117 results on '"Philip L Molyneaux"'
Search Results
2. Residual Lung Abnormalities after COVID-19 Hospitalization: Interim Analysis of the UKILD Post–COVID-19 Study
- Author
-
Iain Stewart, Joseph Jacob, Peter M. George, Philip L. Molyneaux, Joanna C. Porter, Richard J. Allen, Shahab Aslani, J. Kenneth Baillie, Shaney L. Barratt, Paul Beirne, Stephen M. Bianchi, John F. Blaikley, James D. Chalmers, Rachel C. Chambers, Nazia Chadhuri, Christopher Coleman, Guilhem Collier, Emma K. Denneny, Annemarie Docherty, Omer Elneima, Rachael A. Evans, Laura Fabbri, Michael A. Gibbons, Fergus V. Gleeson, Bibek Gooptu, Neil J. Greening, Beatriz Guillen Guio, Ian P. Hall, Neil A. Hanley, Victoria Harris, Ewen M. Harrison, Melissa Heightman, Toby E. Hillman, Alex Horsley, Linzy Houchen-Wolloff, Ian Jarrold, Simon R. Johnson, Mark G. Jones, Fasihul Khan, Rod Lawson, Olivia Leavy, Nazir Lone, Michael Marks, Hamish McAuley, Puja Mehta, Dhruv Parekh, Karen Piper Hanley, Manuela Platé, John Pearl, Krisnah Poinasamy, Jennifer K. Quint, Betty Raman, Matthew Richardson, Pilar Rivera-Ortega, Laura Saunders, Ruth Saunders, Malcolm G. Semple, Marco Sereno, Aarti Shikotra, A. John Simpson, Amisha Singapuri, David J. F. Smith, Mark Spears, Lisa G. Spencer, Stefan Stanel, David R. Thickett, A. A. Roger Thompson, Mathew Thorpe, Simon L. F. Walsh, Samantha Walker, Nicholas David Weatherley, Mark E. Weeks, Jim M. Wild, Dan G. Wootton, Chris E. Brightling, Ling-Pei Ho, Louise V. Wain, and Gisli R. Jenkins
- Subjects
Pulmonary and Respiratory Medicine ,Critical Care and Intensive Care Medicine - Published
- 2023
- Full Text
- View/download PDF
3. Phosphodiesterase 5 inhibitor treatment and survival in interstitial lung disease pulmonary hypertension: A Bayesian retrospective observational cohort study
- Author
-
Timothy J. W. Dawes, Colm McCabe, Konstantinos Dimopoulos, Iain Stewart, Simon Bax, Carl Harries, Chinthaka B. Samaranayake, Aleksander Kempny, Philip L. Molyneaux, Samuel Seitler, Thomas Semple, Wei Li, Peter M. George, Vasileios Kouranos, Felix Chua, Elisabetta A. Renzoni, Maria Kokosi, Gisli Jenkins, Athol U. Wells, Stephen J. Wort, and Laura C. Price
- Subjects
Pulmonary and Respiratory Medicine - Abstract
Background and Objective Pulmonary hypertension is a life-limiting complication of interstitial lung disease (ILD-PH). We investigated whether treatment with phosphodiesterase 5 inhibitors (PDE5i) in patients with ILD-PH was associated with improved survival. Methods Consecutive incident patients with ILD-PH and right heart catheterisation, echocardiography and spirometry data were followed from diagnosis to death, transplantation or censoring with all follow-up and survival data modelled by Bayesian methods. Results The diagnoses in 128 patients were idiopathic pulmonary fibrosis (n = 74, 58%), hypersensitivity pneumonitis (n = 17, 13%), non-specific interstitial pneumonia (n = 12, 9%), undifferentiated ILD (n = 8, 6%) and other lung diseases (n = 17, 13%). Final outcomes were death (n = 106, 83%), transplantation (n = 9, 7%) and censoring (n = 13, 10%). Patients treated with PDE5i (n = 50, 39%) had higher mean pulmonary artery pressure (median 38 mm Hg [interquartile range, IQR: 34, 43] vs. 35 mm Hg [IQR: 31, 38], p = 0.07) and percentage predicted forced vital capacity (FVC; median 57% [IQR: 51, 73] vs. 52% [IQR: 45, 66], p=0.08) though differences did not reach significance. Patients treated with PDE5i survived longer than untreated patients (median 2.18 years [95% CI: 1.43, 3.04] vs. 0.94 years [0.69, 1.51], p = 0.003) independent of all other prognostic markers by Bayesian joint-modelling (HR 0.39, 95% CI: 0.23, 0.59, p
- Published
- 2022
- Full Text
- View/download PDF
4. Microbiome, Metabolism, and Immunoregulation of Asthma: An American Thoracic Society and National Institute of Allergy and Infectious Diseases Workshop Report
- Author
-
Ariangela J. Kozik, Fernando Holguin, Leopoldo N. Segal, Talal A. Chatila, Anne E. Dixon, James E. Gern, Catherine Lozupone, Nicholas Lukacs, Carey Lumeng, Philip L. Molyneaux, Nichole Reisdorph, Ivan Vujkovic-Cvijin, Alkis Togias, and Yvonne J. Huang
- Subjects
Pulmonary and Respiratory Medicine ,Microbiota ,Clinical Biochemistry ,Immunity ,Cell Biology ,Asthma ,United States ,Mice ,National Institute of Allergy and Infectious Diseases (U.S.) ,Hypersensitivity ,Animals ,Humans ,Child ,Molecular Biology - Abstract
This report presents the proceedings from a workshop titled "Microbiome, Metabolism and Immunoregulation of Asthma" that was held virtually May 13 and 14, 2021. The workshop was jointly sponsored by the American Thoracic Society (Assembly on Allergy, Immunology, and Inflammation) and the National Institute of Allergy and Infectious Diseases. It convened an interdisciplinary group of experts with backgrounds in asthma immunology, microbiome science, metabolomics, computational biology, and translational pulmonary research. The main purpose was to identify key scientific gaps and needs to further advance research on microbial and metabolic mechanisms that may contribute to variable immune responses and disease heterogeneity in asthma. Discussions were structured around several topics, including 1) immune and microbial mechanisms of asthma pathogenesis in murine models, 2) the role of microbes in pediatric asthma exacerbations, 3) dysregulated metabolic pathways in asthma associated with obesity, 4) metabolism effects on macrophage function in adipose tissue and the lungs, 5) computational approaches to dissect microbiome-metabolite links, and 6) potential confounders of microbiome-disease associations in human studies. This report summarizes the major points of discussion, which included identification of specific knowledge gaps, challenges, and suggested directions for future research. These include questions surrounding mechanisms by which microbiota and metabolites shape host health versus an allergic or asthmatic state; direct and indirect influences of other biological factors, exposures, and comorbidities on these interactions; and ongoing technical and analytical gaps for clinical translation.
- Published
- 2022
- Full Text
- View/download PDF
5. Wearable In-Ear PPG: Detailed Respiratory Variations Enable Classification of COPD
- Author
-
Harry J. Davies, Patrik Bachtiger, Ian Williams, Philip L. Molyneaux, Nicholas S. Peters, and Danilo P. Mandic
- Subjects
0906 Electrical and Electronic Engineering ,Pulmonary Disease, Chronic Obstructive ,Wearable Electronic Devices ,0903 Biomedical Engineering ,Respiratory Rate ,Heart Rate ,otorhinolaryngologic diseases ,0801 Artificial Intelligence and Image Processing ,Biomedical Engineering ,Humans ,Signal Processing, Computer-Assisted ,sense organs ,Photoplethysmography - Abstract
An ability to extract detailed spirometry-like breath-ing waveforms from wearable sensors promises to greatly improve respiratory health monitoring. Photoplethysmography (PPG) has been researched in depth for estimation of respiration rate, given that it varies with respiration through overall intensity, pulse amplitude and pulse interval. We compare and contrast the extraction of these three respiratory modes from both the ear canal and finger and show a marked improvement in the respiratory power for respiration induced intensity variations and pulse amplitude variations when recording from the ear canal. We next employ a data driven multi-scale method, noise assisted multivariate empirical mode decomposition (NA-MEMD), which allows for simultaneous analysis of all three respiratory modes to extract detailed respiratory waveforms from in-ear PPG. For rigour, we considered in-ear PPG recordings from healthy subjects, both older and young, patients with chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) and healthy subjects with artificially obstructed breathing. Specific in-ear PPG waveform changes are observed for COPD, such as a decreased inspiratory duty cycle and an increased inspiratory magnitude, when compared with expiratory magnitude. These differences are used to classify COPD from healthy and IPF waveforms with a sensitivity of 87% and an overall accuracy of 92%. Our findings indicate the promise of in-ear PPG for COPD screening and unobtrusive respiratory monitoring in ambulatory scenarios and in consumer wearables.
- Published
- 2022
- Full Text
- View/download PDF
6. Rare and Common Variants in KIF15 Contribute to Genetic Risk of Idiopathic Pulmonary Fibrosis
- Author
-
David Zhang, Gundula Povysil, Philippe H. Kobeissy, Qi Li, Binhan Wang, Mason Amelotte, Hager Jaouadi, Chad A. Newton, Toby M. Maher, Philip L. Molyneaux, Imre Noth, Fernando J. Martinez, Ganesh Raghu, Jamie L. Todd, Scott M. Palmer, Carolina Haefliger, Adam Platt, Slavé Petrovski, Joseph A. Garcia, David B. Goldstein, Christine Kim Garcia, and Action for Pulmonary Fibrosis
- Subjects
Pulmonary and Respiratory Medicine ,Respiratory System ,Kinesins ,spindle ,Telomere ,Critical Care and Intensive Care Medicine ,Idiopathic Pulmonary Fibrosis ,IPF ,KIF15 ,cell proliferation ,Genetics ,Humans ,Exome ,Telomerase ,11 Medical and Health Sciences - Abstract
RATIONALE: Genetic studies of Idiopathic Pulmonary Fibrosis (IPF) have improved our understanding of this disease, but not all causal loci have been identified. OBJECTIVE: To identify genes enriched with rare deleterious variants in IPF and familial pulmonary fibrosis. METHODS: We performed gene burden analysis of whole exome data, tested single variants for disease association, conducted KIF15 functional studies, and examined human lung single cell RNA sequencing data. MEASUREMENT AND MAIN RESULTS: Gene burden analysis of 1,725 cases and 23,509 controls identified heterozygous rare deleterious variants in KIF15, a kinesin involved in spindle separation during mitosis, and three telomere-related genes (TERT, RTEL1, PARN). KIF15 was implicated in autosomal dominant models of rare deleterious variants (OR 4.9 [95%CI 2.7, 8.8] P=2.55x10-7) and rare protein-truncating variants (OR 7.6 [3.3, 17.1], P=8.12x10-7). Meta-analysis of the discovery and replication cohorts, including 2,966 cases and 29,817 controls, confirm the involvement of KIF15, plus the three telomere-related genes. A common variant within a KIF15 intron (rs74341405, OR 1.6 [1.4, 1.9], P=5.63x10-10) is associated with IPF risk, confirming a prior report. Lymphoblastoid cells from individuals heterozygous for the common variant have decreased KIF15 and reduced rates of cell growth. Cell proliferation is dependent on KIF15 in the presence of an inhibitor of Eg5/KIF11, which has partially redundant function. KIF15 is expressed specifically in replicating human lung cells, and shows diminished expression in replicating epithelial cells of IPF patients. CONCLUSIONS: Both rare deleterious variants and common variants in KIF15 link a non-telomerase pathway of cell proliferation with IPF susceptibility.
