Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Thorax ; 79(1): 68-74, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37758458

ABSTRACT

BACKGROUND: Autoimmune pulmonary alveolar proteinosis (aPAP) is a rare disease, predisposing to an increased risk of infection. A complete picture of these infections is lacking. RESEARCH QUESTION: Describe the characteristics and clinical outcomes of patients diagnosed with aPAP, and to identify risk factors associated with opportunistic infections. METHODS: We conducted a retrospective cohort including all patients diagnosed with aPAP between 2008 and 2018 in France and Belgium. Data were collected using a standardised questionnaire including demographics, comorbidities, imaging features, outcomes and microbiological data. RESULTS: We included 104 patients, 2/3 were men and median age at diagnosis was 45 years. With a median follow-up of 3.4 years (IQR 1.7-6.6 years), 60 patients (58%), developed at least one infection, including 23 (22%) with opportunistic infections. Nocardia spp was the main pathogen identified (n=10). Thirty-five (34%) patients were hospitalised due to infection. In univariate analysis, male gender was associated with opportunistic infections (p=0.04, OR=3.88; 95% CI (1.02 to 22.06)). Anti-granulocyte macrophage colony-stimulating factor antibody titre at diagnosis was significantly higher among patients who developed nocardiosis (1058 (316-1591) vs 580 (200-1190), p=0.01). Nine patients had died (9%), but only one death was related to infection. INTERPRETATION: Patients with aPAP often presented with opportunistic infections, especially nocardiosis, which highlights the importance of systematic search for slow-growing bacteria in bronchoalveolar lavage or whole lung lavage.


Subject(s)
Autoimmune Diseases , Nocardia Infections , Opportunistic Infections , Pulmonary Alveolar Proteinosis , Humans , Male , Middle Aged , Female , Retrospective Studies , Granulocyte-Macrophage Colony-Stimulating Factor , Autoimmune Diseases/complications , Nocardia Infections/diagnosis , Nocardia Infections/epidemiology , Autoantibodies
2.
Eur Respir J ; 61(6)2023 06.
Article in English | MEDLINE | ID: mdl-37230499

ABSTRACT

BACKGROUND: Standard of care for interstitial lung disease (ILD) with a nonspecific interstitial pneumonia (NSIP) pattern proposes mycophenolate mofetil (MMF) as one of the first-step therapies while rituximab is used as rescue therapy. METHODS: In a randomised, double-blind, two-parallel group, placebo-controlled trial (NCT02990286), patients with connective tissue disease-associated ILD or idiopathic interstitial pneumonia (with or without autoimmune features) and a NSIP pattern (defined on NSIP pathological pattern or on integration of clinicobiological data and a NSIP-like high-resolution computed tomography pattern) were randomly assigned in a 1:1 ratio to receive rituximab (1000 mg) or placebo on day 1 and day 15 in addition to MMF (2 g daily) for 6 months. The primary end-point was the change in percent predicted forced vital capacity (FVC) from baseline to 6 months analysed by a linear mixed model for repeated measures analysis. Secondary end-points included progression-free survival (PFS) up to 6 months and safety. FINDINGS: Between January 2017 and January 2019, 122 randomised patients received at least one dose of rituximab (n=63) or placebo (n=59). The least-squares mean change from baseline to 6 months in FVC (% predicted) was +1.60 (se 1.13) in the rituximab+MMF group and -2.01 (se 1.17) in the placebo+MMF group (between-group difference 3.60, 95% CI 0.41-6.80; p=0.0273). PFS was better in the rituximab+MMF group (crude hazard ratio 0.47, 95% CI 0.23-0.96; p=0.03). Serious adverse events occurred in 26 (41%) patients of the rituximab+MMF group and in 23 (39%) of the placebo+MMF group. Nine infections were reported in the rituximab+MMF group (five bacterial infections, three viral infections, one other) and four bacterial infections in the placebo+MMF group. INTERPRETATION: Combination of rituximab and MMF was superior to MMF alone in patients with ILD and a NSIP pattern. The use of this combination must take into consideration the risk of viral infection.


