mechanisms that underlie disease pathogenesis remain only partially understood. However, the self-perpetuating nature of pulmonary fibrosis has led several researchers to propose the existence of pathological signalling loops. According to this hypothesis, the normal wound-healing process becomes corrupted and results in the progressive accumulation of scar tissue in the lung. In addition, several negative regulators of pulmonary fibrosis are downregulated and, therefore, are no longer capable of inhibiting these feed-forward loops. The combination of pathological signalling loops and loss of a checks and balances system ultimately culminates in a process of unregulated scar formation. This review details specific signalling pathways demonstrated to play a role in the pathogenesis of pulmonary fibrosis. The evidence of detrimental signalling loops is elucidated with regard to epithelial cell injury, cellular senescence and the activation of developmental and ageing pathways. We demonstrate where these loops intersect each other, as well as common mediators that may drive these responses and how the loss of pro-resolving mediators may contribute to the propagation of disease. By focusing on the overlapping signalling mediators among the many pro-fibrotic pathways, it is our hope that the pulmonary fibrosis community will be better equipped to design future trials that incorporate the redundant nature of these pathways as we move towards finding a cure for this unrelenting disease.
Collapse
Affiliation(s)
- Ashley R Rackow
- Dept of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Authors contributed equally to this work
| | - David J Nagel
- Division of Pulmonary Diseases and Critical Care, University of Rochester Medical Center, Rochester, NY, USA.,Authors contributed equally to this work
| | | | | | - Shannon Lacy
- US Army of Veterinary Corps, Fort Campbell, KY, USA
| | | | - Thomas H Thatcher
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - R Matthew Kottmann
- Division of Pulmonary Diseases and Critical Care, University of Rochester Medical Center, Rochester, NY, USA
| | - Patricia J Sime
- Division of Pulmonary Diseases and Critical Care, University of Rochester Medical Center, Rochester, NY, USA.,Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
115
|
The Potential Effects of Curcumin on Pulmonary Fibroblasts of Idiopathic Pulmonary Fibrosis (IPF)-Approaching with Next-Generation Sequencing and Bioinformatics. Molecules 2020; 25:molecules25225458. [PMID: 33233354 PMCID: PMC7700625 DOI: 10.3390/molecules25225458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive interstitial lung disease. Currently, therapeutic options are limited for this fatal disease. Curcumin, with its pleiotropic effects, has been studied for its potential therapeutic utilities in various diseases, including pulmonary fibrosis. However, the detailed mechanisms have not been studied comprehensively. We conducted a next-generation sequencing and bioinformatics study to investigate changes in the profiles of mRNA and microRNA after curcumin treatment in IPF fibroblasts. We identified 23 downregulated and 8 upregulated protein-coding genes in curcumin-treated IPF fibroblasts. Using STRING and IPA, we identified that suppression of cell cycle progression was the main cellular function associated with these differentially expressed genes. We also identified 13 downregulated and 57 upregulated microRNAs in curcumin-treated IPF fibroblasts. Further analysis identified a potential microRNA-mediated gene expression alteration in curcumin-treated IPF fibroblasts, namely, downregulated hsa-miR-6724-5p and upregulated KLF10. Therefore, curcumin might decrease the level of hsa-miR-6724-5p, leading to increased KLF10 expression, resulting in cell cycle arrest in curcumin-treated IPF fibroblasts. In conclusion, our findings might support the potential role of curcumin in the treatment of IPF, but further in-depth study is warranted to confirm our findings.
Collapse
|
116
|
Verleden SE, Braubach P, Kuehnel M, Dickgreber N, Brouwer E, Tittmann P, Laenger F, Jonigk D. Molecular approach to the classification of chronic fibrosing lung disease-there and back again. Virchows Arch 2020; 478:89-99. [PMID: 33169196 DOI: 10.1007/s00428-020-02964-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/21/2020] [Accepted: 10/30/2020] [Indexed: 11/26/2022]
Abstract
Chronic diffuse parenchymal lung disease (DPLD) is an umbrella term for a very heterogeneous group of lung diseases. Over the last decades, clinical, radiological and histopathological criteria have been established to define and separate these entities. More recently the clinical utility of this approach has been challenged as a unifying concept of pathophysiological mechanisms and a shared response to therapy across the disease spectrum have been described. In this review, we discuss molecular motifs for subtyping and the prediction of prognosis focusing on genetics and markers found in the blood, lavage and tissue. As a purely molecular classification so far lacks sufficient sensitivity and specificity for subtyping, it is not routinely used and not implemented in international guidelines. However, a better molecular characterization of lung disease with a more precise identification of patients with, for example, a risk for rapid disease progression would facilitate more accurate treatment decisions and hopefully contribute to better patients' outcomes.
Collapse
Affiliation(s)
- Stijn E Verleden
- Institute of Pathology, Hannover Medical School, Hannover, Germany.
- BREATHE Lab, Department of CHROMETA, KU Leuven, Leuven, Belgium.
| | - Peter Braubach
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Mark Kuehnel
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Nicolas Dickgreber
- Department of Respiratory Medicine and Thoracic Oncology, Ibbenbueren General Hospital, Ibbenbueren, Germany
| | - Emily Brouwer
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Pauline Tittmann
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Florian Laenger
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research, Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), Hannover, Germany
| |
Collapse
|
117
|
Li X, Goobie GC, Zhang Y. Toll-interacting protein impacts on inflammation, autophagy, and vacuole trafficking in human disease. J Mol Med (Berl) 2020; 99:21-31. [PMID: 33128579 DOI: 10.1007/s00109-020-01999-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/30/2020] [Accepted: 10/23/2020] [Indexed: 02/08/2023]
Abstract
Toll-interacting protein (TOLLIP) is a ubiquitous intracellular adaptor protein involved in multiple intracellular signaling pathways. It plays a key role in mediating inflammatory intracellular responses, promoting autophagy, and enabling vacuole transport within the cell. TOLLIP is being increasingly recognized for its role in disease pathophysiology through involvement in these three primary pathways. Recent research also indicates that TOLLIP is involved in nuclear-cytoplasmic transfer, although this area requires further exploration. TOLLIP is involved in the pathophysiologic pathways associated with neurodegenerative diseases, pulmonary diseases, cardiovascular disease, inflammatory bowel disease, and malignancy. We postulate that TOLLIP plays an integral role in the disease pathophysiology of other conditions involved in vacuole trafficking and autophagy. We suggest that future research in this field should investigate the role of TOLLIP in the pathogenesis of these multiple conditions. This research has the potential to inform disease mechanisms and identify novel opportunities for therapeutic advances in multiple disease processes.
Collapse
Affiliation(s)
- Xiaoyun Li
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Gillian C Goobie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Clinician Investigator Program, Department of Medicine, University of British Columbia, BC, V5Z-3X7, Vancouver, Canada
| | - Yingze Zhang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
118
|
Feng H, Zhao Y, Li Z, Kang J. Real-life experiences in a single center: efficacy of pirfenidone in idiopathic pulmonary fibrosis and fibrotic idiopathic non-specific interstitial pneumonia patients. Ther Adv Respir Dis 2020; 14:1753466620963015. [PMID: 33070705 PMCID: PMC7580189 DOI: 10.1177/1753466620963015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background: Pirfenidone is the first antifibrotic drug approved for the treatment of idiopathic pulmonary fibrosis (IPF) and it is used in the treatment of other interstitial pneumonias, such as unclassifiable interstitial lung disease (ILD) and connective tissue-related ILD. This study examined the efficacy of pirfenidone in patients with IPF and fibrotic idiopathic non-specific interstitial pneumonia (f-iNSIP). Methods: In a retrospective real-life study, 67 IPF and 24 f-iNSIP patients were enrolled and classified into a pirfenidone group and a non-antifibrotic group. The level of forced vital capacity (FVC) and diffusing capacity of lung for carbon monoxide (DLco) at baseline, 6, 12 and 24 months were recorded. The level of KL-6 in serum was detected by chemiluminescence enzyme immunoassay (CLEIA). The prognosis and safety outcomes were collected from patients. Results: In IPF patients, pirfenidone decreased the mean change of FVC and DLco, and decreased the proportion of patients with a ⩾10% decline in FVC or a ⩾15% decline in DLco compared with the non-antifibrotic group. There was no difference in the mean change of FVC and DLco between smokers and non-smokers who received pirfenidone treatment. There was an improvement in progression-free survival, defined as the time to the first occurrence of acute exacerbation or death related to pulmonary fibrosis. Moreover, the ratio of patients who experienced acute exacerbation and death related to pulmonary fibrosis was lower in the pirfenidone group. There was no change in lung function and prognosis between the pirfenidone and non-antifibrotic groups in f-iNSIP patients. The KL-6 level slightly decreased after 1 year of pirfenidone treatment but not significantly. Gastrointestinal and skin-related adverse events were most common, and four patients ceased treatment due to the side effects. Conclusions: Pirfenidone safely reduced disease progression by improving the lung function and progression-free survival in IPF patients, with acceptable side effects. The efficacy of pirfenidone on f-iNSIP was not significant, suggesting the need for further studies. The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Haoshen Feng
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, PR China
| | - Yabin Zhao
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, the First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, Liaoning 110001, China
| | - Zhenhua Li
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Jian Kang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| |
Collapse
|
119
|
Paudel KR, Dharwal V, Patel VK, Galvao I, Wadhwa R, Malyla V, Shen SS, Budden KF, Hansbro NG, Vaughan A, Yang IA, Kohonen-Corish MRJ, Bebawy M, Dua K, Hansbro PM. Role of Lung Microbiome in Innate Immune Response Associated With Chronic Lung Diseases. Front Med (Lausanne) 2020; 7:554. [PMID: 33043031 PMCID: PMC7530186 DOI: 10.3389/fmed.2020.00554] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
Respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), lung fibrosis, and lung cancer, pose a huge socio-economic burden on society and are one of the leading causes of death worldwide. In the past, culture-dependent techniques could not detect bacteria in the lungs, therefore the lungs were considered a sterile environment. However, the development of culture-independent techniques, particularly 16S rRNA sequencing, allowed for the detection of commensal microbes in the lung and with further investigation, their roles in disease have since emerged. In healthy individuals, the predominant commensal microbes are of phylum Firmicutes and Bacteroidetes, including those of the genera Veillonella and Prevotella. In contrast, pathogenic microbes (Haemophilus, Streptococcus, Klebsiella, Pseudomonas) are often associated with lung diseases. There is growing evidence that microbial metabolites, structural components, and toxins from pathogenic and opportunistic bacteria have the capacity to stimulate both innate and adaptive immune responses, and therefore can contribute to the pathogenesis of lung diseases. Here we review the multiple mechanisms that are altered by pathogenic microbiomes in asthma, COPD, lung cancer, and lung fibrosis. Furthermore, we focus on the recent exciting advancements in therapies that can be used to restore altered microbiomes in the lungs.