- Published
- 2022
- Full Text
- View/download PDF
7. Nocturnal hypoxemia associates with symptom progression and mortality in patients with progressive fibrotic interstitial lung disease
- Author
-
Katherine J. Myall, Alex G. West, Jennifer L. Martinovic, Jodie L. Lam, Diana Roque, Zhe Wu, Toby M. Maher, Philip L. Molyneaux, Eui-Sik Suh, and Brian D. Kent
- Subjects
Pulmonary and Respiratory Medicine ,Cardiology and Cardiovascular Medicine ,Critical Care and Intensive Care Medicine - Published
- 2023
- Full Text
- View/download PDF
8. Genome-wide Enrichment of TERT Rare Variants in Idiopathic Pulmonary Fibrosis Patients of Latino Ancestry
- Author
-
David Zhang, Qi Li, Gundula Povysil, Chad A. Newton, Toby M. Maher, Philip L. Molyneaux, Imre Noth, Fernando J. Martinez, Ganesh Raghu, Jamie L. Todd, Scott M. Palmer, Adam Platt, Slavé Petrovski, David B. Goldstein, Christine Kim Garcia, and Action for Pulmonary Fibrosis
- Subjects
Latino ,Pulmonary and Respiratory Medicine ,non-European ancestry ,Respiratory System ,Hispanic or Latino ,Critical Care and Intensive Care Medicine ,Idiopathic Pulmonary Fibrosis ,IPF ,Genetics ,Humans ,Genetic Predisposition to Disease ,Telomerase ,11 Medical and Health Sciences ,Genome-Wide Association Study - Abstract
Genome-wide rare variant studies of IPF patients of non-European ancestry have been understudied. Here, we evaluate the enrichment of rare genetic variants of 241 unrelated non-European cases, representing individuals of Latino, African, South Asian, East Asian, and Other Admixed ancestry. Gene burden analysis of deleterious rare (protein-truncating and missense) variants demonstrate an excess of TERT rare damaging variants (OR 67.1, 95% CI [23.1, 195.0], P = 9.4 x 10-14) in non-European subjects. Analysis by ancestry demonstrated an excess of rare, damaging TERT variants in the Latino subgroup (OR 80.9, 95% CI [17.3, 383.8], P = 2.6 x 10-8). Although the non-European group did not show enrichment of PARN, RTEL1, and KIF15 rare deleterious variants, these groups all showed a trend in the same direction as the European ancestry group. For TERT and KIF15, the inclusion of IPF patients of non-European ancestry led to a higher odds ratios and increased evidence in favor of rare deleterious variant contributions, thus demonstrating the increased power of multi-ethnic studies over single-ethnicity studies. To our knowledge, this is the first study that confirms the involvement of rare deleterious TERT variants for IPF patients of Latino and non-European ancestry. To better understand the genetic underpinnings of IPF patients of all ancestries, additional work will be needed to broaden patient recruitment to normalize imbalances.
- Published
- 2022
- Full Text
- View/download PDF
9. Cluster analysis of transcriptomic datasets to identify endotypes of idiopathic pulmonary fibrosis
- Author
-
Luke M Kraven, Adam R Taylor, Philip L Molyneaux, Toby M Maher, John E McDonough, Marco Mura, Ivana V Yang, David A Schwartz, Yong Huang, Imre Noth, Shwu Fan Ma, Astrid J Yeo, William A Fahy, R Gisli Jenkins, and Louise V Wain
- Subjects
Pulmonary and Respiratory Medicine ,respiratory system ,Article ,respiratory tract diseases - Abstract
BackgroundConsiderable clinical heterogeneity in idiopathic pulmonary fibrosis (IPF) suggests the existence of multiple disease endotypes. Identifying these endotypes would improve our understanding of the pathogenesis of IPF and could allow for a biomarker-driven personalised medicine approach. We aimed to identify clinically distinct groups of patients with IPF that could represent distinct disease endotypes.MethodsWe co-normalised, pooled and clustered three publicly available blood transcriptomic datasets (total 220 IPF cases). We compared clinical traits across clusters and used gene enrichment analysis to identify biological pathways and processes that were over-represented among the genes that were differentially expressed across clusters. A gene-based classifier was developed and validated using three additional independent datasets (total 194 IPF cases).FindingsWe identified three clusters of patients with IPF with statistically significant differences in lung function (p=0.009) and mortality (p=0.009) between groups. Gene enrichment analysis implicated mitochondrial homeostasis, apoptosis, cell cycle and innate and adaptive immunity in the pathogenesis underlying these groups. We developed and validated a 13-gene cluster classifier that predicted mortality in IPF (high-risk clusters vs low-risk cluster: HR 4.25, 95% CI 2.14 to 8.46, p=3.7×10−5).InterpretationWe have identified blood gene expression signatures capable of discerning groups of patients with IPF with significant differences in survival. These clusters could be representative of distinct pathophysiological states, which would support the theory of multiple endotypes of IPF. Although more work must be done to confirm the existence of these endotypes, our classifier could be a useful tool in patient stratification and outcome prediction in IPF.
- Published
- 2022
- Full Text
- View/download PDF
10. Pulmonary Rehabilitation in Idiopathic Pulmonary Fibrosis and COPD
- Author
-
Jessica A. Walsh, Peter M. George, Oliver Polgar, Philip L. Molyneaux, Susie Schofield, Ruth E Barker, William D.-C. Man, Claire M. Nolan, Toby M. Maher, Suhani Patel, and Karen A. Ingram
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,COPD ,Vital capacity ,business.industry ,medicine.medical_treatment ,Hazard ratio ,Interstitial lung disease ,respiratory system ,Critical Care and Intensive Care Medicine ,medicine.disease ,respiratory tract diseases ,Idiopathic pulmonary fibrosis ,FEV1/FVC ratio ,Internal medicine ,Cohort ,Medicine ,Pulmonary rehabilitation ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background The adherence to and clinical efficacy of pulmonary rehabilitation in idiopathic pulmonary fibrosis (IPF), particularly in comparison to people with chronic obstructive pulmonary disease (COPD), remains uncertain. The objectives of this real-world study were to compare the responses of patients with IPF with a matched group of patients with COPD undergoing the same supervised, outpatient pulmonary rehabilitation program, and to determine whether pulmonary rehabilitation is associated with survival in IPF. Research question Do people with IPF improve to the same extent with pulmonary rehabilitation as a matched group of individuals with COPD, and are non-completion of and/or non-response to pulmonary rehabilitation associated with one-year all-cause mortality in IPF? Study design and methods Using propensity score matching, 163 patients with IPF were matched 1:1 with a control group of 163 patients with COPD referred to pulmonary rehabilitation. We compared between-group pulmonary rehabilitation completion rates and response. Survival status in the IPF cohort was recorded over one-year following pulmonary rehabilitation discharge. Cox proportional-hazards regression explored the association between pulmonary rehabilitation status and all-cause mortality. Results Similar pulmonary rehabilitation completion rates (IPF: 69%; COPD: 63%; p=0.24) and improvements in exercise response were observed in both groups with no significant mean (95% confidence interval (CI)) between-group differences in incremental shuttle walk (ISW) change (2 (-18 to 22) meters). Pulmonary rehabilitation non-completion (hazard ratio (HR) (95%CI) 5.62 (2.24 to 14.08)) and non-response (HR (95%CI) 3.91 (1.54 to 9.93)) were independently associated with increased one-year all-cause mortality in IPF. Interpretation Compared with a matched group of patients with COPD, this real-word study demonstrates that patients with IPF have similar completion rates and magnitude of response to pulmonary rehabilitation. In IPF, non-completion of and non-response to pulmonary rehabilitation were associated with increased all-cause mortality. These data reinforce the benefits of pulmonary rehabilitation in patients with IPF.
- Published
- 2022
- Full Text
- View/download PDF
11. Short‐term lung function changes predict mortality in patients with fibrotic hypersensitivity pneumonitis
- Author
-
Claudio Macaluso, Cristina Boccabella, Maria Kokosi, Nishanth Sivarasan, Vasilis Kouranos, Peter M. George, George Margaritopoulos, Philip L. Molyneaux, Felix Chua, Toby M. Maher, Gisli R. Jenkins, Andrew G. Nicholson, Sujal R. Desai, Anand Devaraj, Athol U. Wells, Elisabetta A. Renzoni, Carmel J. W. Stock, and Action for Pulmonary Fibrosis
- Subjects
Pulmonary and Respiratory Medicine ,short-term lung function change ,Science & Technology ,Respiratory System ,fibrotic hypersensitivity pneumonitis ,ANTIGEN ,predictor ,respiratory system ,mortality ,FVC ,respiratory tract diseases ,DLCO ,SURVIVAL ,PATTERNS ,IDIOPATHIC PULMONARY-FIBROSIS ,Life Sciences & Biomedicine ,FORCED VITAL CAPACITY ,11 Medical and Health Sciences - Abstract
Background and objective A proportion of patients with fibrotic hypersensitivity pneumonitis (fHP) follow a progressive disease course despite immunosuppressive treatment. Little is known about predictors of mortality in fHP. We aimed to investigate the impact of short-term lung function changes in fHP on mortality. Methods Baseline demographics for 145 consecutive patients with a multi-disciplinary team diagnosis of fHP, as well as baseline and 1-year follow-up of lung function, baseline echocardiographic findings, bronchoalveolar lavage (BAL) cellularity and all-cause mortality were recorded. Changes in forced vital capacity (FVC) ≥ 5% and ≥10%, and diffusion capacity of the lung for carbon monoxide (DLCO) ≥ 10% and ≥15% at 1 year were calculated. Cox proportional hazards analysis was performed to test for associations with mortality. Results Baseline lung function severity, age, presence of honeycombing on computed tomography (CT) and echocardiographic pulmonary arterial systolic pressure (PASP) ≥ 40 mm Hg were associated with early mortality, while BAL lymphocytosis was associated with improved survival. A decline in FVC ≥ 5% (hazard ratio [HR]: 3.10, 95% CI: 2.00–4.81, p
- Published
- 2022
- Full Text
- View/download PDF
12. Adventitial mast cell activation disrupts endothelial cell/pericyte interactions during early life allergic asthma
- Author
-
Régis Joulia, Franz Puttur, Helen Stölting, Lewis J. Entwistle, Anna Voitovich, May Al-Sahaf, Katie Bonner, Elizabeth Scotney, Philip L Molyneaux, Richard J Hewitt, Simone A. Walker, Laura Yates, Sejal Saglani, and Clare M. Lloyd
- Abstract
SummaryAllergic asthma generally starts during early life and is linked to significant tissue remodelling and lung dysfunction. Although angiogenesis is a feature of the disrupted airway, the impact of allergic asthma on the pulmonary microcirculation during early life is unknown. Here, using quantitative imaging in precision-cut lung slices (PCLS), we report that exposure of neonatal mice to house dust mite (HDM) extract disrupts endothelial cell/pericyte interactions in adventitial areas. Central to the blood vessel structure, the loss of pericyte coverage was driven by mast cell (MCs) proteases, such as tryptase, that can induce pericyte retraction and loss of the critical adhesion molecule N-Cadherin. Furthermore, spatial transcriptomics of paediatric asthmatic endobronchial biopsies revealed intense remodelling associated with increased expression of MC proteases in regions enriched in blood vessels. These data provide previously unappreciated insights into the pathophysiology of allergic asthma with potential long-term vascular defects.
- Published
- 2023
- Full Text
- View/download PDF
13. Lung cancer screening provides an opportunity for early diagnosis and treatment of interstitial lung disease
- Author
-
Richard J Hewitt, Emily C Bartlett, Rea Ganatra, Haroun Butt, Vasilis Kouranos, Felix Chua, Maria Kokosi, Philip L Molyneaux, Sujal R Desai, Athol U Wells, R Gisli Jenkins, Elisabetta A Renzoni, Samuel V Kemp, Anand Devaraj, and Peter M George
- Subjects
Pulmonary and Respiratory Medicine ,Lung Neoplasms ,Humans ,Lung Diseases, Interstitial ,Tomography, X-Ray Computed ,Lung ,Early Detection of Cancer ,Idiopathic Pulmonary Fibrosis ,Retrospective Studies - Abstract
Interstitial lung abnormalities (ILA) can be incidentally detected in patients undergoing low-dose CT screening for lung cancer. In this retrospective study, we explore the downstream impact of ILA detection on interstitial lung disease (ILD) diagnosis and treatment. Using a targeted approach in a lung cancer screening programme, the rate of de novo ILD diagnosis was 1.5%. The extent of abnormality on CT and severity of lung function impairment, but not symptoms were the most important factors in differentiating ILA from ILD. Disease modifying therapies were commenced in 39% of ILD cases, the majority being antifibrotic therapy for idiopathic pulmonary fibrosis.