Subject(s)
Idiopathic Interstitial Pneumonias , Lung Diseases, Interstitial , Humans , Rituximab/therapeutic use , Rituximab/adverse effects , Mycophenolic Acid/therapeutic use , Immunosuppressive Agents/adverse effects , Lung , Treatment Outcome , Lung Diseases, Interstitial/drug therapy , Idiopathic Interstitial Pneumonias/drug therapy , Double-Blind Method
3.
Am J Transplant ; 22(4): 1236-1244, 2022 04.
Article in English | MEDLINE | ID: mdl-34854205

ABSTRACT

Carriers of germline telomerase-related gene (TRG) mutations can show poor prognosis, with an increase in common hematological complications after lung transplantation (LT) for pulmonary fibrosis. The aim of this study was to describe the outcomes after LT in recipients carrying a germline TRG mutation and to identify the predictors of survival. In a multicenter cohort of LT patients, we retrospectively reviewed those carrying pathogenic TRG variations (n = 38; TERT, n = 23, TERC, n = 9, RTEL1, n = 6) between 2009 and 2018. The median age at LT was 54 years (interquartile range [IQR] 46-59); 68% were male and 71% had idiopathic pulmonary fibrosis. During the diagnosis of pulmonary fibrosis, 28 (74%) had a hematological disease, including eight with myelodysplasia. After a median follow-up of 26 months (IQR 15-46), 38 patients received LT. The overall post-LT median survival was 3.75 years (IQR 1.8-NA). The risk of death after LT was increased for patients with myelodysplasia (HR 4.1 [95% CI 1.5-11.5]) or short telomere (HR 2.2 [1.0-5.0]) before LT. After LT, all patients had anemia, 66% had thrombocytopenia, and 39% had neutropenia. Chronic lung allograft dysfunction frequency was 29% at 4 years. The present findings support the use of LT in TRG mutation carriers without myelodysplasia. Hematological evaluation should be systematically performed before LT.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung Transplantation , Telomerase , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/surgery , Lung Transplantation/adverse effects , Male , Middle Aged , Mutation , Retrospective Studies , Telomerase/genetics
4.
Respiration ; 101(1): 34-45, 2022.
Article in English | MEDLINE | ID: mdl-34515219

ABSTRACT

BACKGROUND: There is growing evidence of gender-specific phenotypic differences among patients with idiopathic pulmonary fibrosis (IPF), which may affect patient outcomes. OBJECTIVES: We present the characteristics of patients with IPF at inclusion in the French Rare Disease Cohort - Interstitial Lung Disease (RaDiCo-ILD) with the aim of characterizing gender-specific phenotypic differences. METHODS: Patients with IPF who were enrolled in the national, multicentre RaDiCo-ILD cohort were included. Demographic characteristics, comorbidities, health-related quality of life (HRQoL) scores, pulmonary function, chest imaging, and IPF treatment were collected at inclusion and described by gender. RESULTS: The cohort included 724 patients with IPF (54% of RaDiCo-ILD cohort), of whom 82.9% were male. The proportion of male and female patients with a prior history of smoking was 75.0% and 26.8%, respectively. Emphysema was present in 17.0% (95% confidence interval [CI]: 10.0, 24.0) of men and 5.4% (95% CI: 1.2, 9.6) of women. At inclusion, females had poorer HRQoL than males based on St. George's Respiratory Questionnaire scores (48.5 [95% CI: 43.9, 53.0] and 41.5 [39.4, 43.6], respectively). The mean forced vital capacity per cent predicted was 77.7% (95% CI: 76.2, 79.3) and 87.4% (83.4, 91.4) for males and females, respectively. Honeycombing on high-resolution computed tomography (HRCT) was present in 70.8% (95% CI: 61.0, 80.6) of males and 45.8% (95% CI: 35.1, 56.5) of females. CONCLUSIONS: This analysis of patients with IPF at inclusion in the RaDiCo-ILD cohort provides evidence that comorbid emphysema, lung volume reduction, and honeycombing on HRCT are more common characteristics of males than females.


Subject(s)
Emphysema , Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Cohort Studies , Female , Humans , Idiopathic Pulmonary Fibrosis/diagnostic imaging , Idiopathic Pulmonary Fibrosis/epidemiology , Male , Quality of Life , Rare Diseases
5.
N Engl J Med ; 379(23): 2209-2219, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30345907