Collapse
Affiliation(s)
- Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Vivek Dharwal
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Vyoma K Patel
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Izabela Galvao
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Ridhima Wadhwa
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Vamshikrishna Malyla
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Sj Sijie Shen
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Kurtis F Budden
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Annalicia Vaughan
- Faculty of Medicine, Thoracic Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Ian A Yang
- Faculty of Medicine, Thoracic Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Maija R J Kohonen-Corish
- Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.,School of Medicine, Western Sydney University, Sydney, NSW, Australia.,St George and Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Mary Bebawy
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Kamal Dua
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
120
|
Fan Y, He R, Zou L, Meng J. [Clinical value of biomarkers in diagnosis and treatment of idiopathic pulmonary fibrosis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1062-1065. [PMID: 32895164 DOI: 10.12122/j.issn.1673-4254.2020.07.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial pneumonia characterized by progressive accumulation of fibroblastic foci and destruction of the alveolar structure. Due to an incomplete understanding of the mechanism of the occurrence and progression of IPF, currently no effective means have been available for its early screening or treatment. With a poor overall prognosis, the patients with IPF have a median survival of only 2-4 years. In recent years, several studies have confirmed that dozens of molecules are involved in the development of IPF and can be used as potential biomarkers. These biomarkers play important roles in early diagnosis (such as SP-D, MMP-7, and osteopontin), prognostic evaluation (such as telomerase length, KL-6, mtDNA, HSP-70, LOXL2, CXCL13, miRNA, ICAM-1, and CCL18), and guiding treatment of IPF (such as TOLLIP rs3750920 genotype, SAMS score, and SP-D), and also provide potential therapeutic targets (such as TERT, TERR, RTEC, and PARN).
Collapse
Affiliation(s)
- Yubin Fan
- Department of Respiratory and Critical Care Medicine, Xiangya Hospital; Organ Fibrosis Research Center, Central South University, Changsha 410008, China
| | - Rongling He
- Department of Respiratory and Critical Care Medicine, Xiangya Hospital; Organ Fibrosis Research Center, Central South University, Changsha 410008, China
| | - Lijun Zou
- Department of Respiratory and Critical Care Medicine, Xiangya Hospital; Organ Fibrosis Research Center, Central South University, Changsha 410008, China
| | - Jie Meng
- Department of Respiratory and Critical Care Medicine, Xiangya Hospital; Organ Fibrosis Research Center, Central South University, Changsha 410008, China
| |
Collapse
|
121
|
Sarmah P, Bharali R, Khatonier R, Khan A. Polymorphism in Toll interacting protein (TOLLIP) gene and its association with Visceral Leishmaniasis. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
122
|
Zhu T, Ma Z, Wang H, Jia X, Wu Y, Fu L, Li Z, Zhang C, Yu G. YAP/TAZ affects the development of pulmonary fibrosis by regulating multiple signaling pathways. Mol Cell Biochem 2020; 475:137-149. [PMID: 32813142 DOI: 10.1007/s11010-020-03866-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022]
Abstract
YAP and TAZ are important co-activators of various biological processes in human body. YAP/TAZ plays a vital role in the development of pulmonary fibrosis. Dysregulation of the YAP/TAZ signaling pathway is one of the most important causes of pulmonary fibrosis. Therefore, considering its crucial role, summary of the signal mechanism of YAP/TAZ is of certain guiding significance for the research of YAP/TAZ as a therapeutic target. The present review provided a detailed introduction to various YAP/TAZ-related signaling pathways and clarified the specific role of YAP/TAZ in these pathways. In the meantime, we summarized and evaluated possible applications of YAP/TAZ in the treatment of pulmonary fibrosis. Overall, our study is of guiding significance for future research on the functional mechanism of YAP/TAZ underlying lung diseases as well as for identification of novel therapeutic targets specific to pulmonary fibrosis.
Collapse
Affiliation(s)
- Ting Zhu
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Zhifeng Ma
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Haiyong Wang
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Xiaoxiao Jia
- Department of Pathology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, China
| | - Yuanlin Wu
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Linhai Fu
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Zhupeng Li
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China
| | - Chu Zhang
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China.
| | - Guangmao Yu
- Department of Thoracic Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Yuecheng District, Shaoxing, 312000, China.
| |
Collapse
|
123
|
Kim KJ, Moon SJ, Park KS, Tagkopoulos I. Network-based modeling of drug effects on disease module in systemic sclerosis. Sci Rep 2020; 10:13393. [PMID: 32770109 PMCID: PMC7414841 DOI: 10.1038/s41598-020-70280-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/10/2020] [Indexed: 01/13/2023] Open
Abstract
The network-based proximity between drug targets and disease genes can provide novel insights regarding the repercussions, interplay, and repositioning of drugs in the context of disease. Current understanding and treatment for reversing of the fibrotic process is limited in systemic sclerosis (SSc). We have developed a network-based analysis for drug effects that takes into account the human interactome network, proximity measures between drug targets and disease-associated genes, genome-wide gene expression and disease modules that emerge through pertinent analysis. Currently used and potential drugs showed a wide variation in proximity to SSc-associated genes and distinctive proximity to the SSc-relevant pathways, depending on their class and targets. Tyrosine kinase inhibitors (TyKIs) approach disease gene through multiple pathways, including both inflammatory and fibrosing processes. The SSc disease module includes the emerging molecular targets and is in better accord with the current knowledge of the pathophysiology of the disease. In the disease-module network, the greatest perturbing activity was shown by nintedanib, followed by imatinib, dasatinib, and acetylcysteine. Suppression of the SSc-relevant pathways and alleviation of the skin fibrosis was remarkable in the inflammatory subsets of the SSc patients receiving TyKI therapy. Our results show that network-based drug-disease proximity offers a novel perspective into a drug’s therapeutic effect in the SSc disease module. This could be applied to drug combinations or drug repositioning, and be helpful guiding clinical trial design and subgroup analysis.
Collapse
Affiliation(s)
- Ki-Jo Kim
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. .,St. Vincent's Hospital, 93 Jungbu-daero, Paldal-gu, Suwon, Gyeonggi-do, 16247, Republic of Korea.
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyung-Su Park
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ilias Tagkopoulos
- Department of Computer Science, University of California, Davis, CA, USA. .,Genome Center, University of California, Davis, CA, USA. .,AI Institute for Next-Generation Food Systems, AIFS, Davis, CA, USA.
| |
Collapse
|
124
|
Miles T, Hoyne GF, Knight DA, Fear MW, Mutsaers SE, Prêle CM. The contribution of animal models to understanding the role of the immune system in human idiopathic pulmonary fibrosis. Clin Transl Immunology 2020; 9:e1153. [PMID: 32742653 PMCID: PMC7385431 DOI: 10.1002/cti2.1153] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/21/2020] [Accepted: 06/15/2020] [Indexed: 12/21/2022] Open
Abstract
Pulmonary fibrosis occurs in a heterogeneous group of lung disorders and is characterised by an excessive deposition of extracellular matrix proteins within the pulmonary interstitium, leading to impaired gas transfer and a loss of lung function. In the past 10 years, there has been a dramatic increase in our understanding of the immune system and how it contributes to fibrogenic processes within the lung. This review will compare some of the models used to investigate the pathogenesis and treatment of pulmonary fibrosis, in particular those used to study immune cell pathogenicity in idiopathic pulmonary fibrosis, highlighting their advantages and disadvantages in dissecting human disease.