- Published
- 2022
- Full Text
- View/download PDF
14. Lung extracellular matrix modulates KRT5+basal cell activity in pulmonary fibrosis
- Author
-
Richard J. Hewitt, Franz Puttur, David C. A. Gaboriau, Frédéric Fercoq, Maryline Fresquet, William J. Traves, Laura L. Yates, Simone A. Walker, Philip L. Molyneaux, Samuel V. Kemp, Andrew G. Nicholson, Alexandra Rice, Rachel Lennon, Leo M. Carlin, Adam J. Byrne, Toby M. Maher, and Clare M. Lloyd
- Abstract
Aberrant expansion of KRT5+basal cells in the distal lung accompanies progressive alveolar epithelial cell loss and tissue remodelling during fibrogenesis in idiopathic pulmonary fibrosis (IPF). The mechanisms determining activity of KRT5+cells in IPF have not been delineated. Here, we reveal a potential mechanism by which KRT5+cells migrate within the fibrotic lung, navigating regional differences in collagen topography.In vitro, KRT5+cell migratory characteristics and expression of remodelling genes are modulated by extracellular matrix (ECM) composition and organisation. Mass spectrometry-based proteomics revealed compositional differences in ECM components secreted by primary human lung fibroblasts (HLF) from IPF patients compared to controls. Over-expression of ECM glycoprotein, Secreted Protein Acidic and Cysteine Rich (SPARC) in the IPF HLF matrix restricts KRT5+cell migrationin vitro. Together, our findings demonstrate how changes to the ECM in IPF directly influence KRT5+cell behaviour and function contributing to remodelling events in the fibrotic niche.
- Published
- 2022
- Full Text
- View/download PDF
15. How to Understand a Revolution: Guts, Lungs, and Bronchiectasis
- Author
-
Callie M. Drohan, Philip L. Molyneaux, and Robert P. Dickson
- Subjects
Pulmonary and Respiratory Medicine ,Critical Care and Intensive Care Medicine - Abstract
Suppose a scholar seeks to understand the American Revolutionary War: its causes, consequences, and what lessons we may learn from it. They begin by reading biographies of key participants, but soon realize the scope of these books is too narrow. Broadening the scope to study demography (e.g., census reports, immigration records) helps them understand not merely the leaders of the revolution but also the people they led. Yet even this isn’t enough to understand the colonists’ activities, so the scope is broadened further to interactions: in governance (political science), in the marketplace (economics), and in culture (sociology). Finally, our historian realizes they cannot understand the events of Boston, Philadelphia, and Yorktown without also studying England: the leaders, people, and interactions that influenced a revolution from across an ocean.
- Published
- 2022
16. Monocyte Count as a Prognostic Biomarker in Patients with Idiopathic Pulmonary Fibrosis
- Author
-
Klaus Uwe Kirchgaessler, Toby M. Maher, Mark Atwood, Philip L. Molyneaux, Derek Weycker, Argyrios Tzouvelekis, Justin M. Oldham, Michael Kreuter, Joyce S. Lee, and Action for Pulmonary Fibrosis
- Subjects
Male ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Respiratory System ,Critical Care and Intensive Care Medicine ,Placebo ,Risk Assessment ,Autoimmune Disease ,Medical and Health Sciences ,Gastroenterology ,Monocytes ,03 medical and health sciences ,Idiopathic pulmonary fibrosis ,FEV1/FVC ratio ,Rare Diseases ,0302 clinical medicine ,Clinical Research ,Internal medicine ,Pulmonary fibrosis ,medicine ,Humans ,In patient ,030212 general & internal medicine ,Respiratory system ,Lung ,11 Medical and Health Sciences ,Retrospective Studies ,Aged ,pulmonary fibrosis ,business.industry ,Monocyte ,biomarkers ,Pirfenidone ,Middle Aged ,Prognosis ,medicine.disease ,Idiopathic Pulmonary Fibrosis ,medicine.anatomical_structure ,030228 respiratory system ,6.1 Pharmaceuticals ,Female ,prognosis ,business ,medicine.drug - Abstract
Rationale: There is an urgent need for simple, cost-effective prognostic biomarkers for idiopathic pulmonary fibrosis (IPF); biomarkers that show potential include monocyte count. Objectives: We used pooled data from pirfenidone and interferon gamma-1b trials to explore the association between monocyte count and prognosis in patients with IPF. Methods: This retrospective pooled analysis included patients (active and placebo arms) from four Phase III, randomized, placebo-controlled trials: ASCEND (NCT01366209), CAPACITY (NCT00287729 and NCT00287716), and INSPIRE (NCT00075998). Outcomes included IPF progression (≥10% absolute decline in percent predicted forced vital capacity, ≥50 m decline in 6-minute walk distance, or death), all-cause hospitalization, and all-cause mortality over 1 year. The relationship between monocyte count (defined as time-dependent) and outcomes was assessed using bivariate and multivariable models. Measurements and Main Results: This analysis included 2067 patients stratified by monocyte count (at baseline
- Published
- 2021
- Full Text
- View/download PDF
17. Reply to Noboa-Sevilla et al
- Author
-
Janelle Vu Pugashetti, Chad A. Newton, Philip L. Molyneaux, and Justin M. Oldham
- Subjects
Pulmonary and Respiratory Medicine ,Critical Care and Intensive Care Medicine - Published
- 2023
- Full Text
- View/download PDF
18. Reply to: Functional criteria to define progressive pulmonary fibrosis: Searching for the Holy Grail
- Author
-
Janelle, Vu Pugashetti, Chad A, Newton, Philip L, Molyneaux, and Justin M, Oldham
- Published
- 2022
19. The causal relationship between gastro-esophageal reflux disease and idiopathic pulmonary fibrosis: A bidirectional two-sample Mendelian randomization study
- Author
-
Carl J Reynolds, Fabiola Del Greco M, Richard J Allen, Carlos Flores, R Gisli Jenkins, Toby M Maher, Philip L Molyneaux, Imre Noth, Justin M Oldham, Louise V Wain, Jiyuan An, Jue-Sheng Ong, Stuart MacGregor, Tom A. Yates, Paul Cullinan, and Cosetta Minelli
- Abstract
BackgroundGastro-esophageal reflux disease (GERD) is associated with idiopathic pulmonary fibrosis (IPF) in observational studies. It is not known if this association arises because GERD causes IPF, or IPF causes GERD, or because of confounding by factors, such as smoking, associated with both GERD and IPF. We used bidirectional Mendelian randomisation (MR), where genetic variants are used as instrumental variables to address issues of confounding and reverse causation, to examine how, if at all, GERD and IPF are causally related.Methods and resultsA bidirectional two-sample MR was performed to estimate the causal effect of GERD on IPF risk, and of IPF on GERD risk, using genetic data from the largest GERD (78,707 cases and 288,734 controls) and IPF (4,125 cases and 20,464 controls) genome-wide association meta-analyses currently available. GERD increased the risk of IPF, with an odds ratio (OR) of 1.6 (95% Confidence Interval, CI: 1.04-2.49; p=0.032). There was no evidence of a causal effect of IPF on the risk of GERD, with an OR of 0.99 (95%CI: 0.97-1.02; p=0.615).ConclusionWe found that GERD increases the risk of IPF, but found no evidence that IPF increases the risk of GERD. GERD should be considered in future studies of IPF risk, and interest in it as a potential therapeutic target should be renewed. The mechanisms underlying the effect of GERD on IPF should also be investigated.
- Published
- 2022
- Full Text
- View/download PDF
20. The causal relationship between gastro-oesophageal reflux disease and idiopathic pulmonary fibrosis: a bidirectional two-sample Mendelian randomisation study
- Author
-
Carl J. Reynolds, Fabiola Del Greco M, Richard J. Allen, Carlos Flores, R. Gisli Jenkins, Toby M. Maher, Philip L. Molyneaux, Imre Noth, Justin M. Oldham, Louise V. Wain, Jiyuan An, Jue-Sheng Ong, Stuart MacGregor, Tom A. Yates, Paul Cullinan, and Cosetta Minelli
- Subjects
Pulmonary and Respiratory Medicine - Abstract
BackgroundGastro-oesophageal reflux disease (GORD) is associated with idiopathic pulmonary fibrosis (IPF) in observational studies. It is not known if this association arises because GORD causes IPF or because IPF causes GORD, or because of confounding by factors, such as smoking, associated with both GORD and IPF. We used bidirectional Mendelian randomisation (MR), where genetic variants are used as instrumental variables to address issues of confounding and reverse causation, to examine how, if at all, GORD and IPF are causally related.MethodsA bidirectional two-sample MR was performed to estimate the causal effect of GORD on IPF risk and of IPF on GORD risk, using genetic data from the largest GORD (78 707 cases and 288 734 controls) and IPF (4125 cases and 20 464 controls) genome-wide association meta-analyses currently available.ResultsGORD increased the risk of IPF, with an OR of 1.6 (95% CI 1.04–2.49; p=0.032). There was no evidence of a causal effect of IPF on the risk of GORD, with an OR of 0.999 (95% CI 0.997–1.000; p=0.245).ConclusionsWe found that GORD increases the risk of IPF, but found no evidence that IPF increases the risk of GORD. GORD should be considered in future studies of IPF risk and interest in it as a potential therapeutic target should be renewed. The mechanisms underlying the effect of GORD on IPF should also be investigated.
- Published
- 2023
- Full Text
- View/download PDF
21. BAL Is Safe and Well Tolerated in Individuals with Idiopathic Pulmonary Fibrosis: An Analysis of the PROFILE Study
- Author
-
Andrew G. Nicholson, Philip L. Molyneaux, Felix Chua, Toby M. Maher, Elisabetta A. Renzoni, William A. Fahy, Jonathan J Smith, Peter Saunders, Athol U. Wells, and R. Gisli Jenkins
- Subjects
Male ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,MEDLINE ,Critical Care and Intensive Care Medicine ,Bronchoalveolar Lavage ,Gastroenterology ,Idiopathic pulmonary fibrosis ,Bronchoscopy ,Cytology ,Internal medicine ,Correspondence ,medicine ,Humans ,Aged ,Aged, 80 and over ,medicine.diagnostic_test ,business.industry ,Interstitial lung disease ,Middle Aged ,medicine.disease ,Idiopathic Pulmonary Fibrosis ,Clinical trial ,Treatment Outcome ,Female ,business - Published
- 2021
- Full Text
- View/download PDF
22. Genome-wide Enrichment of
- Author
-
David, Zhang, Gundula, Povysil, Chad A, Newton, Toby M, Maher, Philip L, Molyneaux, Imre, Noth, Fernando J, Martinez, Ganesh, Raghu, Jamie L, Todd, Scott M, Palmer, Adam, Platt, Slavé, Petrovski, David B, Goldstein, and Christine Kim, Garcia
- Subjects
Humans ,Genetic Predisposition to Disease ,Hispanic or Latino ,Telomerase ,Idiopathic Pulmonary Fibrosis ,Genome-Wide Association Study - Published
- 2022
23. Reply to Fujimoto
- Author
-
Philip L, Molyneaux and Toby M, Maher
- Subjects
Keratin-19 ,Antigens, Neoplasm ,Humans - Published
- 2022
24. PCSK6 and Survival in Idiopathic Pulmonary Fibrosis
- Author
-
Justin M. Oldham, Richard J. Allen, Jose M. Lorenzo-Salazar, Philip L. Molyneaux, Shwu-Fan Ma, Chitra Joseph, John S. Kim, Beatriz Guillen-Guio, Tamara Hernández-Beeftink, Jonathan A. Kropski, Yong Huang, Cathryn T. Lee, Ayodeji Adegunsoye, Janelle Vu Pugashetti, Angela L. Linderholm, Vivian Vo, Mary E. Strek, Jonathan Jou, Adrian Muñoz-Barrera, Luis A. Rubio-Rodriguez, Richard Hubbard, Nik Hirani, Moira K.B. Whyte, Simon Hart, Andrew G. Nicholson, Lisa Lancaster, Helen Parfrey, Doris Rassl, William Wallace, Eleanor Valenzi, Yingze Zhang, Josyf Mychaleckyj, Amy Stockwell, Naftali Kaminski, Paul J. Wolters, Maria Molina-Molina, William A. Fahy, Fernando J. Martinez, Ian P. Hall, Martin D. Tobin, Toby M. Maher, Timothy S. Blackwell, Brian L. Yaspan, R Gisli Jenkins, Carlos Flores, Louise V Wain, and Imre Noth
- Abstract
RationaleIdiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by limited treatment options and high mortality. Novel therapies and prognostic biomarkers are needed.ObjectiveTo identify and validate molecular determinants of IPF survival.MethodsA staged genome-wide association study (GWAS) was performed using paired genomic and survival data. Stage I cases were drawn from centers across the US and Europe and stage II cases from Vanderbilt University. Cox proportional hazards regression was used to identify gene variants associated with differential transplant-free survival (TFS). Stage I variants with nominal significance (p−5) were advanced for stage II testing and meta-analyzed to identify those reaching genome-wide significance (p−8). Downstream analyses were performed for genes and proteins associated with variants reaching genome-wide significance.Main ResultsAfter quality controls, 1481 stage I cases and 397 stage II cases were included in the analysis. After filtering, 9,075,629 variants were tested in stage I, with 158 meeting advancement criteria. Four variants associated with TFS with consistent effect direction were identified in stage II, including one in an intron of proprotein convertase subtilisin/kexin type 6 (PCSK6) reaching genome-wide significance (HR 4.11; 95%CI 2.54-6.67; p=9.45×10−9). PCSK6 protein was highly expressed in IPF lung parenchyma and negatively correlated with survival. Peripheral blood PCSK6 gene expression and plasma concentration were associated with reduced transplant-free survival.ConclusionsWe identified four novel variants associated with IPF survival, including one in PCSK6 that reached genome-wide significance. Downstream analyses suggested that PCSK6 protein may serve as prognostic biomarker in IPF and potential therapeutic target.