ABSTRACT

BACKGROUND: Given the phenotypic similarities between rheumatoid arthritis (RA)-associated interstitial lung disease (ILD) (hereafter, RA-ILD) and idiopathic pulmonary fibrosis, we hypothesized that the strongest risk factor for the development of idiopathic pulmonary fibrosis, the gain-of-function MUC5B promoter variant rs35705950, would also contribute to the risk of ILD among patients with RA. METHODS: Using a discovery population and multiple validation populations, we tested the association of the MUC5B promoter variant rs35705950 in 620 patients with RA-ILD, 614 patients with RA without ILD, and 5448 unaffected controls. RESULTS: Analysis of the discovery population revealed an association of the minor allele of the MUC5B promoter variant with RA-ILD when patients with RA-ILD were compared with unaffected controls (adjusted odds ratio, 3.8; 95% confidence interval [CI], 2.8 to 5.2; P=9.7×10-17). The MUC5B promoter variant was also significantly overrepresented among patients with RA-ILD, as compared with unaffected controls, in an analysis of the multiethnic case series (adjusted odds ratio, 5.5; 95% CI, 4.2 to 7.3; P=4.7×10-35) and in a combined analysis of the discovery population and the multiethnic case series (adjusted odds ratio, 4.7; 95% CI, 3.9 to 5.8; P=1.3×10-49). In addition, the MUC5B promoter variant was associated with an increased risk of ILD among patients with RA (adjusted odds ratio in combined analysis, 3.1; 95% CI, 1.8 to 5.4; P=7.4×10-5), particularly among those with evidence of usual interstitial pneumonia on high-resolution computed tomography (adjusted odds ratio in combined analysis, 6.1; 95% CI, 2.9 to 13.1; P=2.5×10-6). However, no significant association with the MUC5B promoter variant was observed for the diagnosis of RA alone. CONCLUSIONS: We found that the MUC5B promoter variant was associated with RA-ILD and more specifically associated with evidence of usual interstitial pneumonia on imaging. (Funded by Société Française de Rhumatologie and others.).


Subject(s)
Arthritis, Rheumatoid/genetics , Gain of Function Mutation , Lung Diseases, Interstitial/genetics , Mucin-5B/genetics , Aged , Arthritis, Rheumatoid/complications , Female , Genetic Predisposition to Disease , Genotype , Humans , Idiopathic Pulmonary Fibrosis/genetics , Lung/chemistry , Lung/pathology , Lung Diseases, Interstitial/complications , Male , Middle Aged , Mucin-5B/analysis , Odds Ratio , Promoter Regions, Genetic
6.
Eur Respir J ; 57(2)2021 02.
Article in English | MEDLINE | ID: mdl-32646919

ABSTRACT

QUESTION ADDRESSED BY THE STUDY: Methotrexate (MTX) is a key anchor drug for rheumatoid arthritis (RA) management. Fibrotic interstitial lung disease (ILD) is a common complication of RA. Whether MTX exposure increases the risk of ILD in patients with RA is disputed. We aimed to evaluate the association of prior MTX use with development of RA-ILD. METHODS: Through a case-control study design with discovery and international replication samples, we examined the association of MTX exposure with ILD in 410 patients with chronic fibrotic ILD associated with RA (RA-ILD) and 673 patients with RA without ILD. Estimates were pooled over the different samples using meta-analysis techniques. RESULTS: Analysis of the discovery sample revealed an inverse relationship between MTX exposure and RA-ILD (adjusted OR 0.46, 95% CI 0.24-0.90; p=0.022), which was confirmed in the replication samples (pooled adjusted OR 0.39, 95% CI 0.19-0.79; p=0.009). The combined estimate using both the derivation and validation samples revealed an adjusted OR of 0.43 (95% CI 0.26-0.69; p=0.0006). MTX ever-users were less frequent among patients with RA-ILD compared to those without ILD, irrespective of chest high-resolution computed tomography pattern. In patients with RA-ILD, ILD detection was significantly delayed in MTX ever-users compared to never-users (11.4±10.4 years and 4.0±7.4 years, respectively; p<0.001). ANSWER TO THE QUESTION: Our results suggest that MTX use is not associated with an increased risk of RA-ILD in patients with RA, and that ILD was detected later in MTX-treated patients.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Lung Diseases, Interstitial , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/drug therapy , Case-Control Studies , Humans , Lung Diseases, Interstitial/chemically induced , Lung Diseases, Interstitial/drug therapy , Methotrexate/adverse effects
7.
Eur Respir J ; 53(2)2019 02.
Article in English | MEDLINE | ID: mdl-30523160