Collapse
Affiliation(s)
- Tylah Miles
- Institute for Respiratory Health Nedlands WA Australia.,Centre for Respiratory Health School of Biomedical Sciences University of Western Australia Nedlands WA Australia
| | - Gerard F Hoyne
- Centre for Cell Therapy and Regenerative Medicine School of Biomedical Sciences University of Western Australia Nedlands WA Australia.,School of Health Sciences University of Notre Dame Australia Fremantle WA Australia
| | - Darryl A Knight
- Providence Health Care Research Institute Vancouver BC Canada.,University of British Columbia Vancouver BC Canada
| | - Mark W Fear
- Burn Injury Research Unit School of Biomedical Sciences The University of Western Australia Crawley WA Australia
| | - Steven E Mutsaers
- Institute for Respiratory Health Nedlands WA Australia.,Centre for Respiratory Health School of Biomedical Sciences University of Western Australia Nedlands WA Australia.,Centre for Cell Therapy and Regenerative Medicine School of Biomedical Sciences University of Western Australia Nedlands WA Australia
| | - Cecilia M Prêle
- Centre for Respiratory Health School of Biomedical Sciences University of Western Australia Nedlands WA Australia.,Centre for Cell Therapy and Regenerative Medicine School of Biomedical Sciences University of Western Australia Nedlands WA Australia.,Ear Science Institute Australia Nedlands WA Australia
| |
Collapse
|
125
|
Singh S, Sharma BB, Bairwa M, Gothi D, Desai U, Joshi JM, Talwar D, Singh A, Dhar R, Sharma A, Ahluwalia B, Mangal DK, Jain NK, Pilania K, Hadda V, Koul PA, Luhadia SK, Swarnkar R, Gaur SN, Ghoshal AG, Nene A, Jindal A, Jankharia B, Ravindran C, Choudhary D, Behera D, Christopher DJ, Khilnani GC, Samaria JK, Singh H, Gupta KB, Pilania M, Gupta ML, Misra N, Singh N, Gupta PR, Chhajed PN, Kumar R, Chawla R, Jenaw RK, Chawla R, Guleria R, Agarwal R, Narsimhan R, Katiyar S, Mehta S, Dhooria S, Chowdhury SR, Jindal SK, Katiyar SK, Chaudhri S, Gupta N, Singh S, Kant S, Udwadia ZF, Singh V, Raghu G. Management of interstitial lung diseases: A consensus statement of the Indian Chest Society (ICS) and National College of Chest Physicians (NCCP). Lung India 2020; 37:359-378. [PMID: 32643655 PMCID: PMC7507933 DOI: 10.4103/lungindia.lungindia_275_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 04/29/2020] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Interstitial lung disease (ILD) is a complex and heterogeneous group of acute and chronic lung diseases of several known and unknown causes. While clinical practice guidelines (CPG) for idiopathic pulmonary fibrosis (IPF) have been recently updated, CPG for ILD other than IPF are needed. METHODS A working group of multidisciplinary clinicians familiar with clinical management of ILD (pulmonologists, radiologist, pathologist, and rheumatologist) and three epidemiologists selected by the leaderships of Indian Chest Society and National College of Chest Physicians, India, posed questions to address the clinically relevant situation. A systematic search was performed on PubMed, Embase, and Cochrane databases. A modified GRADE approach was used to grade the evidence. The working group discussed the evidence and reached a consensus of opinions for each question following face-to-face discussions. RESULTS Statements have been made for each specific question and the grade of evidence has been provided after performing a systematic review of literature. For most of the questions addressed, the available evidence was insufficient and of low to very low quality. The consensus of the opinions of the working group has been presented as statements for the questions and not as an evidence-based CPG for the management of ILD. CONCLUSION This document provides the guidelines made by consensus of opinions among experts following discussion of systematic review of evidence pertaining to the specific questions for management of ILD other than IPF. It is hoped that this document will help the clinician understand the accumulated evidence and help better management of idiopathic and nonidiopathic interstitial pneumonias.
Collapse
Affiliation(s)
- Sheetu Singh
- Department of Respiratory Medicine, SMS Medical College, Jaipur, Rajasthan, India
| | | | - Mohan Bairwa
- Centre for Community Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Dipti Gothi
- Department of Pulmonary, Sleep and Critical Care Medicine, ESI-PGIMSR, Delhi, India
| | - Unnati Desai
- Department of Pulmonary Medicine, TNMC and BYL Nair Hospital, Mumbai, Maharashtra, India
| | - Jyotsna M Joshi
- Department of Pulmonary Medicine, TNMC and BYL Nair Hospital, Mumbai, Maharashtra, India
| | - Deepak Talwar
- Division of Pulmonary and Critical Care Medicine, Metro Centre for Respiratory Diseases, Metro Multi Speciality Hospital, Noida, Uttar Pradesh, India
| | - Abhijeet Singh
- Division of Pulmonary and Critical Care Medicine, Metro Centre for Respiratory Diseases, Metro Multi Speciality Hospital, Noida, Uttar Pradesh, India
| | - Raja Dhar
- Department of Pulmonology, Fortis Hospital, Kolkata, West Bengal, India
| | - Ambika Sharma
- Department of Respiratory Medicine, SMS Medical College, Jaipur, Rajasthan, India
| | - Bineet Ahluwalia
- Department of Respiratory Medicine, SMS Medical College, Jaipur, Rajasthan, India
| | - Daya K Mangal
- Department of Public Health and Epidemiology, IIHMR University, Jaipur, Rajasthan, India
| | | | - Khushboo Pilania
- Department of Radio Diagnosis, Max Super Specialty Hospital, Noida, Uttar Pradesh, India
| | - Vijay Hadda
- Department of Pulmonary, Critical Care and Sleep Medicine, AIIMS, New Delhi, India
| | - Parvaiz A Koul
- Department of Internal and Pulmonary Medicine, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| | - Shanti Kumar Luhadia
- Department of Respiratory Medicine, Geetanjali Medical College and Hospital, Udaipur, Rajasthan, India
| | - Rajesh Swarnkar
- Department of Respiratory, Critical Care, Sleep and Interventional Pulmonology, Getwell Hospital and Research Institute, Nagpur, Maharashtra, India
| | - Shailender Nath Gaur
- Department of Respiratory Medicine, School of Medical Science and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Aloke G Ghoshal
- Department of Respiratory Medicine, National Allergy Asthma Bronchitis Institute, Kolkata, West Bengal, India
| | - Amita Nene
- Department of Respiratory Medicine, Bombay Hospital and Medical Research Center, Mumbai, Maharashtra, India
| | - Arpita Jindal
- Department of Pathology, SMS Medical College, Jaipur, Rajasthan, India
| | - Bhavin Jankharia
- Department of Radiodiagnosis, Jankharia Imaging, Mumbai, Maharashtra, India
| | - Chetambath Ravindran
- Department of Pulmonary Medicine, DM Wayanad Institute of Medical Sciences, Wayanad, Kerala, India
| | - Dhruv Choudhary
- Department of Pulmonary and Critical Care Medicine, Pt. B.D.S PGIMS, Rohtak, Haryana, India
| | | | - DJ Christopher
- Department of Pulmonary Medicine, Christian Medical College, Vellore, Tamil Nadu, India
| | - Gopi C Khilnani
- Department of Pulmonary Medicine, PSRI, Institute of Pulmonary, Critical Care and Sleep Medicine, New Delhi, India
| | - Jai Kumar Samaria
- Department of Chest Diseases, Institute of Medical Sciences, BHU, Varanasi, Uttar Pradesh, India
| | | | | | - Manju Pilania
- Department of Community Medicine, RUHS College of Medical Sciences, Jaipur, Rajasthan, India
| | - Manohar L Gupta
- Department of Pulmonary and Sleep Medicine, Santokba Durlabhji Memorial Hospital, Jaipur, Rajasthan, India
| | - Narayan Misra
- Department of Pulmonary Medicine, MKCG Medical College and Hospital, Brahmapur, Odisha, India
| | - Nishtha Singh
- Department of Pulmonary Medicine, Asthma Bhawan, Jaipur, Rajasthan, India
| | - Prahlad R Gupta
- Department of Pulmonary Medicine, NIMS University, Jaipur, Rajasthan, India
| | - Prashant N. Chhajed
- Lung Care and Sleep Center, Institute of Pulmonology Medical Research and Development, Mumbai, Maharashtra, India
| | - Raj Kumar
- Department of Respiratory Medicine, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Rajesh Chawla
- Department of Respiratory Medicine, Critical Care and Sleep Disorders, Indraprastha Apollo Hospitals, New Delhi, India
| | - Rajendra K Jenaw
- Department of Respiratory Medicine, SMS Medical College, Jaipur, Rajasthan, India
| | - Rakesh Chawla
- Department of Respiratory Medicine, Critical Care and Sleep disorders, Jaipur Golden Hospital and Saroj Superspeciality Hospital, Delhi, India
| | - Randeep Guleria
- Department of Pulmonary, Critical Care and Sleep Medicine, AIIMS, New Delhi, India
| | - Ritesh Agarwal
- Department of Pulmonary Medicine, PGIMER, Chandigarh, India
| | - R Narsimhan
- Department of Respiratory Medicine, Apollo Hospitals, Chennai, Tamil Nadu, India
| | - Sandeep Katiyar
- Department of Pulmonary Medicine, Apollo Spectra Hospital, Kanpur, Uttar Pradesh, India
| | - Sanjeev Mehta
- Department of Pulmonology, The Chest and Allergy Center, Mumbai, Maharashtra, India
| | | | - Sushmita R Chowdhury
- Department of Pulmonary Medicine, Apollo Gleneagles Hospital, Kolkata, West Bengal, India
| | | | | | - Sudhir Chaudhri
- Department of Respiratory Medicine, GSVM Medical College and Hospital, Kanpur, Uttar Pradesh, India
| | - Neeraj Gupta
- Department of Respiratory Medicine, JLN Medical College & Hospital, Ajmer, India
| | - Sunita Singh
- Department of Pathology, PGIMS, Rohtak (Haryana), KGMU, Lucknow, Uttar Pradesh, India
| | - Surya Kant
- Department of Respiratory Medicine, KG Medical University, Lucknow (Uttar Pradesh), India
| | - Zarir F. Udwadia
- Department of Pulmonary Medicine, Hinduja Hospital, Mumbai (Maharashtra), India
| | - Virendra Singh
- Department of Pulmonary Medicine, Asthma Bhawan, Jaipur, Rajasthan, India
| | - Ganesh Raghu
- Department of Medicine, University of Washington, Seattle, USA
| |
Collapse
|
126
|
Advances in Targeted Therapy for Progressive Fibrosing Interstitial Lung Disease. Lung 2020; 198:597-608. [PMID: 32591895 DOI: 10.1007/s00408-020-00370-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/17/2020] [Indexed: 12/25/2022]
Abstract
Progressive fibrosing interstitial lung disease (PF-ILD) has been redefined as a new clinical syndrome that shares similar genetics, pathophysiology, and natural history to idiopathic pulmonary fibrosis (IPF). IPF is the most common form of idiopathic interstitial pneumonias, which is progressive in nature and is associated with significant mortality. Therapies targeting an inflammatory and/or immune response have not been consistently effective or well tolerated in patients with IPF. The two antifibrotic drugs approved for IPF treatment, nintedanib and pirfenidone, have been shown to reduce lung function decline in PF-ILD. Novel uses of antifibrotic therapy are emerging due to a paucity of evidence-based treatments for multiple ILD subtypes. In this review, we describe the current body of knowledge on antifibrotic therapy and immunomodulators in PF-ILD, drawing from experience in IPF where appropriate.