- Published
- 2022
- Full Text
- View/download PDF
25. Rituximab versus intravenous cyclophosphamide in patients with connective tissue disease-associated interstitial lung disease in the UK (RECITAL): a double-blind, double-dummy, randomised, controlled, phase 2b trial
- Author
-
Toby M Maher, Veronica A Tudor, Peter Saunders, Michael A Gibbons, Sophie V Fletcher, Christopher P Denton, Rachel K Hoyles, Helen Parfrey, Elisabetta A Renzoni, Maria Kokosi, Athol U Wells, Deborah Ashby, Matyas Szigeti, Philip L Molyneaux, Mohammed Akil, Daphne Babalis, Nazia Chaudhuri, Felix Chua, Arnab Data, Dhananjay Desai, Shrish Dubey, Natalie Dwyer, Marcus Flather, Richard Fordham, Carlota Grossi Sampedro, Frances Hall, Ira Jakupovic, Gregory Keir, Bipen Patel, Henry Penn, Arvind Rajasekaran, Lisa G Spencer, Vicky Tsipouri, Zhe Wu, Georgio Xydopoulos, and Fernando Zanghelini
- Subjects
Pulmonary and Respiratory Medicine ,Treatment Outcome ,Double-Blind Method ,Adrenal Cortex Hormones ,Quality of Life ,Humans ,Rituximab ,Lung Diseases, Interstitial ,Connective Tissue Diseases ,Cyclophosphamide ,United Kingdom - Abstract
Background: rituximab is often used as rescue therapy in interstitial lung disease (ILD) associated with connective tissue disease (CTD), but has not been studied in clinical trials. This study aimed to assess whether rituximab is superior to cyclophosphamide as a treatment for severe or progressive CTD associated ILD.Methods: we conducted a randomised, double-blind, double-dummy, phase 2b trial to assess the superiority of rituximab compared with cyclophosphamide. Patients aged 18-80 years with severe or progressive ILD related to scleroderma, idiopathic inflammatory myositis, or mixed CTD, recruited across 11 specialist ILD or rheumatology centres in the UK, were randomly assigned (1:1) to receive rituximab (1000 mg at weeks 0 and 2 intravenously) or cyclophosphamide (600 mg/m2 body surface area every 4 weeks intravenously for six doses). The primary endpoint was rate of change in forced vital capacity (FVC) at 24 weeks compared with baseline, analysed using a mixed-effects model with random intercepts, adjusted for baseline FVC and CTD type. Prespecified secondary endpoints reported in this Article were change in FVC at 48 weeks versus baseline; changes from baseline in 6 min walk distance, diffusing capacity of the lung for carbon monoxide (DLCO), physician-assessed global disease activity (GDA) score, and quality-of-life scores on the St George's Respiratory Questionnaire (SGRQ), King's Brief Interstitial Lung Disease (KBILD) questionnaire, and European Quality of Life Five-Dimension (EQ-5D) questionnaire at 24 and 48 weeks; overall survival, progression-free survival, and time to treatment failure; and corticosteroid use. All endpoints were analysed in the modified intention-to-treat population, which comprised all patients who received at least one dose of study drug. This trial is registered with ClinicalTrials.gov (NCT01862926).Findings: between Dec 1, 2014, and March 31, 2020, we screened 145 participants, of whom 101 participants were randomly allocated: 50 (50%) to receive cyclophosphamide and 51 (50%) to receive rituximab. 48 (96%) participants in the cyclophosphamide group and 49 (96%) in the rituximab group received at least one dose of treatment and were included in analyses; 43 (86%) participants in the cyclophosphamide group and 42 (82%) participants in the rituximab group completed 24 weeks of treatment and follow-up. At 24 weeks, FVC was improved from baseline in both the cyclophosphamide group (unadjusted mean increase 99 mL [SD 329]) and the rituximab group (97 mL [234]); in the adjusted mixed-effects model, the difference in the primary endpoint at 24 weeks was -40 mL (95% CI -153 to 74; p=0·49) between the rituximab group and the cyclophosphamide group. KBILD quality-of-life scores were improved at 24 weeks by a mean 9·4 points (SD 20·8) in the cyclophosphamide group and 8·8 points (17·0) in the rituximab group. No significant differences in secondary endpoints were identified between the treatment groups, with the exception of change in GDA score at week 48, which favoured cyclophosphamide (difference 0·90 [95% CI 0·11 to 1·68]). Improvements in lung function and respiratory-related quality-of-life measures were observed in both treatment groups. Lower corticosteroid exposure over 48 weeks of follow-up was recorded in the rituximab group. Two (4%) of 48 participants who received cyclophosphamide and three (6%) of 49 who received rituximab died during the study, all due to complications of CTD or ILD. Overall survival, progression-free survival, and time to treatment failure did not significantly differ between the two groups. All participants reported at least one adverse event during the study. Numerically fewer adverse events were reported by participants receiving rituximab (445 events) than those receiving cyclophosphamide (646 events). Gastrointestinal and respiratory disorders were the most commonly reported adverse events in both groups. There were 62 serious adverse events of which 33 occurred in the cyclophosphamide group and 29 in the rituximab group.Interpretation: rituximab was not superior to cyclophosphamide to treat patients with CTD-ILD, although participants in both treatment groups had increased FVC at 24 weeks, in addition to clinically important improvements in patient-reported quality of life. Rituximab was associated with fewer adverse events. Rituximab should be considered as a therapeutic alternative to cyclophosphamide in individuals with CTD-ILD requiring intravenous therapy.Funding: efficacy and Mechanism Evaluation Programme (Medical Research Council and National Institute for Health Research, UK).
- Published
- 2022
26. Airway mucins promote immunopathology in virus-exacerbated chronic obstructive pulmonary disease
- Author
-
Aran Singanayagam, Joseph Footitt, Matthias Marczynski, Giorgia Radicioni, Michael T. Cross, Lydia J. Finney, Maria-Belen Trujillo-Torralbo, Maria Calderazzo, Jie Zhu, Julia Aniscenko, Thomas B. Clarke, Philip L. Molyneaux, Nathan W. Bartlett, Miriam F. Moffatt, William O. Cookson, Jadwiga Wedzicha, Christopher M. Evans, Richard C. Boucher, Mehmet Kesimer, Oliver Lieleg, Patrick Mallia, Sebastian L. Johnston, British Medical Association, British Lung Foundation, and Wellcome Trust
- Subjects
Innate immunity ,Inflammation ,Pulmonology ,Immunology ,Respiratory Mucosa ,General Medicine ,respiratory system ,Mucin 5AC ,digestive system ,Mucin-5B ,Disease Models, Animal ,Mice ,Mucus ,Pulmonary Disease, Chronic Obstructive ,fluids and secretions ,Adenosine Triphosphate ,Humans ,COPD ,Animals ,sense organs ,11 Medical and Health Sciences - Abstract
The respiratory tract surface is protected from inhaled pathogens by a secreted layer of mucus rich in mucin glycoproteins. Abnormal mucus accumulation is a cardinal feature of chronic respiratory diseases but the relationship between mucus and pathogens during exacerbations is poorly understood. We identified elevations in airway MUC5AC and MUC5B concentrations during spontaneous and experimentally-induced chronic obstructive pulmonary disease (COPD) exacerbations. MUC5AC was more sensitive to changes in expression during exacerbation and was therefore more predictably associated with virus load, inflammation, symptom severity, decrements in lung function, and secondary bacterial infections. MUC5AC was functionally related to inflammation as Muc5ac-deficient (Muc5ac-/-) mice had attenuated rhinovirus (RV)-induced airway inflammation and exogenous MUC5AC glycoprotein administration augmented inflammatory responses and increased release of extracellular adenosine triphosphate (ATP) in mice and human airway epithelial cell cultures. Hydrolysis of ATP suppressed MUC5AC augmentation of rhinovirus-induced inflammation in mice. Therapeutic suppression of mucin production using an epidermal growth factor receptor (EGFR) antagonist ameliorated immunopathology in a mouse COPD exacerbation model. The coordinated virus induction of MUC5AC and MUC5B suggests that non-Th2 mechanisms trigger mucin hypersecretion during exacerbations. Our data identifies a pro-inflammatory role for MUC5AC during viral infection and suggest that MUC5AC inhibition may ameliorate COPD exacerbations.
- Published
- 2022
- Full Text
- View/download PDF
27. Rare and Common Variants in
- Author
-
David, Zhang, Gundula, Povysil, Philippe H, Kobeissy, Qi, Li, Binhan, Wang, Mason, Amelotte, Hager, Jaouadi, Chad A, Newton, Toby M, Maher, Philip L, Molyneaux, Imre, Noth, Fernando J, Martinez, Ganesh, Raghu, Jamie L, Todd, Scott M, Palmer, Carolina, Haefliger, Adam, Platt, Slavé, Petrovski, Joseph A, Garcia, David B, Goldstein, and Christine Kim, Garcia
- Subjects
Humans ,Kinesins ,Exome ,Telomere ,Telomerase ,Idiopathic Pulmonary Fibrosis - Published
- 2022
28. A comparison of long term outcomes in patients managed with VV-ECMO in the first and second waves of the COVID-19 pandemic in the UK
- Author
-
Benjamin Garfield, Paolo Bianchi, Deepa J Arachchillage, Francisca Caetano, Sujal Desai, James Doyle, Clara Hernandez Caballero, Anne-Marie Doyle, Sachin Mehta, Alexander Law, Sian Jaggar, Maria Kokosi, Philip L Molyneaux, Maurizio Passariello, Meena Naja, Carole Ridge, Joana Alçada, Brijesh Patel, Suveer Singh, and Stephane Ledot
- Abstract
BackgroundEarly studies of veno-venous extracorporeal membrane oxygenation (VV-ECMO) in COVID-19 have revealed similar outcomes to historical cohorts. Changes in the disease and treatments has led to differences in the patients supported on VV-ECMO in the 1st and 2nd waves. We aimed to compare these two groups in both the acute and follow-up phase.MethodsIn this retrospective study, we identified the differences between patients supported on ECMO for COVID-19 between wave 1 (17/03/2020-31/08/2020) and wave 2 (01/09/2020-25/05/2021). We examined mortality at censoring date (30/11/2021) and decannulation, patient characteristics, complications and lung function and quality of life (QOL – by EQ5D3L) at first follow-up.FindingsOne-hundred and twenty-three patients were included in our analysis. Survival at censoring date [Chi-sqaured 6.35, p=0.012] and decannulation [90.4% vs 70.0%, pnd wave, whilst duration of ECMO run was longer [12.0(18.0-30.0) days vs. 29.5(15.5-58.3)] days (p=0.005)). Wave 2 patients had longer application of non-invasive ventilation (NIV) prior to ECMO and a higher incidence of barotrauma. Patient age and NIV use were independently associated with increased mortality [OR 1.07(1.01-1.14), p=0.025 and 3.37(1.12–12.60), p=0.043 respectively]. QOL and lung function, apart from KCOc was similar at follow up across the waves.ConclusionMost patients with COVID-19 supported on ECMO in both waves survived in the short and longer term. At follow-up patients had similar lung function and QOL across the 2 waves. This suggests that ECMO has an ongoing role in the management of a carefully selected group of patients with COVID-19.Trial RegistrationResearch Ethics Committee (20/EM/0204)
- Published
- 2022
- Full Text
- View/download PDF
29. Serum markers of pulmonary epithelial damage in systemic sclerosis‐associated interstitial lung disease and disease progression
- Author
-
Maria Kokosi, Toby M. Maher, Felix Chua, Athol U. Wells, Rachel K. Hoyles, Vasilis Kouranos, Peter M. George, Jackie Donovan, Veronica Alfieri, Dina Visca, Philip L. Molyneaux, Angelo De Lauretis, Cécile Daccord, George Margaritopoulos, Christopher P. Denton, Elisabetta A. Renzoni, David Abraham, Carmel Stock, Voon H Ong, Piersante Sestini, and Action for Pulmonary Fibrosis
- Subjects
Krebs von den Lungen‐ ,INVOLVEMENT ,CLEARANCE ,associated interstitial lung disease ,Respiratory System ,systemic sclerosis‐ ,CYFRA 21‐ ,Gastroenterology ,DLCO ,FIBROSIS ,Medicine ,Prospective Studies ,Prospective cohort study ,Lung ,11 Medical and Health Sciences ,HUMAN MUC1 MUCIN ,Interstitial lung disease ,respiratory system ,PREDICTS ,Krebs von den Lungen-6 ,Cohort ,Disease Progression ,biomarker ,CYFRA 21-1 ,Biomarker (medicine) ,DETERIORATION ,Life Sciences & Biomedicine ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,disease progression ,MUC1 allele ,systemic sclerosis-associated interstitial lung disease ,VON DEN LUNGEN-6 ,FEV1/FVC ratio ,Antigens, Neoplasm ,Internal medicine ,Humans ,Retrospective Studies ,Keratin-19 ,Science & Technology ,Scleroderma, Systemic ,business.industry ,Retrospective cohort study ,KL-6 LEVELS ,medicine.disease ,SURFACTANT PROTEIN-D ,SEVERITY ,Lung Diseases, Interstitial ,business ,Biomarkers - Abstract
Background and objective The course of systemic sclerosis-associated interstitial lung disease (SSc-ILD) is highly variable, and accurate prognostic markers are needed. KL-6 is a mucin-like glycoprotein (MUC1) expressed by type II pneumocytes, while CYFRA 21-1 is expressed by alveolar and bronchiolar epithelial cells. Both are released into the blood from cell injury. Methods Serum KL-6 and CYFRA 21-1 levels were measured in a retrospective (n = 189) and a prospective (n = 118) cohort of SSc patients. Genotyping of MUC1 rs4072037 was performed. Linear mixed-effect models were used to evaluate the relationship with change in lung function parameters over time, while association with survival was evaluated with Cox proportional hazard analysis. Results In both cohorts, KL-6 and CYFRA 21-1 were highest in patients with lung involvement, and in patients with extensive rather than limited ILD. KL-6 was higher in patients carrying the MUC1 rs4072037 G allele in both cohorts. In patients with SSc-ILD, serum KL-6, but not CYFRA 21-1, was significantly associated with DLCO decline in both cohorts (P = 0.001 and P = 0.004, respectively), and with FVC decline in the retrospective cohort (P = 0.005), but not the prospective cohort. When combining the cohorts and subgrouping by severity (median CPI = 45.97), KL-6 remained predictive of decline in DLCO in both milder (P = 0.007) and more severe disease (P = 0.02) on multivariable analysis correcting for age, gender, ethnicity, smoking history and MUC1 allele carriage. Conclusion Our results suggest serum KL-6 predicts decline in lung function in SSc, suggesting its clinical utility in risk stratification for progressive SSc-ILD.