ABSTRACT

Regulator of telomere length 1 (RTEL1) mutations have been evidenced in 5-9% of familial pulmonary fibrosis; however, the phenotype of patients with interstitial lung disease (ILD) and RTEL1 mutations is poorly understood.Whole exome sequencing was performed in 252 probands with ILD and we included all patients with ILD and RTEL1 mutation. RTEL1 expression was evaluated by immunochemistry in the lungs of controls, as well as in RTEL1 and telomerase reverse transcriptase (TERT) mutation carriers.We identified 35 subjects from 17 families. Median age at diagnosis of ILD was 53.1 years (range 28.0-80.6). The most frequent pulmonary diagnoses were idiopathic pulmonary fibrosis (n=20, 57%), secondary ILD (n=7, 20%) and unclassifiable fibrosis or interstitial pneumonia with autoimmune features (n=7, 20%). The median transplant-free and overall survival periods were 39.2 months and 45.3 months, respectively. Forced vital capacity at diagnosis was the only factor associated with decreased transplant-free survival. Extra-pulmonary manifestations were less frequent as compared to other telomere-related gene mutation carriers. A systematic analysis of the literature identified 110 patients with ILD and RTEL1 mutations (including this series) and confirmed the heterogeneity of the pulmonary phenotype, the prevalence of non-idiopathic diseases and the low prevalence of extra-pulmonary manifestations.Immunohistochemistry showed that RTEL1 was expressed by bronchial and alveolar epithelial cells, as well as by alveolar macrophages and lymphocytes, but not by fibroblasts.


Subject(s)
DNA Helicases/genetics , Gene Expression Regulation , Lung Diseases, Interstitial/genetics , Lung Diseases/metabolism , Mutation , Adult , Aged , Aged, 80 and over , Exome , Female , Follow-Up Studies , Heterozygote , Humans , Lung Diseases/genetics , Male , Middle Aged , Pedigree , Phenotype , Sequence Analysis, DNA , Telomerase/genetics , Vital Capacity
8.
Respir Res ; 19(1): 74, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29695229

ABSTRACT

BACKGROUND: Whole lung lavage is the current standard therapy for pulmonary alveolar proteinosis (PAP) that is characterized by the alveolar accumulation of surfactant. Rituximab showed promising results in auto-immune PAP (aPAP) related to anti-GM-CSF antibody. METHODS: We aimed to assess efficacy of rituximab in aPAP in real life and all patients with aPAP in France that received rituximab were retrospectively analyzed. RESULTS: Thirteen patients were included. No patients showed improvement 6 months after treatment, but, 4 patients (30%) presented a significant decrease of alveolar-arterial difference in oxygen after 1 year. One patient received lung transplantation and one patient was lost of follow-up within one year. Although a spontaneous improvement cannot be excluded in these 4 patients, improvement was more frequent in patients naïve to prior specific therapy and with higher level of anti-GM-CSF antibodies evaluated by ELISA. No serious adverse event was evidenced. CONCLUSIONS: These data do not support rituximab as a second line therapy for patients with refractory aPAP.


Subject(s)
Immunologic Factors/therapeutic use , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/drug therapy , Rituximab/therapeutic use , Adult , Autoantibodies , Bronchoalveolar Lavage/trends , Cohort Studies , Female , Follow-Up Studies , France/epidemiology , Humans , Male , Middle Aged , Pulmonary Alveolar Proteinosis/epidemiology , Retrospective Studies
10.
Eur Respir J ; 49(5)2017 05.
Article in English | MEDLINE | ID: mdl-28495692

ABSTRACT

Despite its high prevalence and mortality, little is known about the pathogenesis of rheumatoid arthritis-associated interstitial lung disease (RA-ILD). Given that familial pulmonary fibrosis (FPF) and RA-ILD frequently share the usual pattern of interstitial pneumonia and common environmental risk factors, we hypothesised that the two diseases might share additional risk factors, including FPF-linked genes. Our aim was to identify coding mutations of FPF-risk genes associated with RA-ILD.We used whole exome sequencing (WES), followed by restricted analysis of a discrete number of FPF-linked genes and performed a burden test to assess the excess number of mutations in RA-ILD patients compared to controls.Among the 101 RA-ILD patients included, 12 (11.9%) had 13 WES-identified heterozygous mutations in the TERT, RTEL1, PARN or SFTPC coding regions. The burden test, based on 81 RA-ILD patients and 1010 controls of European ancestry, revealed an excess of TERT, RTEL1, PARN or SFTPC mutations in RA-ILD patients (OR 3.17, 95% CI 1.53-6.12; p=9.45×10-4). Telomeres were shorter in RA-ILD patients with a TERT, RTEL1 or PARN mutation than in controls (p=2.87×10-2).Our results support the contribution of FPF-linked genes to RA-ILD susceptibility.