Collapse
|
127
|
Caliskan C, Seeliger B, Jäger B, Fuge J, Welte T, Terwolbeck O, Freise J, van Moorsel CHM, Zhang Y, Prasse A. Genetic Variation in CCL18 Gene Influences CCL18 Expression and Correlates with Survival in Idiopathic Pulmonary Fibrosis-Part B. J Clin Med 2020; 9:jcm9061993. [PMID: 32630441 PMCID: PMC7356861 DOI: 10.3390/jcm9061993] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/03/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease with high mortality. CC-chemokine ligand 18 (CCL18) is predictive of survival in IPF. We described correlation of CCL18 serum levels with the genotype of rs2015086 C > T polymorphism the CCL18-gene, which was associated with survival in a pre-antifibrotic cohort (Part-A). Herein (Part-B), we aimed to validate these findings and to study the effects of antifibrotics. Two cohorts were prospectively recruited, cohort-A (n = 61, pre-antifibrotic) and cohort B (n = 101, received antifibrotics). Baseline CCL18 serum level measurement by enzyme-linked immunosorbent assay (ELISA, serially in cohort B) and genotyping of rs2015086 was performed and correlated with clinical outcomes. The CT genotype was present in 15% and 31% of patients. These patients had higher CCL18 levels compared to the TT-genotype (cohort-A: 234 vs. 115.8 ng/mL, p < 0.001; cohort B: 159.5 vs. 120 ng/mL, p = 0.0001). During antifibrotic therapy, CCL18 increased (p = 0.0036) regardless of rs2015086-genotype and antifibrotic-agent. In cohort-A, baseline CCL18-cutoff (>120 ng/mL) and CT-genotype were associated with mortality (p = 0.041 and p = 0.0051). In cohort-B, the CCL18-cutoff (>140 ng/mL) was associated with mortality (p = 0.003) and progression (p = 0.004), but not the CT/CC-genotype. In conclusion, we validated the correlation between rs2015086-genotype and CCL18 serum levels, which was predictive of (progression-free)-survival in two prospective validation cohorts.
Collapse
Affiliation(s)
- Canay Caliskan
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), 30265 Hannover, Germany; (C.C.); (B.S.); (J.F.); (T.W.); (J.F.)
| | - Benjamin Seeliger
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), 30265 Hannover, Germany; (C.C.); (B.S.); (J.F.); (T.W.); (J.F.)
| | - Benedikt Jäger
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany; (B.J.); (O.T.)
| | - Jan Fuge
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), 30265 Hannover, Germany; (C.C.); (B.S.); (J.F.); (T.W.); (J.F.)
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), 30265 Hannover, Germany; (C.C.); (B.S.); (J.F.); (T.W.); (J.F.)
| | - Oliver Terwolbeck
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany; (B.J.); (O.T.)
| | - Julia Freise
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), 30265 Hannover, Germany; (C.C.); (B.S.); (J.F.); (T.W.); (J.F.)
| | - Coline H. M. van Moorsel
- Dept of Pulmonology, Interstitial Lung Diseases Center of Excellence, St Antonius Hospital, 3435 CM Nieuwegein, The Netherlands;
| | - Yingze Zhang
- Department of Medicine and Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Antje Prasse
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), 30265 Hannover, Germany; (C.C.); (B.S.); (J.F.); (T.W.); (J.F.)
- Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany; (B.J.); (O.T.)
- Correspondence: ; Tel.: +49-(0)-511-532-3530
| |
Collapse
|
128
|
Torrisi SE, Kahn N, Vancheri C, Kreuter M. Evolution and treatment of idiopathic pulmonary fibrosis. Presse Med 2020; 49:104025. [PMID: 32437841 DOI: 10.1016/j.lpm.2020.104025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 01/02/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and devastating disease of unknown etiology, characterized by irreversible morphological changes, ultimately leading to lung fibrosis and death. In recent years, significant progress has been achieved in understanding the pathogenesis of IPF. Moreover, we assisted to the conceptual change of the pathogenic hypothesis that currently considers IPF as a primarily fibrotic driven disease. However, despite the undeniable progress, the diagnosis of IPF remains still very complex requiring the presence of a team of experts to achieve the highest level of diagnostic confidence. The advent of antifibrotics has radically changed the treatment landscape of IPF and new promising drugs are currently under evaluation. Furthermore, a more extensive use of non-pharmacological treatments has also to be encouraged in all patients both to reduce symptoms and improve quality of life.
Collapse
Affiliation(s)
- Sebastiano Emanuele Torrisi
- Center for interstitial and rare lung diseases, Pneumology and respiratory critical care medicine, Thoraxklinik, University of Heidelberg, and Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany; Regional Referral Centre for Rare Lung Diseases, University Hospital "Policlinico", Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Nicolas Kahn
- Center for interstitial and rare lung diseases, Pneumology and respiratory critical care medicine, Thoraxklinik, University of Heidelberg, and Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Carlo Vancheri
- Regional Referral Centre for Rare Lung Diseases, University Hospital "Policlinico", Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Michael Kreuter
- Center for interstitial and rare lung diseases, Pneumology and respiratory critical care medicine, Thoraxklinik, University of Heidelberg, and Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research (DZL), Heidelberg, Germany.
| |
Collapse
|
129
|
Borie R, Kannengiesser C, Dupin C, Debray MP, Cazes A, Crestani B. Impact of genetic factors on fibrosing interstitial lung diseases. Incidence and clinical presentation in adults. Presse Med 2020; 49:104024. [PMID: 32437840 DOI: 10.1016/j.lpm.2020.104024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
At least 10% of patients with pulmonary fibrosis, whether idiopathic or secondary, present heritable pulmonary fibrosis suspected on familial aggregation of pulmonary fibrosis, specific syndromes or early age of diagnosis. Approximately 30% of those patients have an identified mutation mostly in telomere related genes (TRG) more rarely in surfactant homeostasis or other genes. TRG mutation may be associated with hematological and hepatic diseases that may worsen after lung transplantation requiring a specific care and adapted immunosuppression. Surfactant genes mutations are usually associated with ground-glass opacities and cysts on CT scan and may improve with steroids, hydroxychloroquine or azithromycin. Moreover relatives should benefit from a genetic analysis associated with a clinical evaluation according to the gene involved. Genetics of pulmonary fibrosis raise specific problems from diagnosis, therapy or genetic counseling varying from one gene to another.
Collapse
Affiliation(s)
- Raphael Borie
- Unité 1152, Inserm, DHU FIRE, service de pneumologie A, centre de référence des maladies pulmonaires rares, université Paris Diderot, hôpital Bichat, AP-HP, 75013 Paris, France.
| | - Caroline Kannengiesser
- Unité 1152, Inserm, laboratoire de génétique, université Paris Diderot, hôpital Bichat, AP-HP, 75013 Paris, France
| | - Clairelyne Dupin
- Unité 1152, Inserm, DHU FIRE, service de pneumologie A, centre de référence des maladies pulmonaires rares, université Paris Diderot, hôpital Bichat, AP-HP, 75013 Paris, France
| | - Marie-Pierre Debray
- Unité 1152, Inserm, service de radiologie, hôpital Bichat, AP-HP, 75018 Paris, France
| | - Aurélie Cazes
- Inserm, unité 1152, service d'antomopathologie, université Paris Diderot, hôpital Bichat, AP-HP, 75018 Paris, France
| | - Bruno Crestani
- Unité 1152, Inserm, DHU FIRE, service de pneumologie A, centre de référence des maladies pulmonaires rares, université Paris Diderot, hôpital Bichat, AP-HP, 75013 Paris, France
| |
Collapse
|
130
|
Shi H, Yin D, Bonella F, Kreuter M, Oltmanns U, Li X, Peng S, Wei L. Efficacy, safety, and tolerability of combined pirfenidone and N-acetylcysteine therapy: a systematic review and meta-analysis. BMC Pulm Med 2020; 20:128. [PMID: 32380989 PMCID: PMC7204217 DOI: 10.1186/s12890-020-1121-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 03/24/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND While antifibrotic drugs significantly decrease lung function decline in idiopathic pulmonary fibrosis (IPF), there is still an unmet need to halt disease progression. Antioxidative therapy with N-acetylcysteine (NAC) is considered a potential additional therapy that can be combined with antifibrotics in some patients in clinical practice. However, data on the efficacy, tolerability, and safety of this combination are scarce. We performed a systematic review and meta-analysis to appraise the safety, tolerability, and efficacy of the combination compared to treatment with pirfenidone alone. METHODS We systematically reviewed all the published studies with combined pirfenidone (PFD) and NAC (PFD + NAC) treatment in IPF patients. The primary outcomes referred to decline in pulmonary function tests (PFTs) and the rates of IPF patients with side effects. RESULTS In the meta-analysis, 6 studies with 319 total IPF patients were included. The PFD + NAC group was comparable to the PFD alone group in terms of the predicted forced vital capacity (FVC%) and predicted diffusion capacity for carbon monoxide (DLco%) from treatment start to week 24. Side effects and treatment discontinuation rates were also comparable in both groups. CONCLUSION This systematic review and meta-analysis suggests that combination with NAC does not alter the efficacy, safety, or tolerability of PFD in comparison to PFD alone in IPF patients.