- Published
- 2020
- Full Text
- View/download PDF
30. Presence of pleomorphic features but not growth patterns improves prognostic stratification of epithelioid malignant pleural mesothelioma by 2‐tier nuclear grade
- Author
-
Eric Lim, Michael Dusmet, S Begum, Loic Lang-Lazdunski, Aliya N. Husain, William O.C.M. Cookson, Miriam F. Moffatt, Jan Lukas Robertus, Emma Beddow, Vladimir Anikin, Yu Zhi Zhang, Andrew G. Nicholson, Sanjay Popat, Philip L. Molyneaux, Alexandra Rice, John Le Quesne, Jonathan Finch, Simon Jordan, Nizar Asadi, C. Brambilla, and Action for Pulmonary Fibrosis
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Histology ,Composite score ,Pleural Neoplasms ,pleomorphic features ,nuclear grade ,Prognostic stratification ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Pathology ,Overall survival ,medicine ,Humans ,Mesothelioma ,growth patterns ,Nuclear grade ,Aged ,Aged, 80 and over ,Science & Technology ,Pleural mesothelioma ,business.industry ,Epithelioid Cells ,Mesothelioma, Malignant ,1103 Clinical Sciences ,Cell Biology ,General Medicine ,Middle Aged ,Prognosis ,medicine.disease ,Micropapillary pattern ,030104 developmental biology ,mesothelioma ,030220 oncology & carcinogenesis ,Female ,Neoplasm Grading ,heterogeneity ,business ,Life Sciences & Biomedicine - Abstract
AIMS: Nuclear grade has been recently validated as a powerful prognostic tool in epithelioid malignant pleural mesothelioma (E-MPM). In other studies histological parameters including pleomorphic features and growth patterns were also shown to exert prognostic impact. The primary aims of our study are (1) externally validate the prognostic role of pleomorphic features in E-MPM and (2) investigate if evaluating growth pattern in addition to 2-tier nuclear grade improves prognostication. METHODS AND RESULTS: 614 consecutive cases of E-MPM from our institution over a period of 15 years were retrospectively reviewed, of which 51 showed pleomorphic features. E-MPM with pleomorphic features showed significantly worse overall survival compared those without (5.4 months vs 14.7 months). Tumours with predominantly micropapillary pattern showed the worst survival (6.2 months) followed by solid (10.5 months), microcystic (15.3 months), discohesive (16.1 months), trabecular (17.6 months) and tubulo-papillary (18.6 months). Sub-classification of growth patterns into high grade (solid, micropapillary) and low grade (all others) led to good separation of overall survival (10.5 months vs. 18.0 months) but did not predict survival independent of 2-tier nuclear grade. A composite score comprised of growth pattern and 2-tier nuclear grade did not improve prognostication compared with nuclear grade alone. Intra-tumoural heterogeneity in growth patterns is ubiquitous. CONCLUSIONS: Our findings support the incorporation of E-MPM with pleomorphic features in the epithelioid subtype as a highly aggressive variant distinct from 2-tier nuclear grade. E-MPM demonstrates extensive heterogeneity in growth pattern but its evaluation does not offer additional prognostic utility to 2-tier nuclear grade.
- Published
- 2020
- Full Text
- View/download PDF
31. Methods in Lung Microbiome Research
- Author
-
Stavros Garantziotis, Leopoldo N. Segal, Robert P. Dickson, Janice M. Leung, Tricia D. LeVan, Georgios D Kitsios, Philip L. Molyneaux, Jose C. Clemente, Yvonne J. Huang, David N O'Dwyer, Sharon M. Carney, Bethany B. Moore, Kirsten M. Kloepfer, and Michael J. Cox
- Subjects
Microbiological Techniques ,0301 basic medicine ,Pulmonary and Respiratory Medicine ,Lung microbiome ,analysis ,Respiratory System ,Clinical Biochemistry ,microbiome ,Health outcomes ,Models, Biological ,Rigour ,Specimen Handling ,lung ,methods ,Translational Research, Biomedical ,03 medical and health sciences ,0302 clinical medicine ,Anti-Infective Agents ,microbiome, lung, reporting, analysis, methods, review ,Animals ,Humans ,Microbiome ,1102 Cardiorespiratory Medicine and Haematology ,Molecular Biology ,reporting ,Host Microbial Interactions ,Whole Genome Sequencing ,Microbiota ,Sputum ,Reproducibility of Results ,Environmental Exposure ,Cell Biology ,Research opportunities ,Data science ,Bacterial Typing Techniques ,Body Fluids ,Translational Review ,030104 developmental biology ,Breath Tests ,030228 respiratory system ,Models, Animal ,Dysbiosis ,Metagenomics ,Epidemiologic Methods ,Psychology - Abstract
The lung microbiome is associated with host immune response and health outcomes in experimental models and patient cohorts. Lung microbiome research is increasing in volume and scope; however, there are no established guidelines for study design, conduct and reporting of lung microbiome studies. Standardized approaches to yield reliable and reproducible data that can be synthesized across studies, will ultimately improve the scientific rigor and impact of published work and greatly benefit microbiome research. In this review, we identify and address several key elements of microbiome research: conceptual modeling and hypothesis framing, study design, experimental methodology and pitfalls, data analysis and reporting considerations. Finally, we explore possible future directions and research opportunities. Our goal is to aid investigators who are interested in this burgeoning research area and will hopefully provide the foundation for formulating consensus approaches in lung microbiome research.
- Published
- 2020
- Full Text
- View/download PDF
32. Defining genetic risk factors for scleroderma-associated interstitial lung disease
- Author
-
Felix Chua, Christopher P. Denton, Dina Visca, Peter M. George, Cécile Daccord, Elisabetta A. Renzoni, Philip L. Molyneaux, Maria Kokosi, Louise V. Wain, Angelo De Lauretis, Toby M. Maher, David Abraham, Vasilis Kouranos, Athol U. Wells, George Margaritopoulos, Carmel Stock, Svetlana I. Nihtyanova, Voon H Ong, Action for Pulmonary Fibrosis, Versus Arthritis, The Scleroderma Society, British Lung Foundation, National Institute for Health Research, and Imperial College Healthcare NHS Trust- BRC Funding
- Subjects
CD226 ,Genetic association ,Genetics ,IRAK1 ,IRF5 ,SSc-ILD ,STAT4 ,0301 basic medicine ,medicine.medical_specialty ,Single-nucleotide polymorphism ,SUSCEPTIBILITY ,Gastroenterology ,FUNCTIONAL POLYMORPHISM ,03 medical and health sciences ,Idiopathic pulmonary fibrosis ,0302 clinical medicine ,Rheumatology ,Internal medicine ,CYCLOPHOSPHAMIDE ,medicine ,Genetic predisposition ,RS2004640 ,Allele ,skin and connective tissue diseases ,Allele frequency ,030203 arthritis & rheumatology ,Science & Technology ,integumentary system ,business.industry ,SYSTEMIC-SCLEROSIS ,Case-control study ,Interstitial lung disease ,1103 Clinical Sciences ,General Medicine ,respiratory system ,medicine.disease ,Arthritis & Rheumatology ,030104 developmental biology ,IDIOPATHIC PULMONARY-FIBROSIS ,business ,Life Sciences & Biomedicine - Abstract
Although several genetic associations with scleroderma (SSc) are defined, very little is known on genetic susceptibility to SSc-associated interstitial lung disease (SSc-ILD). A number of common polymorphisms have been associated with SSc-ILD, but most have not been replicated in separate populations. Four SNPs in IRF5, and one in each of STAT4, CD226 and IRAK1, selected as having been previously the most consistently associated with SSc-ILD, were genotyped in 612 SSc patients, of European descent, of whom 394 had ILD. The control population (n = 503) comprised individuals of European descent from the 1000 Genomes Project. After Bonferroni correction, two of the IRF5 SNPs, rs2004640 (OR (95% CI)1.30 (1.10–1.54), pcorr = 0.015) and rs10488631 (OR 1.48 (1.14–1.92), pcorr = 0.022), and the STAT4 SNP rs7574865 (OR 1.43 (1.18–1.73), pcorr = 0.0015) were significantly associated with SSc compared with controls. However, none of the SNPs were significantly different between patients with SSc-ILD and controls. Two SNPs in IRF5, rs10488631 (OR 1.72 (1.24–2.39), pcorr = 0.0098), and rs2004640 (OR 1.39 (1.11–1.75), pcorr = 0.03), showed a significant difference in allele frequency between controls and patients without ILD, as did STAT4 rs7574865 (OR 1.86 (1.45–2.38), pcorr = 6.6 × 10−6). A significant difference between SSc with and without ILD was only observed for STAT4 rs7574865, being less frequent in patients with ILD (OR 0.66 (0.51–0.85), pcorr = 0.0084). In conclusion, IRF5 rs2004640 and rs10488631, and STAT4 rs7574865 were significantly associated with SSc as a whole. Only STAT4 rs7574865 showed a significant difference in allele frequency in SSc-ILD, with the T allele being protective against ILD.Key points• We confirm the associations of the IRF5 SNPs rs2004640 and rs10488631, and the STAT4 SNP rs7574865, with SSc as a whole.• None of the tested SNPs were risk factors for SSc-ILD specifically.• The STAT4 rs7574865 T allele was protective against the development of lung fibrosis in SSc patients.• Further work is required to understand the genetic basis of lung fibrosis in association with scleroderma.
- Published
- 2020
- Full Text
- View/download PDF
33. Interaktionen zwischen respiratorischem Mikrobiom und Epithelzellen formen Immunität in der Lunge
- Author
-
Clare M. Lloyd, Rachele Invernizzi, and Philip L. Molyneaux
- Abstract
Das respiratorische Epithel stellt eine physische Grenze zur äußeren Umgebung dar und fungiert als erste Verteidigungslinie gegen potenziell schädliche Umweltreize wie Mikroben und Allergene. Die Epithelzellen in der Lunge gelten jedoch zunehmend auch als aktive Effektoren der Abwehr von Erregern, die zur angeborenen und adaptiven Immunfunktion im unteren Respirationstrakt beitragen. Diese Zellen exprimieren ein breites Spektrum an Pattern-Recognition-Rezeptoren, die spezifisch für Strukturmotive der Erreger und des Wirts sind. Mit Hilfe moderner molekularer Verfahren konnte die Komplexität des Mikrobioms im unteren Respirationstrakt aufgedeckt werden. Das Wechselspiel zwischen Mikrobiota und respiratorischem Epithel ist der Schlüssel zum Verständnis der Aufrechterhaltung einer stabilen Immunhomöostase. Ein Verlust der Epithelintegrität nach Exposition gegenüber einer Infektion kann bei anfälligen Personen eine Entzündung auslösen und letztlich zu einer Lungenerkrankung führen. In der vorliegenden Arbeit erörtern wir den aktuellen Erkenntnisstand in Bezug auf die molekularen und zellulären Mechanismen, über die das Lungenepithel mit dem Lungenmikrobiom interagiert und so eine Immunität in der Lunge formt. Im Mittelpunkt stehen insbesondere die Interaktionen zwischen dem Lungenmikrobiom und den Zellen der leitenden Atemwege bei der Modulierung der Immunzellregulation sowie die Frage, wie Defekte in der Barrierestruktur und -funktion letztlich zu einer Lungenerkrankung führen können. Diese Interaktionen zu verstehen, ist bei der Suche nach wirksameren Therapien bei Atemwegserkrankungen von fundamentaler Bedeutung.
- Published
- 2020
- Full Text
- View/download PDF
34. Safety, tolerability, and efficacy of pirfenidone in patients with rheumatoid arthritis-associated interstitial lung disease: a randomised, double-blind, placebo-controlled, phase 2 study
- Author
-
Joshua J Solomon, Sonye K Danoff, Felix A Woodhead, Shelley Hurwitz, Rie Maurer, Ian Glaspole, Paul F Dellaripa, Bibek Gooptu, Robert Vassallo, P Gerard Cox, Kevin R Flaherty, Huzaifa I Adamali, Michael A Gibbons, Lauren Troy, Ian A Forrest, Joseph A Lasky, Lisa G Spencer, Jeffrey Golden, Mary Beth Scholand, Nazia Chaudhuri, Mark A Perrella, David A Lynch, Daniel C Chambers, Martin Kolb, Cathie Spino, Ganesh Raghu, Hilary J Goldberg, Ivan O Rosas, Shana Haynes-Harp, Fernando Poli, Coimbatore Sree Vidya, Rebecca R. Baron, Timothy Clouser, Tracy Doyle, Anthony Maeda, Kristin B. Highland, Jemima F. Albayda, Sarah E. Collins, Karthik S. Suresh, John M. Davis, Andrew H. Limper, Isabel Amigues, Kristina Eliopoulos, Jeffery J. Swigris, Stephen Humphries, John C. Huntwork, Chris Glynn, Steve R. Duncan, Maria I. Danila, Marilyn K. Glassberg, Elana M. Oberstein, Elizabeth A. Belloli, Linda Briggs, Vivek Nagaraja, Linda Cholewa, Donna DiFranco, Edward Green, Christie Liffick, Tanvi Naik, Genevieve Montas, Dorota Lebiedz-Odrobina, Reba Bissell, Mark Wener, Lisa H. Lancaster, Leslie J. Crawford, Karmela Chan, Robert J. Kaner, Alicia Morris, Xiaoping Wu, Nader A. Khalidi, Christopher J. Ryerson, Alyson W. Wong, Charlene D. Fell, Sharon A. LeClercq, Mark Hyman, Shane Shapera, Shikha Mittoo, Shireen Shaffu, Karl Gaffney, Andrew M. Wilson, Shaney Barratt, Harsha Gunawardena, Rachel K. Hoyles, Joel David, Namrata Kewalramani, Toby M. Maher, Philip L. Molyneaux, Maria A. Kokosi, Matthew J. Cates, Mandizha Mandizha, Abdul Ashish, Gladstone Chelliah, Helen Parfrey, Muhunthan Thillai, Josephine Vila, Sophie V. Fletcher, Paul Beirne, Clair Favager, Jo Brown, Julie K. Dawson, Pilar Rivera Ortega, Sahena Haque, Pippa Watson, Jun K. Khoo, Karen Symons, Peter Youssef, and John A. Mackintosh
- Subjects
Pulmonary and Respiratory Medicine - Abstract
Interstitial lung disease is a known complication of rheumatoid arthritis, with a lifetime risk of developing the disease in any individual of 7·7%. We aimed to assess the safety, tolerability, and efficacy of pirfenidone for the treatment of patients with rheumatoid arthritis-associated interstitial lung disease (RA-ILD).TRAIL1 was a randomised, double-blind, placebo-controlled, phase 2 trial done in 34 academic centres specialising in interstitial lung disease in four countries (the UK, the USA, Australia, and Canada). Adults aged 18-85 years were eligible for inclusion if they met the 2010 American College of Rheumatology and European Alliance of Associations for Rheumatology criteria for rheumatoid arthritis and had interstitial lung disease on a high-resolution CT scan imaging and, when available, lung biopsy. Exclusion criteria include smoking, clinical history of other known causes of interstitial lung disease, and coexistant clinically significant COPD or asthma. Patients were randomly assigned (1:1) to receive 2403 mg oral pirfenidone (pirfenidone group) or placebo (placebo group) daily. The primary endpoint was the incidence of the composite endpoint of a decline from baseline in percent predicted forced vital capacity (FVC%) of 10% or more or death during the 52-week treatment period assessed in the intention-to-treat population. Key secondary endpoints included change in absolute and FVC% over 52 weeks, the proportion of patients with a decline in FVC% of 10% or more, and the frequency of progression as defined by Outcome Measures in Rheumatoid Arthritis Clinical Trials (OMERACT) in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT02808871.From May 15, 2017, to March 31, 2020, 231 patients were assessed for inclusion, of whom 123 patients were randomly assigned (63 [51%] to the pirfenidone group and 60 [49%] to the placebo group). The trial was stopped early (March 31, 2020) due to slow recruitment and the COVID-19 pandemic. The difference in the proportion of patients who met the composite primary endpoint (decline in FVC% from baseline of 10% or more or death) between the two groups was not significant (seven [11%] of 63 patients in the pirfenidone group vs nine [15%] of 60 patients in the placebo group; OR 0·67 [95% CI 0·22 to 2·03]; p=0·48). Compared with the placebo group, patients in the pirfenidone group had a slower rate of decline in lung function, measured by estimated annual change in absolute FVC (-66 vs -146; p=0·0082) and FVC% (-1·02 vs -3·21; p=0·0028). The groups were similar with regards to the decline in FVC% by 10% or more (five [8%] participants in the pirfenidone group vs seven [12%] in the placebo group; OR 0·52 [95% CI 0·14-1·90]; p=0·32) and the frequency of progression as defined by OMERACT (16 [25%] in the pirfenidone group vs 19 [32%] in the placebo group; OR 0·68 [0·30-1·54]; p=0·35). There was no significant difference in the rate of treatment-emergent serious adverse events between the two groups, and there were no treatment-related deaths.Due to early termination of the study and underpowering, the results should be interpreted with caution. Despite not meeting the composite primary endpoint, pirfenidone slowed the rate of decline of FVC over time in patients with RA-ILD. Safety in patients with RA-ILD was similar to that seen in other pirfenidone trials.Genentech.
- Published
- 2022
35. Overzealous degradation of collagen fragment Pro-Gly-Pro by leukotriene A4 hydrolase (LTA4H) perpetuates fibrosis in IPF
- Author
-
Kornelija Suveizdyte, Dhiren F. Patel, Chloe J. Pyle, Patricia P. Ogger, Nicoletta Bruno, Kyle T. Mincham, Philip L. Molyneaux, Toby M. Maher, Adam J. Byrne, and Robert J. Snelgrove
- Published
- 2022
- Full Text
- View/download PDF
36. Immuno-proteomic profiling reveals aberrant immune cell regulation in the airways of individuals with ongoing post-COVD-19 respiratory disease
- Author
-
Bavithra Vijayakumar, Karim Boustani, Patricia P. Ogger, Artemis Papadaki, James Tonkin, Christopher M. Orton, Poonam Ghai, Kornelija Suveizdyte, Richard J. Hewitt, Sujal R. Desai, Anand Devaraj, Robert J. Snelgrove, Philip L. Molyneaux, Justin L. Garner, James E. Peters, Pallav L. Shah, Clare M. Lloyd, James A. Harker, Asthma UK, Rosetrees Trust, Action for Pulmonary Fibrosis, Medical Research Council (MRC), and Wellcome Trust
- Subjects
Adult ,Male ,Immunoproteins ,Proteome ,COVID19 ,Respiratory System ,Immunology ,T cells ,tissue-resident memory ,Article ,Monocytes ,tissue resident memory ,proteomics ,Immunology and Allergy ,Humans ,long COVID ,respiratory viral infection ,Aged ,B-Lymphocytes ,Immunity, Cellular ,SARS-CoV-2 ,COVID-19 ,Middle Aged ,respiratory tract ,Respiration Disorders ,Infectious Diseases ,1107 Immunology ,Female ,airways ,Follow-Up Studies ,T-Lymphocytes, Cytotoxic - Abstract
Some patients hospitalized with acute COVID-19 suffer respiratory symptoms that persist for many months. We delineated the immune-proteomic landscape in the airway and peripheral blood of healthy controls and post-COVID-19 patients 3 to 6 months after hospital discharge. Post-COVID-19 patients showed abnormal airway (but not plasma) proteomes, with elevated concentration of proteins associated with apoptosis, tissue repair and epithelial injury versus healthy individuals. Increased numbers of cytotoxic lymphocytes were observed in individuals with greater airway dysfunction, while increased B cell numbers and altered monocyte subsets were associated with more widespread lung abnormalities. 1 year follow-up of some post-COVID-19 patients indicated that these abnormalities resolved over time. In summary, COVID-19 causes a prolonged change to the airway immune landscape in those with persistent lung disease, with evidence of cell death and tissue repair linked to ongoing activation of cytotoxic T cells., Graphical Abstract, Many individuals recovering from acute SARS-CoV-2 infection suffer prolonged respiratory dysfunction for months to years after viral clearance. Vijayakumar, Boustani, Ogger, Papadaki et al. show that individuals with persistent symptoms 3-6 months after infection have an altered airway immune cell landscape and evidence of ongoing lung damage. Importantly, different immune cell types correlate with the severity of distinct aspects of ongoing respiratory disease.
- Published
- 2022
37. The Respiratory Microbiome in Health and Disease
- Author
-
Rachele Invernizzi and Philip L. Molyneaux
- Published
- 2022
- Full Text
- View/download PDF
38. Idiopathic Pulmonary Fibrosis and Other Interstitial Lung Diseases
- Author
-
Rachele Invernizzi, Stavros Garantziotis, and Philip L. Molyneaux
- Published
- 2022
- Full Text
- View/download PDF
39. Distinct airway epithelial immune responses after infection with SARS-CoV-2 compared to H1N1
- Author
-
Helen, Stölting, Laury, Baillon, Rebecca, Frise, Katie, Bonner, Richard J, Hewitt, Philip L, Molyneaux, Mindy L, Gore, Wendy S, Barclay, Sejal, Saglani, and Andrew, Bush
- Subjects
Influenza A Virus, H1N1 Subtype ,SARS-CoV-2 ,Child, Preschool ,Influenza, Human ,Immunity ,COVID-19 ,Humans ,Epithelial Cells ,Interferons ,Middle Aged ,Virus Replication - Abstract
Children are less likely than adults to suffer severe symptoms when infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), while influenza A H1N1 severity is comparable across ages except for the very young or elderly. Airway epithelial cells play a vital role in the early defence against viruses via their barrier and immune functions. We investigated viral replication and immune responses in SARS-CoV-2-infected bronchial epithelial cells from healthy paediatric (n = 6; 2.5-5.6 years old) and adult (n = 4; 47-63 years old) subjects and compared cellular responses following infection with SARS-CoV-2 or Influenza A H1N1. While infection with either virus triggered robust transcriptional interferon responses, including induction of type I (IFNB1) and type III (IFNL1) interferons, markedly lower levels of interferons and inflammatory proteins (IL-6, IL-8) were released following SARS-CoV-2 compared to H1N1 infection. Only H1N1 infection caused disruption of the epithelial layer. Interestingly, H1N1 infection resulted in sustained upregulation of SARS-CoV-2 entry factors FURIN and NRP1. We did not find any differences in the epithelial response to SARS-CoV-2 infection between paediatric and adult cells. Overall, SARS-CoV-2 had diminished potential to replicate, affect morphology and evoke immune responses in bronchial epithelial cells compared to H1N1.
- Published
- 2021
40. Survival effects of pulmonary vasodilators in group 3 pulmonary hypertension
- Author
-
Philip L. Molyneaux, Peter M. George, Athol U. Wells, Felix Chua, Vasilis Kouranos, Konstantinos Dimopoulos, Aleksander Kempny, Laura C. Price, Timothy Dawes, Elisabetta A. Renzoni, Colm McCabe, S. John Wort, Maria Kokosi, and Simon Bax
- Subjects
medicine.medical_specialty ,business.industry ,Internal medicine ,Cardiology ,medicine ,medicine.disease ,business ,Pulmonary hypertension ,Pulmonary vasodilators - Published
- 2021
- Full Text
- View/download PDF
41. Evaluation of CCL18 serum levels and genetic variant as prognostic biomarkers in SSc-ILD
- Author
-
Voon H Ong, Torsten Witte, Carmel Stock, Christopher P. Denton, Felix Chua, Peter M. George, Antje Prasse, Elisabetta A. Renzoni, Vasilios Kouranos, Benjamin Seeliger, David Abraham, Veronica Alfieri, Maria Kokosi, Athol U. Wells, and Philip L. Molyneaux
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,CCL18 ,Genetic variants ,Medicine ,business - Published
- 2021
- Full Text
- View/download PDF
42. Late Breaking Abstract - Fully automated airway measurement correlates with radiological disease progression in Idiopathic Pulmonary Fibrosis
- Author
-
Evan Morgan, Carola-Bibane Schönlieb, Philip L. Molyneaux, Tom McLellan, Fahdi Kanavati, Darren Gallagher, Kirl Kirov, Michael S. Roberts, Alessandro Ruggiero, and Muhunthan Thillai
- Subjects
Idiopathic pulmonary fibrosis ,Pathology ,medicine.medical_specialty ,Fully automated ,business.industry ,Radiological weapon ,Disease progression ,Medicine ,business ,Airway ,medicine.disease - Published
- 2021
- Full Text
- View/download PDF
43. Pulmonary Rehabilitation in Idiopathic Pulmonary Fibrosis and COPD: A Propensity-Matched Real-World Study
- Author
-
Claire M, Nolan, Oliver, Polgar, Susie J, Schofield, Suhani, Patel, Ruth E, Barker, Jessica A, Walsh, Karen A, Ingram, Peter M, George, Philip L, Molyneaux, Toby M, Maher, and William D-C, Man
- Subjects
Pulmonary Disease, Chronic Obstructive ,Exercise Tolerance ,Humans ,Exercise ,Idiopathic Pulmonary Fibrosis ,Exercise Therapy - Abstract
The adherence to and clinical efficacy of pulmonary rehabilitation in idiopathic pulmonary fibrosis (IPF), particularly in comparison with COPD, remains uncertain. The objectives of this real-world study were to compare the responses of patients with IPF with a matched group of patients with COPD undergoing the same supervised, outpatient pulmonary rehabilitation program and to determine whether pulmonary rehabilitation is associated with survival in IPF.Do people with IPF improve to the same extent with pulmonary rehabilitation as a matched group of individuals with COPD, and are noncompletion of or nonresponse to pulmonary rehabilitation, or both, associated with 1-year all-cause mortality in IPF?Using propensity score matching, 163 patients with IPF were matched 1:1 with a control group of 163 patients with COPD referred for pulmonary rehabilitation. We compared between-group pulmonary rehabilitation completion rates and response. Survival status in the IPF cohort was recorded over 1 year after pulmonary rehabilitation discharge. Cox proportional hazards regression explored the association between pulmonary rehabilitation status and all-cause mortality.Similar pulmonary rehabilitation completion rates (IPF, 69%; COPD, 63%; P = .24) and improvements in exercise response were observed in both groups with no significant mean between-group differences in incremental shuttle walk test (ISWT) change (mean, 2 m [95% CI, -18 to 22 m]). Pulmonary rehabilitation noncompletion (hazard ratio [HR], 5.62 [95% CI, 2.24-14.08]) and nonresponse (HR, 3.91 [95% CI, 1.54-9.93]) were associated independently with increased 1-year all-cause mortality in IPF.This real-word study demonstrated that patients with IPF have similar completion rates and magnitude of response to pulmonary rehabilitation compared with a matched group of patients with COPD. In IPF, noncompletion of and nonresponse to pulmonary rehabilitation were associated with increased all-cause mortality. These data reinforce the benefits of pulmonary rehabilitation in patients with IPF.
- Published
- 2021
44. Immuno-proteomic profiling reveals abundant airway CD8 T cells and ongoing epithelial injury in prolonged post-COVID19 respiratory disease
- Author
-
Karim Boustani, Philip L. Molyneaux, Patricia P. Ogger, Poonam Ghai, James A. Harker, Robert J. Snelgrove, Pallav L. Shah, Christopher M. Orton, Kornelija Suveizdyte, Artermis Papadaki, James Tonkin, Justin L. Garner, Bavithra Vijayakumar, Clare M. Lloyd, Richard J. Hewitt, and James E. Peters
- Subjects
medicine.diagnostic_test ,business.industry ,Respiratory disease ,medicine.disease ,Epithelial Damage ,Bronchoalveolar lavage ,Immune system ,Immunology ,medicine ,Cytotoxic T cell ,Respiratory system ,Airway ,business ,CD8 - Abstract
SummarySome patients hospitalized with acute COVID19 suffer respiratory symptoms that persist for many months. To characterize the local and systemic immune responses associated with this form of ‘Long COVID’, we delineated the immune and proteomic landscape in the airway and peripheral blood of normal volunteers and patients from 3 to 6 months after hospital discharge. The bronchoalveolar lavage (but not peripheral blood) proteome was abnormal in patients with post-COVID19 lung disease with significantly elevated concentration of proteins associated with apoptosis, tissue repair and epithelial injury. This correlated with an increase in cytotoxic lymphocytes (especially tissue resident CD8+T cells), lactate dehydrogenase and albumin (biomarkers of cell death and barrier integrity). Follow-up of a subset of these patients greater than 1-year post-COVID19 indicated these abnormalities resolved over time. Collectively, these data indicate that COVID-19 results in a prolonged change to the airway immune landscape in those with persistent lung disease, with evidence of cell death and tissue repair linked to ongoing activation of cytotoxic T cells.HighlightsThe post-COVID19 airway is characterized by increased cytotoxic lymphocytes.Distinct airway proteomes are associated with the airway immune cell landscape.The peripheral blood does not predict immune-proteome alterations in the airway post-COVID19.Persistent abnormalities in the airway immune-proteome post-COVID19 airways correlate with ongoing epithelial damage.
- Published
- 2021
- Full Text
- View/download PDF
45. Airway-specific autoantibodies identify a subset of patients with fibrotic interstitial lung disease
- Author
-
Philip L. Molyneaux, Karim Boustani, Toby M. Maher, James A. Harker, Richard J. Hewitt, Quan-Zhen Li, Rachele Invernizzi, and Poonam Ghai
- Subjects
biology ,Microarray ,business.industry ,Interstitial lung disease ,Autoantibody ,Disease ,respiratory system ,medicine.disease ,respiratory tract diseases ,Idiopathic pulmonary fibrosis ,Immunology ,medicine ,biology.protein ,Antibody ,business ,Airway ,Hypersensitivity pneumonitis - Abstract
BackgroundFibrotic interstitial lung disease (fILD) has previously been associated with the presence of autoantibody. While studies have focused on systemic autoimmunity, the role of local autoantibodies in the airway remains unknown. We therefore extensively characterised the airway and peripheral autoantibody profiles in patients with fILD and assessed association with disease severity and outcome.MethodsBronchoalveolar lavage (BAL) was collected from a cohort of fILD patients and total airway antibody concentrations were quantified. An autoantigen microarray was used to measure IgG and IgA autoantibodies against 124 autoantigens in BAL from 40 idiopathic pulmonary fibrosis (IPF), 20 chronic hypersensitivity pneumonitis (CHP), 20 connective tissue disease-associated ILD (CTD-ILD) patients and 20 controls.ResultsA subset of patients with fILD but not healthy controls had a local autoimmune signature in their airways that was not present systemically, regardless of disease. The proportion of patients with IPF with a local autoantibody signature was comparable to that of CTD-ILD, which has a known autoimmune pathology, identifying a potentially novel subset of patients. The presence of an airway autoimmune signature was not associated with reduced survival probability or changes in lung function in the cohort as a whole. Patients with IPF had increased airway total IgA and IgG1 while subjects with CHP had increased airway IgA, IgG1 and IgG4. In patients with CHP, increased airway total IgA was associated with reduced survival probability.ConclusionThe presence of airway autoantibodies identifies a unique subset of patients with fILD and the mechanisms by which these autoantibodies contribute to disease requires further investigation.
- Published
- 2021
- Full Text
- View/download PDF
46. Autoantibodies are present in the bronchoalveolar lavage but not circulation in patients with fibrotic interstitial lung disease
- Author
-
Richard J. Hewitt, James A. Harker, Quan-Zhen Li, Karim Boustani, Philip L. Molyneaux, Rachele Invernizzi, Poonam Ghai, Toby M. Maher, Asthma UK, Rosetrees Trust, The Royal Society, National Institute for Health Research, and British Lung Foundation
- Subjects
Pulmonary and Respiratory Medicine ,medicine.diagnostic_test ,biology ,business.industry ,Autoantibody ,Interstitial lung disease ,Connective tissue ,Disease ,respiratory system ,medicine.disease ,respiratory tract diseases ,Idiopathic pulmonary fibrosis ,medicine.anatomical_structure ,Bronchoalveolar lavage ,Immunology ,medicine ,biology.protein ,Antibody ,business ,Hypersensitivity pneumonitis - Abstract
BackgroundFibrotic interstitial lung disease (fILD) has previously been associated with the presence of autoantibody. While studies have focused on systemic autoimmunity, the role of local autoantibodies in the airways remains unknown. We therefore extensively characterised the airway and peripheral autoantibody profiles in patients with fILD, and assessed association with disease severity and outcome.MethodsBronchoalveolar lavage (BAL) fluid was collected from a cohort of fILD patients and total BAL antibody concentrations were quantified. An autoantigen microarray was used to measure IgG and IgA autoantibodies against 122 autoantigens in BAL from 40 idiopathic pulmonary fibrosis (IPF), 20 chronic hypersensitivity pneumonitis (CHP), 20 connective tissue disease-associated ILD (CTD-ILD) patients and 20 controls.ResultsA subset of patients with fILD but not healthy controls had a local autoimmune signature in their BAL that was not present systemically, regardless of disease. The proportion of patients with IPF with a local autoantibody signature was comparable to that of CTD-ILD, which has a known autoimmune pathology, identifying a potentially novel subset of patients. The presence of an airway autoimmune signature was not associated with reduced survival probability or changes in lung function in the cohort as a whole. Patients with IPF had increased BAL total IgA and IgG1 while subjects with CHP had increased BAL IgA, IgG1 and IgG4. In patients with CHP, increased BAL total IgA was associated with reduced survival probability.ConclusionAirway autoantibodies that are not present systemically identify a group of patients with fILD and the mechanisms by which these autoantibodies contribute to disease requires further investigation.
- Published
- 2021
47. Cluster analysis of transcriptomic datasets to identify endotypes of Idiopathic Pulmonary Fibrosis
- Author
-
Philip L. Molyneaux, Louise V. Wain, Toby M. Maher, R. Gisli Jenkins, Imre Noth, Adam Taylor, David A. Shwartz, Ivana V. Yang, Marco Mura, S.F. Ma, Yong Huang, Luke M. Kraven, Astrid Yeo, John E. McDonough, and William A. Fahy
- Subjects
Transcriptome ,Idiopathic pulmonary fibrosis ,business.industry ,Hazard ratio ,Medicine ,In patient ,Mitochondrial homeostasis ,Disease ,Computational biology ,business ,medicine.disease ,Lung function ,Confidence interval - Abstract
BackgroundConsiderable clinical heterogeneity in Idiopathic Pulmonary Fibrosis (IPF) suggests the existence of multiple disease endotypes. Identifying these endotypes could allow for a biomarker-driven personalised medicine approach in IPF. To improve our understanding of the pathogenesis of IPF by identifying clinically distinct groups of patients with IPF that could represent distinct disease endotypes.MethodsWe co-normalised, pooled and clustered three publicly available blood transcriptomic datasets (total 220 IPF cases). We compared clinical traits across clusters and used gene enrichment analysis to identify biological pathways and processes that were over-represented among the genes that were differentially expressed across clusters. A gene-based classifier was developed and validated using three additional independent datasets (total 194 IPF cases).FindingsWe identified three clusters of IPF patients with statistically significant differences in lung function (P=0·009) and mortality (P=0·009) between groups. Gene enrichment analysis implicated dysregulation of mitochondrial homeostasis, apoptosis, cell cycle and innate and adaptive immunity in the pathogenesis underlying these groups. We developed and validated a 13-gene cluster classifier that predicted mortality in IPF (high-risk clusters vs low-risk cluster: hazard ratio= 4·25, 95% confidence interval= [2·14, 8·46], P=3·7×10−5).InterpretationWe have identified blood gene expression signatures capable of discerning groups of IPF patients with significant differences in survival. These clusters could be representative of distinct pathophysiological states, which would support the theory of multiple endotypes of IPF. Although more work must be done to confirm the existence of these endotypes, our classifier could be a useful tool in patient stratification and outcome prediction in IPF.FundingL.V.W. holds a GSK/British Lung Foundation Chair in Respiratory Research (C17-1). R.G.J. is supported by a National Institute for Health Research (NIHR) Research Professorship (NIHR reference RP-2017-08-ST2-014). P.L.M. is supported by an Action for Pulmonary Fibrosis Mike Bray fellowship. T.M. Maher is supported by a National Institute for Health Research Clinician Scientist Fellowship (CS-2013-13-017) and a British Lung Foundation Chair in Respiratory Research (C17-3). I.N. is supported by a National Heart, Lung, and Blood Institute (NHLBI) grant (R01HL145266). D.A.S. is supported by NHLBI grants (UG3HL151865, R01HL097163, P01HL092870, X01HL134585 and UH3HL123442) and a United States Department of Defense grant (W81XWH-17-1-0597). The GSE110147 study was supported by the Roche Multi Organ Transplant Academic Enrichment Fund, Lawson Research Institute Internal Research Fund and Western Strategic Support for CIHR Success, Seed Grant. The research was partially supported by the NIHR Leicester Biomedical Research Centre; the views expressed are those of the author(s) and not necessarily those of the National Health Service (NHS), the NIHR, or the Department of Health.Putting research into contextEvidence before this studyWe searched PubMed Central in February 2020 with the search terms “idiopathic pulmonary fibrosis”, “gene expression” and “cluster analysis” with no restrictions on publication date or language. Previous transcriptomic cluster analyses have found that differences in gene expression can be used to predict disease status, severity and outcome in IPF. A previous transcriptomic prognostic biomarker has been developed that can predict outcome in IPF using blood expression data from 52 genes.Added value of this studyBy utilising new methods of data co-normalisation and machine learning, we were able to combine multiple publicly available datasets and perform one of the largest transcriptomic studies in IPF to-date with a total of 416 IPF cases across the discovery and validation stages. We identified three clusters of patients, one of which appeared to contain, on average, the healthiest subjects with favourable lung function and survival over time. These clusters were defined using expression from groups of genes that were significantly enriched for many different biological pathways and processes, including metabolic changes, apoptosis, cell cycle and immune response, and so could be representative of distinct pathophysiological states. Additionally, we developed a 13-gene expression-based classifier to assign individuals with IPF to one of the clusters and validated this classifier using three additional independent cohort of IPF patients (totalling 194 IPF cases). As the clusters were associated with survival, our classifier could potentially be used to predict outcome in IPF.Implications of all the available evidenceOur findings support the hypothesis that the disease consists of multiple endotypes. The clusters identified in this study could provide some valuable insight into the underlying biological processes that may be driving the considerable clinical heterogeneity in IPF. With further development, our gene expression-based classifier could be a useful tool for patient stratification and outcome prediction in IPF.
- Published
- 2021
- Full Text
- View/download PDF
48. Common idiopathic pulmonary fibrosis risk variants are associated with hypersensitivity pneumonitis
- Author
-
Haruhiko Furusawa, Anna L Peljto, Avram D Walts, Jonathan Cardwell, Philip L Molyneaux, Joyce S Lee, Evans R Fernández Pérez, Paul J Wolters, Ivana V Yang, David A Schwartz, and Action for Pulmonary Fibrosis
- Subjects
Pulmonary and Respiratory Medicine ,Risk Factors ,Respiratory System ,Humans ,1103 Clinical Sciences ,respiratory system ,hypersensitivity pneumonitis ,Fibrosis ,Lung ,Idiopathic Pulmonary Fibrosis ,Article ,respiratory tract diseases ,Alveolitis, Extrinsic Allergic - Abstract
A subset of patients with hypersensitivity pneumonitis (HP) develop lung fibrosis that is clinically similar to idiopathic pulmonary fibrosis (IPF). To address the aetiological determinants of fibrotic HP, we investigated whether the common IPF genetic risk variants were also relevant in study subjects with fibrotic HP. Our findings indicate that common genetic variants in TERC, DSP, MUC5B and IVD were significantly associated with fibrotic HP. These findings provide support for a shared etiology and pathogenesis between fibrotic HP and IPF.
- Published
- 2021
49. PAciFy Cough-a multicentre, double-blind, placebo-controlled, crossover trial of morphine sulphate for the treatment of pulmonary Fibrosis Cough
- Author
-
Zhe Wu, Winston Banya, Nazia Chaudhuri, Ira Jakupovic, Toby M. Maher, Brijesh Patel, Lisa G. Spencer, Muhunthan Thillai, Alex West, John Westoby, Marlies Wijsenbeek, Jaclyn Smith, Philip L. Molyneaux, Pulmonary Medicine, National Institute for Health Research, British Lung Foundation, Molyneaux, Philip L [0000-0003-1301-8800], and Apollo - University of Cambridge Repository
- Subjects
Cross-Over Studies ,Morphine ,Medicine (miscellaneous) ,Interstitial lung disease ,1103 Clinical Sciences ,Idiopathic Pulmonary Fibrosis ,respiratory tract diseases ,Study Protocol ,Idiopathic Pulmonary Fibrosis/drug therapy ,Treatment Outcome ,Cardiovascular System & Hematology ,SDG 3 - Good Health and Well-being ,Cough ,Double-Blind Method ,General & Internal Medicine ,Quality of Life ,Humans ,Morphine/adverse effects ,Pharmacology (medical) ,Cough/diagnosis ,1102 Cardiorespiratory Medicine and Haematology - Abstract
Funder: The Jon Moulton Charity Trust, BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive disease that leads to lung scarring. Cough is reported by 85% of patients with IPF and can be a distressing symptom with a significant impact on patients' quality of life. There are no proven effective therapies for IPF-related cough. Whilst morphine is frequently used as a palliative agent for breathlessness in IPF, its effects on cough have never been tested. PAciFy Cough is a multicenter, double-blind, placebo-controlled, crossover trial of morphine sulphate for the treatment of cough in IPF. METHODS: We will recruit 44 subjects with IPF prospectively from three interstitial lung disease units in the UK, namely the Royal Brompton Hospital, Manchester University NHS Foundation Trust (MFT) and Aintree University Hospital NHS Foundation Trust. Patients will be randomised (1:1) to either placebo twice daily or morphine sulphate 5 mg twice daily for 14 days. They will then crossover after a 7-day washout period. The primary endpoint is the percent change in daytime cough frequency (coughs per hour) from baseline as assessed by objective cough monitoring at day 14 of treatment. DISCUSSION: This multicentre, randomised trial will assess the effect of opioids on cough counts and cough associated quality of life in IPF subjects. If proven to be an effective intervention, it represents a readily available treatment for patients. TRIAL REGISTRATION: The study was approved by the UK Medicines and Healthcare Regulatory Agency (Ref: CTA 21268/0224/001-0001 - EUDRACT 2019-003571-19 - Protocol Number RBH2019/001) on 08 April 2020, in compliance with the European Clinical Trials Directive and the Medicines for Human Use (Clinical Trials) Regulations 2004 and its subsequent amendments. The study was provided with ethical approval by the London Brent Research Ethics Committee (Ref: 20/LO/0368) on 21 May 2020 and is registered with clinicaltrials.gov (NCT04429516) on 12 June 2020, available at https://clinicaltrials.gov/ct2/show/NCT04429516.
- Published
- 2021
- Full Text
- View/download PDF
50. Blood Transcriptomics Predicts Progression of Pulmonary Fibrosis and Associated Natural Killer Cells
- Author
-
Jose D. Herazo-Maya, Philip L. Molyneaux, Shu Yi Liao, Rekha Vij, Ayodeji Adegunsoye, Catherine A. Bonham, Andrew J. Barros, Avraham Unterman, Fernando J. Martinez, Shwu Fan Ma, Naftali Kaminski, Justin M. Oldham, Yong Huang, Mary E. Strek, John S. Kim, Toby M. Maher, Imre Noth, Bethany B. Moore, and Action for Pulmonary Fibrosis
- Subjects
Pulmonary and Respiratory Medicine ,Male ,Respiratory System ,Critical Care and Intensive Care Medicine ,Disease activity ,Transcriptome ,Cohort Studies ,Idiopathic pulmonary fibrosis ,Predictive Value of Tests ,Pulmonary fibrosis ,Medicine ,Humans ,relative decline of FVC ,Longitudinal Studies ,Respiratory system ,Relative decline of forced vital capacity ,11 Medical and Health Sciences ,Aged ,cell type composition deconvolution ,business.industry ,Original Articles ,respiratory system ,Middle Aged ,medicine.disease ,Idiopathic Pulmonary Fibrosis ,respiratory tract diseases ,Killer Cells, Natural ,longitudinal changes of blood gene expression ,Cross-Sectional Studies ,multigene predictor for progression ,Immunology ,Disease Progression ,Female ,business ,Biomarkers - Abstract
Rationale: Disease activity in idiopathic pulmonary fibrosis (IPF) remains highly variable, poorly understood, and difficult to predict. Objectives: To identify a predictor using short-term longitudinal changes in gene expression that forecasts future FVC decline and to characterize involved pathways and cell types. Methods: Seventy-four patients from COMET (Correlating Outcomes with Biochemical Markers to Estimate Time-Progression in IPF) cohort were dichotomized as progressors (≥10% FVC decline) or stable. Blood gene-expression changes within individuals were calculated between baseline and 4 months and regressed with future FVC status, allowing determination of expression variations, sample size, and statistical power. Pathway analyses were conducted to predict downstream effects and identify new targets. An FVC predictor for progression was constructed in COMET and validated using independent cohorts. Peripheral blood mononuclear single-cell RNA-sequencing data from healthy control subjects were used as references to characterize cell type compositions from bulk peripheral blood mononuclear RNA-sequencing data that were associated with FVC decline. Measurements and Main Results: The longitudinal model reduced gene-expression variations within stable and progressor groups, resulting in increased statistical power when compared with a cross-sectional model. The FVC predictor for progression anticipated patients with future FVC decline with 78% sensitivity and 86% specificity across independent IPF cohorts. Pattern recognition receptor pathways and mTOR pathways were downregulated and upregulated, respectively. Cellular deconvolution using single-cell RNA-sequencing data identified natural killer cells as significantly correlated with progression. Conclusions: Serial transcriptomic change predicts future FVC decline. An analysis of cell types involved in the progressor signature supports the novel involvement of natural killer cells in IPF progression.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.