Subject(s)
Arthritis, Rheumatoid/genetics , Genetic Predisposition to Disease , Lung Diseases, Interstitial/genetics , Pulmonary Fibrosis/genetics , Adult , Aged , Arthritis, Rheumatoid/complications , Case-Control Studies , DNA Helicases/genetics , Europe , Exome , Female , Genetic Association Studies , Heterozygote , Humans , Lung Diseases, Interstitial/complications , Male , Middle Aged , Mutation , Phenotype , Pulmonary Fibrosis/complications , Risk Factors , Sequence Analysis, DNA , Software , Telomerase/genetics
11.
Eur Respir J ; 48(6): 1721-1731, 2016 12.
Article in English | MEDLINE | ID: mdl-27836952

ABSTRACT

Telomerase reverse transcriptase (TERT) or telomerase RNA (TERC) gene mutation is a major monogenic cause of pulmonary fibrosis. Sequencing of TERT/TERC genes is proposed to patients with familial pulmonary fibrosis. Little is known about the possible predictors of this mutation and its impact on prognosis.We retrospectively analysed all the genetic diagnoses made between 2007-2014 in patients with pulmonary fibrosis. We evaluated the prevalence of TERT/TERC disease-associated variant (DAV), factors associated with a DAV, and the impact of the DAV on survival.237 patients with pulmonary fibrosis (153 with familial pulmonary fibrosis, 84 with telomere syndrome features without familial pulmonary fibrosis) were tested for TERT/TERC DAV. DAV was diagnosed in 40 patients (16.8%), including five with non-idiopathic interstitial pneumonia. Prevalence of TERT/TERC DAV did not significantly differ between patients with familial pulmonary fibrosis or with only telomere syndrome features (18.2% versus 16.4%). Young age, red blood cell macrocytosis, and low platelet count were associated with the presence of DAV; the probability of DAV was increased for patients 40-60 years. Transplant-free survival was lower with than without TERT/TERC DAV (4.2 versus 7.2 years; p=0.046).TERT/TERC DAV were associated with specific clinical and biological features and reduced transplant-free survival.


Subject(s)
Idiopathic Pulmonary Fibrosis/genetics , RNA/genetics , Telomerase/genetics , Telomere/genetics , Adult , Aged , Aged, 80 and over , Cause of Death , Female , France/epidemiology , Humans , Idiopathic Pulmonary Fibrosis/mortality , Logistic Models , Male , Middle Aged , Multivariate Analysis , Mutation , Retrospective Studies , Survival Analysis , Young Adult
12.
Am J Respir Cell Mol Biol ; 51(1): 11-25, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24450438

ABSTRACT

Idiopathic pulmonary fibrosis has been associated with the reactivation of developmental pathways, notably the Hedgehog-Glioma-associated oncogene homolog (GLI) pathway. In this study, we determined whether the Hedgehog pathway was activated in bleomycin-induced lung injury in mice, and whether targeting the Hedgehog-Gli pathway could decrease bleomycin-induced lung fibrosis. After intratracheal injection of bleomycin on Day 0, C57Bl6 mice received GDC-0449 (an inhibitor of Smoothened, the transducer of the pathway), or 2,2'-[[Dihydro-2-(4-pyridinyl)-1,3(2H,4H)-pyrimidinediyl]bis(methylene)]bis[N,N dimethylbenzenamine (GANT61; an inhibitor of GLI transcription factors in the nucleus), from Day 7 to Day 13. At Day 14, whole-lung homogenates were obtained for morphological analysis, assessment of cell apoptosis and proliferation, collagen quantification, and evaluation of profibrotic (transforming growth factor-ß, connective tissue growth factor, plasminogen activator inhibitor 1, vascular endothelial growth factor-A) and proinflammatory mediators (IL-1ß) expression. We showed that the Hedgehog pathway was activated in bleomycin-induced lung fibrosis on Day 14 after injury, with an increased lung expression of the ligand, Sonic Hedgehog, and with increased messenger RNA expression and nuclear localization of GLI1 and GLI2. Inhibition of Smoothened with GDC-0449 did not influence the development of bleomycin-induced lung fibrosis. By contrast, the inhibition of GLI activity with GANT61 decreased lung fibrosis and lung collagen accumulation, and promoted an antifibrotic and anti-inflammatory environment. Our results identify the hedgehog-Gli pathway as a profibrotic pathway in experimental fibrosis. Inhibition of the Hedgehog-Gli pathway at the level of GLI transcriptional activity could be a therapeutic option in fibrotic lung diseases.