Collapse
Affiliation(s)
- Hanyu Shi
- Department of Respiratory and Critical Care Medicine, Special Medical Center of Chinese People's Armed Police Forces, Tianjin, China
- Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Dawei Yin
- Department of Respiratory and Critical Care Medicine, Special Medical Center of Chinese People's Armed Police Forces, Tianjin, China
- Logistics University of Chinese People's Armed Police Forces, Tianjin, China
| | - Francesco Bonella
- Department of Pneumology, Ruhrlandklinik, Centre for Interstitial and Rare Lung Disease, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Michael Kreuter
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, Heidelberg, Germany
| | - Ute Oltmanns
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, Heidelberg, Germany
- Department of Pneumology, Helios Klinikum Pforzheim, Pforzheim, Germany
| | - Xuren Li
- Department of Respiratory and Critical Care Medicine, Special Medical Center of Chinese People's Armed Police Forces, Tianjin, China
| | - Shouchun Peng
- Department of Respiratory and Critical Care Medicine, Special Medical Center of Chinese People's Armed Police Forces, Tianjin, China
| | - Luqing Wei
- Department of Respiratory and Critical Care Medicine, Special Medical Center of Chinese People's Armed Police Forces, Tianjin, China.
- Department of Respiratory and Critical Care Medicine, Special Medical Center of the Chinese People's Armed Police Forces, 220, Cheng-Lin Road, Tianjin, China.
| |
Collapse
|
131
|
Behr J, Prasse A, Wirtz H, Koschel D, Pittrow D, Held M, Klotsche J, Andreas S, Claussen M, Grohé C, Wilkens H, Hagmeyer L, Skowasch D, Meyer JF, Kirschner J, Gläser S, Kahn N, Welte T, Neurohr C, Schwaiblmair M, Bahmer T, Oqueka T, Frankenberger M, Kreuter M. Survival and course of lung function in the presence or absence of antifibrotic treatment in patients with idiopathic pulmonary fibrosis: long-term results of the INSIGHTS-IPF registry. Eur Respir J 2020; 56:13993003.02279-2019. [DOI: 10.1183/13993003.02279-2019] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/07/2020] [Indexed: 11/05/2022]
Abstract
ObjectiveThere is a paucity of observational data on antifibrotic therapy for idiopathic pulmonary fibrosis (IPF). We aimed to assess the course of disease of IPF patients with and without antifibrotic therapy under real-life conditions.MethodsWe analysed data from a non-interventional, prospective cohort study of consecutively enrolled IPF patients from 20 interstitial lung disease expert centres in Germany. Data quality was ensured by automated plausibility checks, on-site monitoring, and source data verification. Propensity scores were applied to account for known differences in baseline characteristics between patients with and without antifibrotic therapy.ResultsAmong the 588 patients suitable for analysis, the mean±sd age was 69.8±9.1 years, and 81.0% were male. The mean±sd duration of disease since diagnosis was 1.8±3.4 years. The mean±sd value at baseline for forced vital capacity (FVC) and diffusion capacity (DLCO) were 68.6±18.8% predicted and 37.8±18.5% predicted, respectively. During a mean±sd follow-up of 1.2±0.7 years, 194 (33.0%) patients died. The 1-year and 2-year survival rates were 87% versus 46% and 62% versus 21%, respectively, for patients with versus without antifibrotic therapy. The risk of death was 37% lower in patients with antifibrotic therapy (hazard ratio 0.63, 95% CI 0.45; 0.87; p=0.005). The results were robust (and remained statistically significant) on multivariable analysis. Overall decline of FVC and DLCO was slow and did not differ significantly between patients with or without antifibrotic therapy.ConclusionsSurvival was significantly higher in IPF patients with antifibrotic therapy, but the course of lung function parameters was similar in patients with and without antifibrotic therapy. This suggests that in clinical practice, premature mortality of IPF patients eventually occurs despite stable measurements for FVC and DLCO.
Collapse
|
132
|
Ni S, Song M, Guo W, Guo T, Shen Q, Peng H. Biomarkers and their potential functions in idiopathic pulmonary fibrosis. Expert Rev Respir Med 2020; 14:593-602. [PMID: 32187497 DOI: 10.1080/17476348.2020.1745066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Idiopathic pulmonary fibrosis (IPF) is a chronic, devastating, and progressive lung disease that is characterized by fibrosis and respiratory failure. IPF holds high morbidity and poor prognosis and still faces considerable problems of reliable diagnosis and valid prognosis. A growing body of literature have reported changes in the level of various biomarkers in IPF patients, which means that they are expected to become a new tool for the clinical practice of IPF.Areas covered: We reviewed the recent literature about biomarkers and focus on the role they play in IPF. We systematically searched Medline/PubMed through February 2020. Many works of literature have shown that a variety of biomolecules and genomics played multiple roles in the diagnosis or differential diagnosis, prognosis, and indication of acute deterioration of IPF and so on.Expert opinion: Significant advances have been made in the role of biomarkers for IPF these years; however, current data indicate that a single biomarker is unlikely to have a transformative effect on clinical practice; therefore, the combined effect of various biomarkers can be considered to improve the accuracy of diagnosis and prognosis. Further research of biomarkers may provide new insights for the diagnosis, prognosis, and even therapy of IPF.
Collapse
Affiliation(s)
- Shanshan Ni
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Min Song
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Wei Guo
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Ting Guo
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Qinxue Shen
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| | - Hong Peng
- Department of Respiratory and Critical Care Medicine, the Second Xiangya Hospital of Central South University; Research Unit of Respiratory Disease, Central South University; The Respiratory Disease Diagnosis and Treatment Center of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
133
|
Diagnostic and Prognostic Biomarkers for Chronic Fibrosing Interstitial Lung Diseases With a Progressive Phenotype. Chest 2020; 158:646-659. [PMID: 32268131 DOI: 10.1016/j.chest.2020.03.037] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/05/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
Biomarkers have the potential to become central to the clinical evaluation and monitoring of patients with chronic fibrosing interstitial lung diseases (ILDs) with a progressive phenotype. Here we summarize the current understanding of putative serum, BAL fluid, and genetic biomarkers in this setting, according to their hypothesized pathobiologic mechanisms: evidence of epithelial cell dysfunction (eg, Krebs von den Lungen-6 antigen), fibroblast proliferation and extracellular matrix production or turnover (eg, matrix metalloproteinase-1), or immune dysregulation (eg, CC chemokine ligand 18). While most of the available data come from idiopathic pulmonary fibrosis (IPF), the prototypic progressive fibrosing ILD, data are available in the broader patient population of chronic fibrosing ILDs. A number of these biomarkers show promise, however, none have been validated. In this review article, we assess both the status of proposed biomarkers for chronic fibrosing lung diseases with a progressive phenotype in predicting disease risk or predisposition, diagnosis, prognosis, and treatment response and provide a direct comparison between IPF and other chronic fibrotic ILDs. We also reflect on the current clinical usefulness and future direction of research for biomarkers in the setting of chronic fibrosing ILDs with a progressive phenotype.
Collapse
|
134
|
Abstract
Idiopathic pulmonary fibrosis (IPF) plays a special role within the group of interstitial lung diseases (ILDs) due to its inexorable progression and its specific medical treatment. With a median survival of only 2-3 years from the time of diagnosis, the prognosis is worse than many carcinomas.In contrast to other ILDs, IPF does not respond to anti-inflammatory treatment with corticosteroids but rather demands a specific medical therapy. Even though this cannot cure the disease, it can prolong survival. Lung transplantation is the only cure for progressive lung fibrosis. The clinical course is individual and difficult to predict. Acute exacerbations accelerate the clinical course and lead to high mortality.The underlying pathomechanisms of IPF, with its complex immunological and inflammatory processes and external impacts, have been the focus of recent research. Lifestyle and environmental influences are held responsible for much of its natural history. Smoking, pneumotoxic medications, and inhalation of dusts are well-known risk factors. Likewise, genetic and hereditary factors play a crucial role.This short review focuses on the peculiarities of IPF within the group of ILDs, especially in relation to its underlying mechanisms and clinical progression.