Subject(s)
Anilides/pharmacology , Bleomycin/toxicity , Glioma/drug therapy , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/antagonists & inhibitors , Pulmonary Fibrosis/prevention & control , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Collagen/genetics , Collagen/metabolism , Fluorescent Antibody Technique , Glioma/metabolism , Glioma/pathology , Immunoenzyme Techniques , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Smoothened Receptor , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Zinc Finger Protein GLI1
13.
Rev Prat ; 64(7): 957-62, 2014 Sep.
Article in French | MEDLINE | ID: mdl-25362779

ABSTRACT

The etiological diagnosis of diffuse interstitial lung disease (ILD) is based on the integration of simple information gathered by interview, physical examination and discriminating arguments made by some investigations of the lower respiratory tract (HR-CT scan, pulmonary function tests, biological and bronchoalveolar lavage results). Obtaining the diagnosis of ILD is an important step but lung biopsy is rarely necessary: the key of ILD etiological diagnosis lies to obtain optimal information while imposing minimal patient aggression. The diagnostic approach is best when the case is subject to a multidisciplinary discussion between clinician, radiologist and pathologist experienced in the field of ILD.


Subject(s)
Diagnostic Techniques, Respiratory System , Lung Diseases, Interstitial/diagnosis , Bronchoscopy , Diagnosis, Differential , Humans , Lung Diseases, Interstitial/etiology , Respiratory Function Tests , Tomography, X-Ray Computed
15.
Am J Pathol ; 181(6): 2126-37, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23031257

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease of unknown cause. Key signaling developmental pathways are aberrantly expressed in IPF. The hedgehog pathway plays a key role during fetal lung development and may be involved in lung fibrogenesis. We determined the expression pattern of several Sonic hedgehog (SHH) pathway members in normal and IPF human lung biopsies and primary fibroblasts. The effect of hedgehog pathway inhibition was assayed by lung fibroblast proliferation and differentiation with and without transforming growth factor (TGF)-ß1. We showed that the hedgehog pathway was reactivated in the IPF lung. Importantly, we deciphered the cross talk between the hedgehog and TGF-ß pathway in human lung fibroblasts. TGF-ß1 modulated the expression of key components of the hedgehog pathway independent of Smoothened, the obligatory signal transducer of the pathway. Smoothened was required for TGF-ß1-induced myofibroblastic differentiation of control fibroblasts, but differentiation of IPF fibroblasts was partially resistant to Smoothened inhibition. Furthermore, functional hedgehog pathway machinery from the primary cilium, as well as GLI-dependent transcription in the nucleus, was required for the TGF-ß1 effects on normal and IPF fibroblasts during myofibroblastic differentiation. These data identify the GLI transcription factors as potential therapeutic targets in lung fibrosis.


Subject(s)
Cell Differentiation , Hedgehog Proteins/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Transforming Growth Factor beta/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cilia/drug effects , Cilia/pathology , Female , Gene Expression Regulation/drug effects , Hedgehog Proteins/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Immunohistochemistry , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Middle Aged , Models, Biological , Myofibroblasts/drug effects , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Veratrum Alkaloids/pharmacology
16.
Respir Med Res ; 83: 100948, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36630775

ABSTRACT

BACKGROUND: Since the latest 2017 French guidelines, knowledge about idiopathic pulmonary fibrosis has evolved considerably. METHODS: Practical guidelines were drafted on the initiative of the Coordinating Reference Center for Rare Pulmonary Diseases, led by the French Language Pulmonology Society (SPLF), by a coordinating group, a writing group, and a review group, with the involvement of the entire OrphaLung network, pulmonologists practicing in various settings, radiologists, pathologists, a general practitioner, a health manager, and a patient association. The method followed the "Clinical Practice Guidelines" process of the French National Authority for Health (HAS), including an online vote using a Likert scale. RESULTS: After a literature review, 54 guidelines were formulated, improved, and then validated by the working groups. These guidelines addressed multiple aspects of the disease: epidemiology, diagnostic procedures, quality criteria and interpretation of chest CT scans, lung biopsy indication and procedures, etiological workup, methods and indications for family screening and genetic testing, assessment of the functional impairment and prognosis, indication and use of antifibrotic agents, lung transplantation, management of symptoms, comorbidities and complications, treatment of chronic respiratory failure, diagnosis and management of acute exacerbations of fibrosis. CONCLUSION: These evidence-based guidelines are intended to guide the diagnosis and practical management of idiopathic pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung Transplantation , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/epidemiology , Idiopathic Pulmonary Fibrosis/therapy , Lung/pathology , Prognosis , Tomography, X-Ray Computed/methods
17.
Radiology ; 263(1): 253-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22332068

ABSTRACT

PURPOSE: To evaluate the tolerance and level of enhancement achievable after inhalation of stable krypton. MATERIALS AND METHODS: This study was approved by the institutional review board and the local ethics committee. Written informed consent was obtained from all subjects. The study was planned as a Fleming two-stage design, enabling one to assess the effectiveness of a newer treatment or technique on a small number of patients. At the end of each stage, the results are computed, and the trial can be stopped if the effectiveness is less than a minimum success rate or greater than an expected success rate. After informed consent was obtained, a total of 32 patients (ie, two successive series of 16 patients each) with severe emphysema underwent a dual-source, dual-energy chest computed tomographic (CT) examination after inhalation of a mixture of stable krypton (80%) and oxygen (20%), with reconstruction of diagnostic and ventilation images. For each patient, two regions of interest were selected on a diagnostic image, one in a region of severe emphysema (presumed to be poorly ventilated or not ventilated) and a second one in a region devoid of structural abnormalities (presumed to be normally ventilated), with measurements of attenuation values on the corresponding ventilation image. RESULTS: All examinations were successfully performed, without adverse effects. Differences in attenuation between normal lung and emphysematous areas were found in 28 patients (88%; 95% confidence interval: 71%, 96.5%). The maximal level of attenuation within normal lung was 18.5 HU. Krypton attenuation difference between normal and emphysematous lung was significant, with a median value of 51.8% (P < .001). CONCLUSION: The level of enhancement after inhalation of krypton and its excellent clinical tolerance makes this gas eligible for ventilation CT examinations.


Subject(s)
Krypton , Pulmonary Disease, Chronic Obstructive/diagnostic imaging , Tomography, X-Ray Computed/methods , Female , Humans , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/physiopathology , Radiographic Image Interpretation, Computer-Assisted , Respiratory Function Tests , Statistics, Nonparametric
18.
Med Sci (Paris) ; 38(6-7): 579-584, 2022.
Article in French | MEDLINE | ID: mdl-35766856

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive and fatal lung disease of unknown origin. It is characterized by aberrant scarring of the alveolar epithelium leading to an accumulation of extracellular matrix (ECM). Fibroblastic foci, consisting of fibroblasts and myofibroblasts, are responsible for the excessive production of ECM. The two therapeutic molecules available on the market to date only allow to slow down the evolution of the disease. In this review, we present the mechanisms involved in the progression of the disease, its treatments and the study models.


Title: La fibrose pulmonaire idiopathique. Abstract: La fibrose pulmonaire idiopathique (FPI) est une maladie pulmonaire chronique, évolutive et mortelle dont l'origine est inconnue. Elle se caractérise par une cicatrisation aberrante de l'épithélium alvéolaire aboutissant à une accumulation de matrice extracellulaire (MEC). Les foyers fibroblastiques, constitués de fibroblastes et de myofibroblastes, sont responsables de la production excessive de MEC. Les deux seules molécules thérapeutiques disponibles sur le marché permettent seulement de ralentir l'évolution de la maladie. Dans cette revue, nous présentons les mécanismes impliqués dans la progression de la maladie, ses traitements et les modèles d'étude.


Subject(s)
Idiopathic Pulmonary Fibrosis , Extracellular Matrix , Fibroblasts , Humans , Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/therapy , Lung , Myofibroblasts
19.
Lancet Respir Med ; 10(1): 26-34, 2022 01.
Article in English | MEDLINE | ID: mdl-34506761