Collapse
|
135
|
Janssen-Heininger Y, Reynaert NL, van der Vliet A, Anathy V. Endoplasmic reticulum stress and glutathione therapeutics in chronic lung diseases. Redox Biol 2020; 33:101516. [PMID: 32249209 PMCID: PMC7251249 DOI: 10.1016/j.redox.2020.101516] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Yvonne Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| |
Collapse
|
136
|
Wang Y, Xiao H, Zhao F, Li H, Gao R, Yan B, Ren J, Yang J. Decrypting the crosstalk of noncoding RNAs in the progression of IPF. Mol Biol Rep 2020; 47:3169-3179. [PMID: 32180083 DOI: 10.1007/s11033-020-05368-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/29/2020] [Indexed: 12/16/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an agnogenic, rare, and lethal disease, with high mortality and poor prognosis and a median survival time as short as 3 to 5 years after diagnosis. No effective therapeutic drugs are still not available not only in clinical practice, but also in preclinical phases. To better and deeper understand pulmonary fibrosis will provide more effective strategies for therapy. Mounting evidence suggests that noncoding RNAs (ncRNAs) and their interactions may contribute to lung fibrosis; however, the mechanisms underlying their roles are largely unknown. In this review, we systematically summarized the recent advances regarding the crucial roles of long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) and crosstalk among them in the development of IPF. The perspective for related genes was well highlighted. In summary, ncRNA and their interactions play a key regulatory part in the progression of IPF and are bound to provide us with new diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Yujuan Wang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Han Xiao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Fenglian Zhao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Han Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Rong Gao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Bingdi Yan
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Jin Ren
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Junling Yang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
137
|
Otoupalova E, Smith S, Cheng G, Thannickal VJ. Oxidative Stress in Pulmonary Fibrosis. Compr Physiol 2020; 10:509-547. [PMID: 32163196 DOI: 10.1002/cphy.c190017] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Oxidative stress has been linked to various disease states as well as physiological aging. The lungs are uniquely exposed to a highly oxidizing environment and have evolved several mechanisms to attenuate oxidative stress. Idiopathic pulmonary fibrosis (IPF) is a progressive age-related disorder that leads to architectural remodeling, impaired gas exchange, respiratory failure, and death. In this article, we discuss cellular sources of oxidant production, and antioxidant defenses, both enzymatic and nonenzymatic. We outline the current understanding of the pathogenesis of IPF and how oxidative stress contributes to fibrosis. Further, we link oxidative stress to the biology of aging that involves DNA damage responses, loss of proteostasis, and mitochondrial dysfunction. We discuss the recent findings on the role of reactive oxygen species (ROS) in specific fibrotic processes such as macrophage polarization and immunosenescence, alveolar epithelial cell apoptosis and senescence, myofibroblast differentiation and senescence, and alterations in the acellular extracellular matrix. Finally, we provide an overview of the current preclinical studies and clinical trials targeting oxidative stress in fibrosis and potential new strategies for future therapeutic interventions. © 2020 American Physiological Society. Compr Physiol 10:509-547, 2020.
Collapse
Affiliation(s)
- Eva Otoupalova
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sam Smith
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Guangjie Cheng
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
138
|
Baddini-Martinez J, Ferreira J, Tanni S, Alves LR, Cabral BF, Carvalho CRR, Cezare TJ, da Costa CH, Gazzana MB, Jezler S, Kairalla RA, Kawano-Dourado L, Lima MS, Mancuzo E, Moreira MAC, Rodrigues MP, Rodrigues SCS, Rubin AS, Rufino RL, Steidle LJM, Storrer K, Baldi BG. Brazilian guidelines for the pharmacological treatment of idiopathic pulmonary fibrosis. Official document of the Brazilian Thoracic Association based on the GRADE methodology. J Bras Pneumol 2020; 46:e20190423. [PMID: 32130337 PMCID: PMC7462709 DOI: 10.36416/1806-3756/e20190423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/12/2020] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a form of chronic interstitial lung disease of unknown cause, which predominantly affects elderly men who are current or former smokers. Even though it is an uncommon disease, it is of great importance because of its severity and poor prognosis. In recent decades, several pharmacological treatment modalities have been investigated for the treatment of this disease, and the classic concepts have therefore been revised. The purpose of these guidelines was to define evidence-based recommendations regarding the use of pharmacological agents in the treatment of IPF in Brazil. We sought to provide guidance on the practical issues faced by clinicians in their daily lives. Patients of interest, Intervention to be studied, Comparison of intervention and Outcome of interest (PICO)-style questions were formulated to address aspects related to the use of corticosteroids, N-acetylcysteine, gastroesophageal reflux medications, endothelin-receptor antagonists, phosphodiesterase-5 inhibitors, pirfenidone, and nintedanib. To formulate the PICO questions, a group of Brazilian specialists working in the area was assembled and an extensive review of the literature on the subject was carried out. Previously published systematic reviews with meta-analyses were analyzed for the strength of the compiled evidence, and, on that basis, recommendations were developed by employing the Grading of Recommendations Assessment, Development and Evaluation approach. The authors believe that the present document represents an important advance to be incorporated in the approach to patients with IPF, aiming mainly to improve its management, and can become an auxiliary tool for defining public policies related to IPF.
Collapse
Affiliation(s)
- José Baddini-Martinez
- . Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto (SP) Brasil
- . Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo (SP) Brasil
| | - Juliana Ferreira
- . Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Suzana Tanni
- . Faculdade de Medicina de Botucatu, Universidade Estadual Paulista - UNESP - Botucatu (SP) Brasil
| | - Luis Renato Alves
- . Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto (SP) Brasil
| | | | | | - Talita Jacon Cezare
- . Faculdade de Medicina de Botucatu, Universidade Estadual Paulista - UNESP - Botucatu (SP) Brasil
| | | | | | - Sérgio Jezler
- . Hospital Geral Roberto Santos, Salvador (BA) Brasil
| | | | | | | | - Eliane Mancuzo
- . Universidade Federal de Minas Gerais, Belo Horizonte (MG) Brasil
| | | | | | | | - Adalberto Sperb Rubin
- . Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre (RS) Brasil
| | | | | | - Karin Storrer
- . Universidade Federal do Paraná - UFPR - Curitiba (PR) Brasil
| | - Bruno Guedes Baldi
- . Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| |
Collapse
|
139
|
Kaner RJ, Bajwa EK, El-Amine M, Gorina E, Gupta R, Lazarus HM, Luckhardt TR, Mouded M, Posada K, Richeldi L, Stauffer J, Tutuncu A, Martinez FJ. Design of Idiopathic Pulmonary Fibrosis Clinical Trials in the Era of Approved Therapies. Am J Respir Crit Care Med 2020; 200:133-139. [PMID: 30985215 DOI: 10.1164/rccm.201903-0592pp] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Robert J Kaner
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and.,2 Department of Genetic Medicine, Weill Cornell Medicine, New York, New York
| | | | | | - Eduard Gorina
- 5 Pliant Therapeutics, Inc., South San Francisco, California
| | - Renu Gupta
- 6 Promedior, Inc., Lexington, Massachusetts
| | - Howard M Lazarus
- 7 Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut
| | - Tracy R Luckhardt
- 8 Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Kaity Posada
- 10 Acceleron Pharma, Inc., Cambridge, Massachusetts
| | - Luca Richeldi
- 11 Fondazione Policlinico A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - John Stauffer
- 12 Genentech, Inc., South San Francisco, California; and
| | | | - Fernando J Martinez
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| |
Collapse
|
140
|
Ricci F, Pugliese L, Cavallo AU, Forcina M, De Stasio V, Presicce M, Di Tosto F, Di Donna C, Spiritigliozzi L, Rogliani P, Floris R, Chiocchi M. Highlights of high-resolution computed tomography imaging in evaluation of complications and co-morbidities in idiopathic pulmonary fibrosis. Acta Radiol 2020; 61:204-218. [PMID: 31237771 DOI: 10.1177/0284185119857435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) represents a condition included in the heterogeneous group of interstitial lung diseases without known causes. The recent ATS/ERS/JRS/ALAT guidelines and the white paper published by the Fleischner Society have well-defined diagnosis and management of idiopathic pulmonary fibrosis. Idiopathic pulmonary fibrosis management is complex because it is also influenced by several co-morbidities and complications. The new frontier in idiopathic pulmonary fibrosis is represented by the effort to understand the complex mechanism of the pathogenesis and progression of disease in order to predict several consequences and co-morbidities. In our review, we tried to distinguish co-morbidities from complications of idiopathic pulmonary fibrosis. In each complication, we have reviewed the existing literature and we have emphasized the complex pathobiological pathway which links the progression of idiopathic pulmonary fibrosis to the development of the complication itself. For every co-morbidity, we tried to identify share common risk factors which explain the coexistence of idiopathic pulmonary fibrosis with its co-morbidities. We then analyzed high-resolution computed tomography (CT) aspects of co-morbidities and complications of idiopathic pulmonary fibrosis that the radiologist should be aware of. In this review, we focused on the role of high-resolution CT imaging in the evaluation of co-morbidities and complications in idiopathic pulmonary fibrosis because their early diagnosis and treatment could change the prognosis in patients with idiopathic pulmonary fibrosis. We have also pointed out that in some cases the final combined quantitative CT tools and conventional visual CT score would allow to get an accurate analysis and quantification of disease progression, co-morbidities, and complications of idiopathic pulmonary fibrosis in order to improve staging systems in idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Francesca Ricci
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Luca Pugliese
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Armando Ugo Cavallo
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Marco Forcina
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Vincenzo De Stasio
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Matteo Presicce
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Federica Di Tosto
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Carlo Di Donna
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Luigi Spiritigliozzi
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Paola Rogliani
- Respiratory Medicine. Department of Systems Medicine, University of Rome "Tor Vergata," Rome, Italy
| | - Roberto Floris
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| | - Marcello Chiocchi
- Department of Biomedicine and Prevention Division of Diagnostic Imaging, University of Rome "Tor Vergata," Rome, Italy
| |
Collapse
|
141
|
Planté-Bordeneuve T, Haouas H, Vanderheyde K, Froidure A. Telomerase-related monogenic lung fibrosis presenting with subacute onset: a case report and review of literature. Acta Clin Belg 2019; 74:445-450. [PMID: 30451599 DOI: 10.1080/17843286.2018.1545375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Objectives: Monogenic pulmonary fibrosis related to telomerase mutations is characterized by a large spectrum of clinical presentations. The disease may affect several organs including bone marrow, liver and skin. This case illustrates some of the most salient features of telomere-related Interstitial Lung Disease(ILD). Methods: Single case study and review of the litterature. Results: We report the case of a 44-year-old man admitted to our unit for subacute pulmonary fibrosis. No underlying cause could be identified. Personal and familial history was highly suggestive of monogenic telomere related lung fibrosis. Genetic investigation confirmed a mutation in the TERT gene, coding for one of the components of telomerase. Given the severe hypoxemia unresponsive to supportive treatment, he was referred for urgent lung transplantation, with a favourable outcome. Genetic counselling was proposed to his family. Conclusions: Telomerase-related monogenic lung fibrosis may present with a subacute onset, requiring urgent lung transplantation. Extra-thoracic clinical manifestations and familial history are key elements pointing towards the diagnosis.
Collapse
Affiliation(s)
| | - Hanae Haouas
- Service de pneumologie, Hopital Notre-Dame de Grâce, Gosselies, Belgium
| | - Kim Vanderheyde
- Service de pneumologie, Hopital Notre-Dame de Grâce, Gosselies, Belgium
| | - Antoine Froidure
- Service de pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
142
|
Abstract
Significance: Redox homeostasis is finely tuned and governed by distinct intracellular mechanisms. The dysregulation of this either by external or internal events is a fundamental pathophysiologic base for many pulmonary diseases. Recent Advances: Based on recent discoveries, it is increasingly clear that cellular redox state and oxidation of signaling molecules are critical modulators of lung disease and represent a final common pathway that leads to poor respiratory outcomes. Critical Issues: Based on the wide variety of stimuli that alter specific redox signaling pathways, improved understanding of the disease and patient-specific alterations are needed for the development of therapeutic targets. Further Directions: For the full comprehension of redox signaling in pulmonary disease, it is essential to recognize the role of reactive oxygen intermediates in modulating biological responses. This review summarizes current knowledge of redox signaling in pulmonary development and pulmonary vascular disease.
Collapse
Affiliation(s)
- Gaston Ofman
- Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Trent E Tipple
- Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
143
|
Somogyi V, Chaudhuri N, Torrisi SE, Kahn N, Müller V, Kreuter M. The therapy of idiopathic pulmonary fibrosis: what is next? Eur Respir Rev 2019; 28:190021. [PMID: 31484664 PMCID: PMC9488691 DOI: 10.1183/16000617.0021-2019] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrosing interstitial lung disease, characterised by progressive scarring of the lung and associated with a high burden of disease and early death. The pathophysiological understanding, clinical diagnostics and therapy of IPF have significantly evolved in recent years. While the recent introduction of the two antifibrotic drugs pirfenidone and nintedanib led to a significant reduction in lung function decline, there is still no cure for IPF; thus, new therapeutic approaches are needed. Currently, several clinical phase I-III trials are focusing on novel therapeutic targets. Furthermore, new approaches in nonpharmacological treatments in palliative care, pulmonary rehabilitation, lung transplantation, management of comorbidities and acute exacerbations aim to improve symptom control and quality of life. Here we summarise new therapeutic attempts and potential future approaches to treat this devastating disease.
Collapse
Affiliation(s)
- Vivien Somogyi
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
- Dept of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Nazia Chaudhuri
- Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK
| | - Sebastiano Emanuele Torrisi
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
- Regional Referral Centre for Rare Lung Diseases, University Hospital "Policlinico", Dept of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Nicolas Kahn
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Veronika Müller
- Dept of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Michael Kreuter
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
144
|
Borie R, Le Guen P, Ghanem M, Taillé C, Dupin C, Dieudé P, Kannengiesser C, Crestani B. The genetics of interstitial lung diseases. Eur Respir Rev 2019; 28:28/153/190053. [PMID: 31554702 PMCID: PMC9488931 DOI: 10.1183/16000617.0053-2019] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022] Open
Abstract
Interstitial lung diseases (ILDs) are a set of heterogeneous lung diseases characterised by inflammation and, in some cases, fibrosis. These lung conditions lead to dyspnoea, cough, abnormalities in gas exchange, restrictive physiology (characterised by decreased lung volumes), hypoxaemia and, if progressive, respiratory failure. In some cases, ILDs can be caused by systemic diseases or environmental exposures. The ability to treat or cure these ILDs varies based on the subtype and in many cases lung transplantation remains the only curative therapy. There is a growing body of evidence that both common and rare genetic variants contribute to the development and clinical manifestation of many of the ILDs. Here, we review the current understanding of genetic risk and ILD. Common and rare genetic variants contribute to the development and clinical manifestation of many interstitial lung diseaseshttp://bit.ly/31loHLh
Collapse
Affiliation(s)
- Raphael Borie
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Pierre Le Guen
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Mada Ghanem
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Camille Taillé
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Clairelyne Dupin
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France.,INSERM U1152, Paris, France
| | - Philippe Dieudé
- INSERM U1152, Paris, France.,Département de Génétique, Hôpital Bichat, AP-HP, Paris, France
| | - Caroline Kannengiesser
- INSERM U1152, Paris, France.,Service de Rhumatologie, Hôpital Bichat, AP-HP, Paris, France
| | - Bruno Crestani
- Service de Pneumologie A, Hôpital Bichat, AP-HP, Paris, France .,INSERM U1152, Paris, France
| |
Collapse
|
145
|
Ballester B, Milara J, Cortijo J. Mucins as a New Frontier in Pulmonary Fibrosis. J Clin Med 2019; 8:jcm8091447. [PMID: 31514468 PMCID: PMC6780288 DOI: 10.3390/jcm8091447] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common idiopathic interstitial pulmonary disease with a median survival of 3–5 years after diagnosis. Recent evidence identifies mucins as key effectors in cell growth and tissue remodeling processes compatible with the processes observed in IPF. Mucins are classified in two groups depending on whether they are secreted (secreted mucins) or tethered to cell membranes (transmembrane mucins). Secreted mucins (MUC2, MUC5AC, MUC5B, MUC6-8 and MUC19) are released to the extracellular medium and recent evidence has shown that a promoter polymorphism in the secreted mucin MUC5B is associated with IPF risk. Otherwise, transmembrane mucins (MUC1, MUC3, MUC4, MUC12-17 and MUC20) have a receptor-like structure, sensing the external environment and activating intracellular signal transduction pathways essential for mucosal maintenance and damage repair. In this context, the extracellular domain can be released to the external environment by metalloproteinase action, increased in IPF, thus activating fibrotic processes. For example, several studies have reported increased serum extracellular secreted KL6/MUC1 during IPF acute exacerbation. Moreover, MUC1 and MUC4 overexpression in the main IPF cells has been observed. In this review we summarize the current knowledge of mucins as promising druggable targets for IPF.
Collapse
Affiliation(s)
- Beatriz Ballester
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain.
- CIBERES, Health Institute Carlos III, 46010 Valencia, Spain.
| | - Javier Milara
- CIBERES, Health Institute Carlos III, 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, 46010 Valencia, Spain
- Research and teaching Unit, University General Hospital Consortium of Valencia, 46014 Valencia, Spain
| |
Collapse
|
146
|
Saito S, Alkhatib A, Kolls JK, Kondoh Y, Lasky JA. Pharmacotherapy and adjunctive treatment for idiopathic pulmonary fibrosis (IPF). J Thorac Dis 2019; 11:S1740-S1754. [PMID: 31632751 PMCID: PMC6783717 DOI: 10.21037/jtd.2019.04.62] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an advancing and fatal lung disease with increasing incidence and prevalence. Nintedanib and pirfenidone were approved by the FDA for the treatment of IPF in 2014 based on positive phase 3 trials, and both of these antifibrotic drugs are conditionally recommended in the 2015 ATS/ERS/JRS/ALAT Clinical Practice Guideline. Although an improvement over previously suggested therapies, their capacity to reduce, but not completely arrest or improve, lung function over time presents an opportunity for novel or add-on pharmacologic agents. The purpose of this review is to deliver a brief overview of the results of phase 3/4 IPF trials with pirfenidone and nintedanib, as well as highlight encouraging results of phase 1/2 trials with novel therapies. Long-term studies indicate that pirfenidone and nintedanib are effective IPF treatments, with acceptable safety and tolerability. The combination of pirfenidone and nintedanib appear safe. Promising results have recently been made public for several phase 2 trials with novel targets, including the autotaxin-lysophosphatidic acid (ATX/LPA) pathway, connective tissue growth factor (CTGF), pentraxin-2, G protein-coupled receptor agonists/antagonists, αvβ6 integrin, and galectin-3. Results of treatments directed at gastro-esophageal reflux in patients with IPF have also been published. Currently, monotherapy with pirfenidone or nintedanib is the mainstay of pharmacological treatment for IPF. Innovative therapies along with combinations of pharmacological agents hold great promise for the future.
Collapse
Affiliation(s)
| | | | | | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Aichi, Japan
| | | |
Collapse
|
147
|
Jessurun NT, Drent M, van Puijenbroek EP, Bekers O, Wijnen PA, Bast A. Drug-induced interstitial lung disease: role of pharmacogenetics in predicting cytotoxic mechanisms and risks of side effects. Curr Opin Pulm Med 2019; 25:468-477. [PMID: 31365381 DOI: 10.1097/mcp.0000000000000590] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The diagnosis of drug-induced interstitial lung disease (DI-ILD) is challenging and mainly made by exclusion of other possible causes. Toxicity can occur as a cause of drug(s) or drug-drug interactions. In this review, we summarize the possible role of pharmacogenetics of metabolizing enzymes in DI-ILD. RECENT FINDINGS Knowledge of the genetic predispositions of enzymes involved in drug metabolization and their relation with proposed cytotoxic mechanisms of DI-ILD, in particular direct cell toxicity and free oxygen radical production is increasing. The cytochrome P450 enzyme family and other enzymes play an important role in the metabolism of all sorts of ingested, injected, or inhaled xenobiotic substances. The liver is the major site for metabolism. Metabolic cytotoxic mechanisms have however also been detected in lung tissue. Polymorphisms in genes coding for enzymes that influence metabolic activity may lead to localized (toxic) reactions and tissue damage. This knowledge may be helpful in preventing the risk of DI-ILD. SUMMARY Drug toxicity can be the consequence of absence or very poor enzyme activity, especially if no other metabolic route is available. In the case of reduced enzyme activity, it is recommended to reduce the dose or to prescribe an alternative drug, which is metabolized by a different, unaffected enzyme system to prevent toxic side effects. However, enhanced enzyme activity may lead to excessive formation of toxic and sometimes reactive metabolites. Therefore, knowing a patient's drug-metabolizing profile before drug prescription is a promising way to prevent or explain DI-ILD.
Collapse
Affiliation(s)
- Naomi T Jessurun
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
- ILD Care Foundation Research Team, Ede, The Netherlands
| | - Marjolein Drent
- ILD Care Foundation Research Team, Ede, The Netherlands
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, The Netherlands
- ILD Center of Excellence, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Eugène P van Puijenbroek
- Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, The Netherlands
- Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Otto Bekers
- ILD Care Foundation Research Team, Ede, The Netherlands
- Department of Clinical Chemistry, Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Petal A Wijnen
- ILD Care Foundation Research Team, Ede, The Netherlands
- Department of Clinical Chemistry, Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Aalt Bast
- ILD Care Foundation Research Team, Ede, The Netherlands
- Department of Pharmacology and Toxicology, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, The Netherlands
- Venlo Campus, Maastricht University, Venlo, The Netherlands
| |
Collapse
|
148
|
Isobe K, Issiki T, Sakamoto S, Sano G, Takai Y, Tochigi N, Homma S. Clinical importance of Bcl-2-like 11 deletion polymorphism in idiopathic pulmonary fibrosis. J Thorac Dis 2019; 11:2981-2989. [PMID: 31463128 DOI: 10.21037/jtd.2019.07.25] [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: 01/08/2023]
Abstract
Background Reactive oxygen species (ROS) can play a role in the pathogenesis of Idiopathic Pulmonary Fibrosis (IPF) by contributing to epithelial damage. Bcl-2-like 11 (BIM) is involved in the generation of ROS via forkhead box O3 (FOXO3), and a BIM deletion polymorphism related to apoptosis has been reported to be specific to Asians. Here we examine the clinical features of IPF in patients with BIM deletion polymorphism. Methods In this single-center retrospective study, we reviewed the medical records of 63 patients with IPF who were treated at our hospital from January 2006 through April 2018. Patients with and without BIM deletion polymorphism were compared in relation to clinical characteristics, pulmonary function test results, frequency of acute exacerbation (AE)-IPF, and redox status [including oxidized glutathione (GSSG), reduced glutathione (GSH), 8-hydroxy-2'-deoxyguanosine (8-OHdG), and 8-isoprostane (8-iso)] at baseline and at 1 and 2 years after treatment. Results No fatal AE-IPF occurred in patients with BIM deletion polymorphism (0% vs. 36.4% of polymorphism-negative cases; P=0.038). Change in forced vital capacity from baseline (ΔFVC) at 2 years was significantly lower in patients with the BIM deletion polymorphism than in those without the polymorphism (‒0.42±0.50 vs. ‒0.88±0.83 L, respectively; P=0.045). Total blood glutathione (tGSH) was significantly higher in patients with the deletion polymorphism than in those without the polymorphism (988±48.3 vs. 858±25.8 µM, respectively; P=0.042). 8-iso was significantly lower in the former group (246.1±81.6 vs. 400.9±334.1 pg/mL, respectively; P=0.022). Conclusions IPF patients with BIM deletion polymorphism had a good redox balance and may thus have a better clinical outcome than patients without this polymorphism.
Collapse
Affiliation(s)
- Kazutoshi Isobe
- Division of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Takuma Issiki
- Division of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Susumu Sakamoto
- Division of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Go Sano
- Division of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Yujiro Takai
- Division of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| | - Naobumi Tochigi
- Division of Surgical Pathology, Toho University School of Medicine, Tokyo, Japan
| | - Sakae Homma
- Division of Respiratory Medicine, Toho University School of Medicine, Tokyo, Japan
| |
Collapse
|
149
|
Moua T, Lee AS, Ryu JH. Comparing effectiveness of prognostic tests in idiopathic pulmonary fibrosis. Expert Rev Respir Med 2019; 13:993-1004. [PMID: 31405303 DOI: 10.1080/17476348.2019.1656069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Idiopathic pulmonary fibrosis (IPF) is a debilitating and progressive fibrotic interstitial lung disease often resulting in death over several years. Prediction of disease course or survival remains of keen interest for clinicians and patients though a commonly used test or tool remain elusive. Areas covered: We undertook a comprehensive review of the published literature highlighting prognostic indicators and predictors of survival in IPF. Baseline and longitudinal clinical, functional, histopathologic, and radiologic findings have been extensively studied as prognostic predictors, both individually and in composite models. Recent approaches include automated quantifiable radiologic scoring, circulating biomarkers, and genetic polymorphisms or abnormalities. This review highlights individual and composite predictors and their relative utility in clinical practice and research studies. Expert opinion: There is a growing body of knowledge highlighting readily available individual and composite predictors of outcome, though none have come to the forefront for common clinical use. Recent advances include quantitative imaging analysis, circulating serologic markers, and genetic testing, which may be more standardized and less prone to lead-time bias or related complications and comorbidities.
Collapse
Affiliation(s)
- Teng Moua
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic , Rochester , MN , USA
| | - Augustine S Lee
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic , Jacksonville , FL , USA
| | - Jay H Ryu
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
150
|
Newton CA, Zhang D, Oldham JM, Kozlitina J, Ma SF, Martinez FJ, Raghu G, Noth I, Garcia CK. Telomere Length and Use of Immunosuppressive Medications in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2019; 200:336-347. [PMID: 30566847 PMCID: PMC6680304 DOI: 10.1164/rccm.201809-1646oc] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/18/2018] [Indexed: 12/17/2022] Open
Abstract
Rationale: Immunosuppression was associated with adverse events for patients with idiopathic pulmonary fibrosis (IPF) in the PANTHER-IPF (Evaluating the Effectiveness of Prednisone, Azathioprine and N-Acetylcysteine in Patients with IPF) clinical trial. The reason why some patients with IPF experience harm is unknown.Objectives: To determine whether age-adjusted leukocyte telomere length (LTL) was associated with the harmful effect of immunosuppression in patients with IPF.Methods: LTL was measured from available DNA samples from PANTHER-IPF (interim analysis, n = 79; final analysis, n = 118). Replication cohorts included ACE-IPF (Anticoagulant Effectiveness in Idiopathic Pulmonary Fibrosis) (n = 101) and an independent observational cohort (University of Texas Southwestern Medical Center-IPF, n = 170). LTL-stratified and medication-stratified survival analyses were performed using multivariable Cox regression models for composite endpoint-free survival.Measurements and Main Results: Of the subjects enrolled in the PANTHER-IPF and ACE-IPF, 62% (49/79) and 56% (28/50) had an LTL less than the 10th percentile of normal, respectively. In PANTHER-IPF, exposure to prednisone/azathioprine/N-acetylcysteine was associated with a higher composite endpoint of death, lung transplantation, hospitalization, or FVC decline for those with an LTL less than the 10th percentile (hazard ratio, 2.84; 95% confidence interval, 1.02-7.87; P = 0.045). This finding was replicated in the placebo arm of ACE-IPF for those exposed to immunosuppression (hazard ratio, 7.18; 95% confidence interval, 1.52-33.84; P = 0.013). A propensity-matched University of Texas Southwestern Medical Center IPF cohort showed a similar association between immunosuppression and composite endpoints (death, lung transplantation, or FVC decline) for those with an LTL less than the 10th percentile (hazard ratio, 3.79; 95% confidence interval, 1.73-8.30; P = 0.00085). An interaction was found between immunosuppression and LTL for the combined PANTHER-IPF and ACE-IPF clinical trials (Pinteraction = 0.048), and the University of Texas Southwestern Medical Center IPF cohort (Pinteraction = 0.00049).Conclusions: LTL is a biomarker that may identify patients with IPF at risk for poor outcomes when exposed to immunosuppression.
Collapse
Affiliation(s)
| | - David Zhang
- Department of Medicine and
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Justin M. Oldham
- Department of Medicine, University of California at Davis, Davis, California
| | - Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Shwu-Fan Ma
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| | | | - Ganesh Raghu
- Department of Medicine, University of Washington Medical Center, Seattle, Washington
| | - Imre Noth
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Christine Kim Garcia
- Department of Medicine and
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|