ABSTRACT

BACKGROUND: The use of cyclophosphamide in patients with acute exacerbation of idiopathic pulmonary fibrosis (IPF) is unknown. Our study was designed to evaluate the efficacy and safety of four cyclophosphamide pulses in addition to high-dose methylprednisolone in this population. METHODS: In this double-blind, placebo-controlled trial done in 35 departments across 31 hospitals in France, adult patients (≥18 years) with acute exacerbation of IPF and those with suspected acute exacerbation of IPF were randomly assigned in a 1:1 ratio using a web-based system to receive either intravenous pulses of cyclophosphamide (600 mg/m2) plus uromitexan as haemorrhagic cystitis prophylaxis (200 mg/m2) at the time of cyclophosphamide administration and then again, 4 h later, or placebo at days 0, 15, 30, and 60. Random assignment was stratified according to the severity of IPF and was block-balanced with variable block sizes of four or six patients. Patients receiving mechanical ventilation, with active infection, with active cancer, or who were registered on the lung transplant waiting list were excluded. All patients received standardised high-dose glucocorticoids. The investigators, patients, and the sponsor were masked to the treatment assignments. The primary endpoint was 3-month all-cause mortality, analysed by a χ2 test adhering to an intention-to-treat principle. The trial is now complete and registered with ClinicalTrials.gov, NCT02460588. FINDINGS: Between Jan 22, 2016, and July 19, 2018, 183 patients were assessed for eligibility, of whom 120 patients were randomly assigned and 119 patients (62 [52%] with severe IPF) received at least one dose of cyclophosphamide (n=60) or placebo (n=59), all of whom were included in the intention-to-treat analysis. The 3-month all-cause mortality was 45% (27/60) in patients given cyclophosphamide compared with 31% (18/59) in the placebo group (difference 14·5% [95% CI -3·1 to 31·6]; p=0·10). Similar results were found after adjustment by IPF severity (odds ratio [OR] 1·89 [95% CI 0·89-4·04]). The risk of death at 3 months, independent of the treatment received, was higher with severe than non-severe IPF (OR 2·62 [1·12-6·12]) and was lower with the use of antifibrotic therapy (OR 0·33 [0·13-0·82]). Adverse events were similar between groups by 6 months (25 [42%] in the cyclophosphamide group vs 30 [51%] in the placebo group) and their proportion, including infections, did not differ. Overall infection was the main adverse event and occurred in 20 (33%) of 60 patients in the cyclophosphamide group versus 21 (36%) of 59 patients in the placebo group. INTERPRETATION: In patients with acute exacerbation of IPF, adding intravenous cyclophosphamide pulses to glucocorticoids increased 3-month mortality. These findings provide evidence against the use of intravenous cyclophosphamide in such patients. FUNDING: Programme Hospitalier de Recherche Clinique of the French Ministry of Health (PHRC 2014-502), Roche Pharmaceuticals.


Subject(s)
Glucocorticoids , Idiopathic Pulmonary Fibrosis , Adult , Cyclophosphamide/adverse effects , Double-Blind Method , Glucocorticoids/adverse effects , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Treatment Outcome
20.
Respiration ; 82(6): 501-8, 2011.
Article in English | MEDLINE | ID: mdl-21934275

ABSTRACT

BACKGROUND: Patients with sarcoidosis frequently exhibit exertional dyspnea. Although the functional limitation might be related to ventilatory problems, some patients exhibited exertional dyspnea without significant abnormalities of pulmonary function tests (PFT). Therefore, the mechanisms responsible for exercise intolerance remain unclear. OBJECTIVES: The aim of this retrospective study was to determine the mechanisms responsible for exercise intolerance. METHODS: Cardiopulmonary exercise testing (CPET) was performed in 157 dyspneic sarcoid patients (stage I: 29; stage II-III: 95; stage IV: 33) and VO2 peak was correlated with radiological stage and resting PFT. RESULTS: VO2 peak was decreased in 73% patients, did not differ according to radiological stage and was correlated with VC, FVC, FEV1, TLC and DLCO. FVC was the major significant predictor of VO2 peak explaining 17% of VO2 variation. Among CPET variables, peak heart rate and VE/VO2 at ventilatory threshold explained 22% of VO2 alteration in stage I and stage II-III, and 9% in stage IV. In stage IV, V(D)/V(T) peak explained 41% of VO2 alteration. CONCLUSION: In conclusion, in the lower stages circulatory impairment and impaired heart rate response to exercise are involved in the exercise capacity limitation, whereas in stage IV the ventilatory and gas exchange impairment may be more important. CPET must be performed to accurately characterize the mechanisms responsible for exercise limitation and help the clinician in the management of the disease.


Subject(s)
Anaerobic Threshold , Dyspnea/metabolism , Exercise Tolerance , Oxygen/metabolism , Pulmonary Gas Exchange , Sarcoidosis/metabolism , Adult , Aged , Dyspnea/physiopathology , Exercise Test/methods , Female , Forced Expiratory Volume , Humans , Male , Middle Aged , Predictive Value of Tests , Rest , Retrospective Studies , Sarcoidosis/physiopathology , Severity of Illness Index , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL