355 results on '"DA Lomas"'
Search Results
2. P121 Cost-Effectiveness of Alpha-1 Antitrypsin (A1AT) Deficiency Case-Finding in Secondary Care: Abstract P121 TABLE 1
- Author
-
B Gooptu, DA Lomas, John R. Hurst, Massimo Pinzani, Douglas Thorburn, S Whiting, and L Dron
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Pathology ,In vivo degradation ,business.industry ,Cost effectiveness ,Alpha (ethology) ,Hepatology ,Gastroenterology ,Secondary care ,Molecular genetics ,Internal medicine ,Medicine ,Case finding ,business ,Genotyping - Abstract
Background Screening for A1AT-deficiency at our institution involves serum protein assay, then phenotyping where A1AT≤1.1g/L. RT-PCR genotyping for S/Z variants is available for clarification. We recently established a multi-disciplinary respiratory/hepatology London A1AT Deficiency Service and re-examined the cost-effectiveness of our diagnostic algorithm. Method We studied all patients who had a serum A1AT request over the three years 1/1/2010–31/12/2012. We went on to examine all requests for phenotyping and/or genotyping over the ten years 1/1/2003–31/12/2012. Results From 4460 requests over 2010–2012, 240(5.4%) had serum A1AT≤1.1g/l. Of these, phenotyping was not available in 33 and of the remainder the following phenotype-prevalence data were observed (n,%): MM (75,36%), MZ (89,43%), MS (29,14%), ZZ (6,3%), SZ (6,3%), SS (1,1%), GM (1,1%). In error, 28 patients had phenotyping with A1AT>1.1g/l: 27 were PiMM and 1 PiMS. Over 2003–2012, 525 phenotyping tests were performed. In 41 the result was unclear or there was evidence of in vivo degradation. The prevalence of the respective phenotypes and mean (SD) A1AT concentrations are reported in the Table. Over the same period, 24 equivocal samples were sent for genotyping. This confirmed 13 = PiMM, 4 = PiZZ, 3 = PiMZ, 2 = PiMS and 2 = PiSZ. The NHS costs of A1AT serum assay, phenotyping and genotyping are £2.45, £35.50 and £87.50. Using the 2010–2012 data, our mean annualised spend on A1AT serum, phenotyping and genotyping was £3,642 £3,170 and £376 respectively. The cost per ZZ/SZ case diagnosed was £1198. The highest A1AT serum concentration recorded in the ZZ/SZ patients were 0.4 and 1.0g/l respectively suggesting that the ≤1.1g/l cut-off is appropriate. The specificity, sensitivity and positive predictive values are 26.5%, 100% and 6.5%. Conclusion 5.4% of serum A1AT assays yielded results ≤1.1g/l. Of these, 6% were found to represent SZ/ZZ variants at risk of clinically relevant disease (0.27%, 1/372 requests). The ≤1.1g/l cut-off was 100% sensitive but only 26.5% specific for clinically relevant deficiency alleles. The ≤1.1g/l cut-off could be lowered with improved specificity and cost-saving if the desire was to detect only ZZ patients.
- Published
- 2013
3. A genome-wide analysis of the response to inhaled β2-agonists in chronic obstructive pulmonary disease
- Author
-
Hardin M, Mh, Cho, Ml, Mcdonald, Wan E, Da, Lomas, Ho, Coxson, William MacNee, Vestbo J, Jc, Yates, Agusti A, Pm, Calverley, Celli B, Crim C, Rennard S, Wouters E, Bakke P, Sp, Bhatt, Kim V, Ramsdell J, and Ea, Regan
4. Fibrosis, biomarkers and liver biopsy in AAT deficiency and relation to liver Z protein polymer accumulation.
- Author
-
Suri A, Zhang Z, Neuschwander-Tetri B, Lomas DA, Heyer-Chauhan N, Burling K, Loomba R, Brenner DA, Nagy R, Wilson A, Carpenter D, Blomenkamp K, and Teckman J
- Abstract
Background and Aims: The course of adults with ZZ alpha-1-antitrypsin deficiency (AATD) liver disease is unpredictable. The utility of markers, including liver biopsy, is undefined., Methods: A prospective cohort, including protocol liver biopsies, was enrolled to address these questions., Results: We enrolled 96 homozygous ZZ AATD adults prospectively at three US sites with standardized clinical evaluations, and protocol liver biopsies. Fibrosis was scored using Ishak (stages 0-6). Also, 51% of the 96 subjects had Ishak score >1 fibrosis (49% Ishak 0-1, 36% Ishak 2-3 and 15% ≥4). Elevated aspartate aminotransferase (AST) more than alanine aminotransferase (ALT), high body mass index (BMI), obesity, AST platelet ratio index and elevated serum Z alpha 1 antitrypsin (AAT) polymer levels were associated with increased fibrosis. Steatosis did not correlate to fibrosis. Increased fibrosis was associated with increased mutant Z polymer globular inclusions (p = .002) and increased diffuse cytoplasmic Z polymer on biopsy (p = .0029) in a direct relationship. Increased globule Z polymer was associated with increased serum AST (p = .007) and increased periportal inflammation on histopathology (p = .004), but there was no relationship of Z polymer hepatocellular accumulation with ALT, gamma glutamine transferase, inflammation in other parts of the lobule, necrosis or steatosis. Serum Z polymer levels were directly correlated to hepatic Z protein polymer content. Lung function, smoking and alcohol consumption patterns were not associated with fibrosis., Conclusion: In AATD high BMI, obesity and elevated AST are associated with increased fibrosis. Liver biopsy features are correlated to some serum tests. Serum Z AAT polymer levels could be a future biomarker to detect fibrosis early and is directly correlated to liver Z content., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
5. Reduction of Z alpha-1 antitrypsin polymers in human iPSC-hepatocytes and mice by LRRK2 inhibitors.
- Author
-
Kent D, Ng SS, Syanda AM, Khoshkenar P, Ronzoni R, Li CZ, Zieger M, Greer C, Hatch S, Segal J, Blackford SJI, Im YR, Chowdary V, Ismaili T, Danovi D, Lewis PA, Irving JA, Sahdeo S, Lomas DA, Ebner D, Mueller C, and Rashid ST
- Abstract
Background: Alpha-1 antitrypsin deficiency (A1ATD) is a life-threatening condition caused by the inheritance of the serpin family A member 1 "Z" genetic variant driving alpha-1 antitrypsin (AAT) protein misfolding in hepatocytes. There are no approved medicines for this disease., Methods: We conducted a high-throughput image-based small molecule screen using patient-derived induced pluripotent stem cell-hepatocytes (iPSC-hepatocytes). Identified targets were validated in vitro using 3 independent patient iPSC lines. The effects of the identified target, leucine-rich repeat kinase 2 (LRRK2), were further evaluated in an animal model of A1ATD through histology and immunohistochemistry and in an autophagy-reporter line. Autophagy induction was assessed through immunoblot and immunofluorescence analyses., Results: Small-molecule screen performed in iPSC-hepatocytes identified LRRK2 as a potentially new therapeutic target. Of the commercially available LRRK2 inhibitors tested, we identified CZC-25146, a candidate with favorable pharmacokinetic properties, as capable of reducing polymer load, increasing normal AAT secretion, and reducing inflammatory cytokines in both cells and PiZ mice. Mechanistically, this effect was achieved through the induction of autophagy., Conclusions: Our findings support the use of CZC-25146 and leucine-rich repeat kinase-2 inhibitors in hepatic proteinopathy research and their further investigation as novel therapeutic candidates for A1ATD., (Copyright © 2024 American Association for the Study of Liver Diseases.)
- Published
- 2024
- Full Text
- View/download PDF
6. A novel pathological mutant reveals the role of torsional flexibility in the serpin breach in adoption of an aggregation-prone intermediate.
- Author
-
Kamuda K, Ronzoni R, Majumdar A, Guan FHX, Irving JA, and Lomas DA
- Subjects
- Humans, Crystallography, X-Ray, Mutation, Models, Molecular, Protein Aggregates, Protein Aggregation, Pathological genetics, Protein Aggregation, Pathological metabolism, Protein Aggregation, Pathological pathology, Protein Conformation, Endoplasmic Reticulum metabolism, Endoplasmic Reticulum genetics, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin chemistry, alpha 1-Antitrypsin metabolism
- Abstract
Mutants of alpha-1-antitrypsin cause the protein to self-associate and form ordered aggregates ('polymers') that are retained within hepatocytes, resulting in a predisposition to the development of liver disease. The associated reduction in secretion, and for some mutants, impairment of function, leads to a failure to protect lung tissue against proteases released during the inflammatory response and an increased risk of emphysema. We report here a novel deficiency mutation (Gly192Cys), that we name the Sydney variant, identified in a patient in heterozygosity with the Z allele (Glu342Lys). Cellular analysis revealed that the novel variant was mostly retained as insoluble polymers within the endoplasmic reticulum. The basis for this behaviour was investigated using biophysical and structural techniques. The variant showed a 40% reduction in inhibitory activity and a reduced stability as assessed by thermal unfolding experiments. Polymerisation involves adoption of an aggregation-prone intermediate and paradoxically the energy barrier for transition to this state was increased by 16% for the Gly192Cys variant with respect to the wild-type protein. However, with activation to the intermediate state, polymerisation occurred at a 3.8-fold faster rate overall. X-ray crystallography provided two crystal structures of the Gly192Cys variant, revealing perturbation within the 'breach' region with Cys192 in two different orientations: in one structure it faces towards the hydrophobic core while in the second it is solvent-exposed. This orientational heterogeneity was confirmed by PEGylation. These data show the critical role of the torsional freedom imparted by Gly192 in inhibitory activity and stability against polymerisation., (© 2024 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
7. Biomarkers Associated With Future Severe Liver Disease in Children With Alpha-1-Antitrypsin Deficiency.
- Author
-
Teckman JH, Buchanan P, Blomenkamp KS, Heyer-Chauhan N, Burling K, and Lomas DA
- Abstract
Background and Aims: Children with alpha-1-antitrypsin deficiency (AATD) exhibit a wide range of liver disease outcomes from portal hypertension and transplant to asymptomatic without fibrosis. Individual outcomes cannot be predicted. Liver injury in AATD is caused by the accumulation in hepatocytes of the mutant Z alpha-1-antitrypsin (AAT) protein, especially the toxic, intracellular polymerized conformation. AATD patients have trace Z polymer detectable in serum with unknown significance., Methods: The Childhood Liver Disease Research Network is an NIH consortium for the study of pediatric liver diseases, including AATD. We obtained data and samples with the aim of identifying biomarkers predictive of severe AATD liver disease., Results: We analyzed prospective AATD Childhood Liver Disease Research Network data and serum samples in 251 subjects from 2007 to 2015 for outcomes and Z polymer levels. Fifty-eight of 251 had clinically evident portal hypertension (CEPH) at enrollment, and 10 developed CEPH during follow-up. Higher Z AAT polymer levels were associated with existing CEPH ( P = .01). In infants without CEPH, higher polymer levels were associated with future CEPH later in childhood, but total AAT was not predictive. Higher gamma-glutamyl transferase (GGT) in the first few months of life was also significantly associated with future CEPH, and risk-threshold GGT levels can be identified. A model was constructed to identify subjects at high risk of future CEPH by combining clinical GGT and polymer levels (area under the curve of 0.83; 95% confidence interval: 0.656-1.00, P = .019)., Conclusion: High circulating Z polymer levels and high GGT early in life are associated with future CEPH in AATD, and the use of predictive cutoffs may assist in future clinical trial design., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
8. Native and Ion Mobility Mass Spectrometry Characterization of Alpha 1 Antitrypsin Variants and Oligomers.
- Author
-
Vickers S, Irving J, Lomas DA, and Thalassinos K
- Subjects
- Plasma, Workflow, Mass Spectrometry, alpha 1-Antitrypsin genetics, Ion Mobility Spectrometry
- Abstract
In this chapter, we describe a method for analyzing both recombinant and plasma-derived alpha 1 antitrypsin and its oligomers by means of native ion mobility mass spectrometry. Our experimental workflow can be applied to other variants of alpha 1 antitrypsin and its oligomers as well as being used to probe their interactions with small molecules in the gas phase., (© 2024. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
9. The under-appreciated world of the serpin family of serine proteinase inhibitors.
- Author
-
Bouton MC, Geiger M, Sheffield WP, Irving JA, Lomas DA, Song S, Satyanarayanan RS, Zhang L, McFadden G, and Lucas AR
- Subjects
- Animals, Humans, Serine Proteinase Inhibitors pharmacology, Serine Proteinase Inhibitors chemistry, Serine Proteinase Inhibitors metabolism, Serine Proteases metabolism, Inflammation, Serpins genetics, Serpins chemistry, Serpins metabolism
- Abstract
In the practice of medicine, many fundamental biological pathways that require tight on/off control, such as inflammation and circulatory homeostasis, are regulated by serine proteinases, but we rarely consider the unique protease inhibitors that, in turn, regulate these proteases. The serpins are a family of proteins with a shared tertiary structure, whose members largely act as serine protease inhibitors, found in all forms of life, ranging from viruses, bacteria, and archaea to plants and animals. These proteins represent up to 2-10% of proteins in the human blood and are the third most common protein family., (© 2023 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2023
- Full Text
- View/download PDF
10. X chromosome associations with chronic obstructive pulmonary disease and related phenotypes: an X chromosome-wide association study.
- Author
-
Hayden LP, Hobbs BD, Busch R, Cho MH, Liu M, Lopes-Ramos CM, Lomas DA, Bakke P, Gulsvik A, Silverman EK, Crapo JD, Beaty TH, Laird NM, Lange C, and DeMeo DL
- Subjects
- Female, Male, Humans, Genetic Predisposition to Disease genetics, Genome-Wide Association Study, Phenotype, X Chromosome, Pulmonary Disease, Chronic Obstructive diagnosis, Pulmonary Disease, Chronic Obstructive epidemiology, Pulmonary Disease, Chronic Obstructive genetics, Pulmonary Emphysema, Emphysema
- Abstract
Background: The association between genetic variants on the X chromosome to risk of COPD has not been fully explored. We hypothesize that the X chromosome harbors variants important in determining risk of COPD related phenotypes and may drive sex differences in COPD manifestations., Methods: Using X chromosome data from three COPD-enriched cohorts of adult smokers, we performed X chromosome specific quality control, imputation, and testing for association with COPD case-control status, lung function, and quantitative emphysema. Analyses were performed among all subjects, then stratified by sex, and subsequently combined in meta-analyses., Results: Among 10,193 subjects of non-Hispanic white or European ancestry, a variant near TMSB4X, rs5979771, reached genome-wide significance for association with lung function measured by FEV
1 /FVC ([Formula: see text] 0.020, SE 0.004, p 4.97 × 10-08 ), with suggestive evidence of association with FEV1 ([Formula: see text] 0.092, SE 0.018, p 3.40 × 10-07 ). Sex-stratified analyses revealed X chromosome variants that were differentially trending in one sex, with significantly different effect sizes or directions., Conclusions: This investigation identified loci influencing lung function, COPD, and emphysema in a comprehensive genetic association meta-analysis of X chromosome genetic markers from multiple COPD-related datasets. Sex differences play an important role in the pathobiology of complex lung disease, including X chromosome variants that demonstrate differential effects by sex and variants that may be relevant through escape from X chromosome inactivation. Comprehensive interrogation of the X chromosome to better understand genetic control of COPD and lung function is important to further understanding of disease pathology. Trial registration Genetic Epidemiology of COPD Study (COPDGene) is registered at ClinicalTrials.gov, NCT00608764 (Active since January 28, 2008). Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints Study (ECLIPSE), GlaxoSmithKline study code SCO104960, is registered at ClinicalTrials.gov, NCT00292552 (Active since February 16, 2006). Genetics of COPD in Norway Study (GenKOLS) holds GlaxoSmithKline study code RES11080, Genetics of Chronic Obstructive Lung Disease., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
11. Keratin 8 is a scaffolding and regulatory protein of ERAD complexes.
- Author
-
Pranke IM, Chevalier B, Premchandar A, Baatallah N, Tomaszewski KF, Bitam S, Tondelier D, Golec A, Stolk J, Lukacs GL, Hiemstra PS, Dadlez M, Lomas DA, Irving JA, Delaunay-Moisan A, van Anken E, Hinzpeter A, Sermet-Gaudelus I, and Edelman A
- Subjects
- HeLa Cells, Humans, Transcription Factors metabolism, Ubiquitin-Protein Ligases metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Endoplasmic Reticulum-Associated Degradation, Keratin-8 metabolism
- Abstract
Early recognition and enhanced degradation of misfolded proteins by the endoplasmic reticulum (ER) quality control and ER-associated degradation (ERAD) cause defective protein secretion and membrane targeting, as exemplified for Z-alpha-1-antitrypsin (Z-A1AT), responsible for alpha-1-antitrypsin deficiency (A1ATD) and F508del-CFTR (cystic fibrosis transmembrane conductance regulator) responsible for cystic fibrosis (CF). Prompted by our previous observation that decreasing Keratin 8 (K8) expression increased trafficking of F508del-CFTR to the plasma membrane, we investigated whether K8 impacts trafficking of soluble misfolded Z-A1AT protein. The subsequent goal of this study was to elucidate the mechanism underlying the K8-dependent regulation of protein trafficking, focusing on the ERAD pathway. The results show that diminishing K8 concentration in HeLa cells enhances secretion of both Z-A1AT and wild-type (WT) A1AT with a 13-fold and fourfold increase, respectively. K8 down-regulation triggers ER failure and cellular apoptosis when ER stress is jointly elicited by conditional expression of the µ
s heavy chains, as previously shown for Hrd1 knock-out. Simultaneous K8 silencing and Hrd1 knock-out did not show any synergistic effect, consistent with K8 acting in the Hrd1-governed ERAD step. Fractionation and co-immunoprecipitation experiments reveal that K8 is recruited to ERAD complexes containing Derlin2, Sel1 and Hrd1 proteins upon expression of Z/WT-A1AT and F508del-CFTR. Treatment of the cells with c407, a small molecule inhibiting K8 interaction, decreases K8 and Derlin2 recruitment to high-order ERAD complexes. This was associated with increased Z-A1AT secretion in both HeLa and Z-homozygous A1ATD patients' respiratory cells. Overall, we provide evidence that K8 acts as an ERAD modulator. It may play a scaffolding protein role for early-stage ERAD complexes, regulating Hrd1-governed retrotranslocation initiation/ubiquitination processes. Targeting K8-containing ERAD complexes is an attractive strategy for the pharmacotherapy of A1ATD., (© 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)- Published
- 2022
- Full Text
- View/download PDF
12. Correction to: Single-Inhaler Triple Therapy and Health-Related Quality of Life in COPD: The IMPACT Study.
- Author
-
Tabberer M, Jones CE, Kilbride S, Halpin DMG, Lomas DA, Pascoe S, Singh D, Wise RA, Criner GJ, Lange P, Dransfield MT, Han MK, Martinez FJ, Kaisermann MC, and Lipson DA
- Published
- 2022
- Full Text
- View/download PDF
13. Hepatobiliary phenotypes of adults with alpha-1 antitrypsin deficiency.
- Author
-
Fromme M, Schneider CV, Pereira V, Hamesch K, Pons M, Reichert MC, Benini F, Ellis P, H Thorhauge K, Mandorfer M, Burbaum B, Woditsch V, Chorostowska-Wynimko J, Verbeek J, Nevens F, Genesca J, Miravitlles M, Nuñez A, Schaefer B, Zoller H, Janciauskiene S, Abreu N, Jasmins L, Gaspar R, Liberal R, Macedo G, Mahadeva R, Gomes C, Schneider KM, Trauner M, Krag A, Gooptu B, Thorburn D, Marshall A, Hurst JR, Lomas DA, Lammert F, Gaisa NT, Clark V, Griffiths W, Trautwein C, Turner AM, McElvaney NG, and Strnad P
- Subjects
- Adult, Aged, Case-Control Studies, Cohort Studies, Female, Humans, Male, Middle Aged, Phenotype, Prevalence, United Kingdom, Cholelithiasis epidemiology, Liver Cirrhosis epidemiology, Liver Neoplasms epidemiology, alpha 1-Antitrypsin Deficiency complications
- Abstract
Objective: Alpha-1 antitrypsin deficiency (AATD) is a common, potentially lethal inborn disorder caused by mutations in alpha-1 antitrypsin (AAT). Homozygosity for the 'Pi*Z' variant of AAT (Pi*ZZ genotype) causes lung and liver disease, whereas heterozygous 'Pi*Z' carriage (Pi*MZ genotype) predisposes to gallstones and liver fibrosis. The clinical significance of the more common 'Pi*S' variant remains largely undefined and no robust data exist on the prevalence of liver tumours in AATD., Design: Baseline phenotypes of AATD individuals and non-carriers were analysed in 482 380 participants in the UK Biobank. 1104 participants of a multinational cohort (586 Pi*ZZ, 239 Pi*SZ, 279 non-carriers) underwent a comprehensive clinical assessment. Associations were adjusted for age, sex, body mass index, diabetes and alcohol consumption., Results: Among UK Biobank participants, Pi*ZZ individuals displayed the highest liver enzyme values, the highest occurrence of liver fibrosis/cirrhosis (adjusted OR (aOR)=21.7 (8.8-53.7)) and primary liver cancer (aOR=44.5 (10.8-183.6)). Subjects with Pi*MZ genotype had slightly elevated liver enzymes and moderately increased odds for liver fibrosis/cirrhosis (aOR=1.7 (1.2-2.2)) and cholelithiasis (aOR=1.3 (1.2-1.4)). Individuals with homozygous Pi*S mutation (Pi*SS genotype) harboured minimally elevated alanine aminotransferase values, but no other hepatobiliary abnormalities. Pi*SZ participants displayed higher liver enzymes, more frequent liver fibrosis/cirrhosis (aOR=3.1 (1.1-8.2)) and primary liver cancer (aOR=6.6 (1.6-26.9)). The higher fibrosis burden was confirmed in a multinational cohort. Male sex, age ≥50 years, obesity and the presence of diabetes were associated with significant liver fibrosis., Conclusion: Our study defines the hepatobiliary phenotype of individuals with the most relevant AATD genotypes including their predisposition to liver tumours, thereby allowing evidence-based advice and individualised hepatological surveillance., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2022. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
14. Genetic variation in genes regulating skeletal muscle regeneration and tissue remodelling associated with weight loss in chronic obstructive pulmonary disease.
- Author
-
Lakshman Kumar P, Wilson AC, Rocco A, Cho MH, Wan E, Hobbs BD, Washko GR, Ortega VE, Christenson SA, Li X, Wells JM, Bhatt SP, DeMeo DL, Lutz SM, Rossiter H, Casaburi R, Rennard SI, Lomas DA, Labaki WW, Tal-Singer R, Bowler RP, Hersh CP, Tiwari HK, Dransfield M, Thalacker-Mercer A, Meyers DA, Silverman EK, and McDonald MN
- Subjects
- Adult, Bayes Theorem, Genetic Variation, Humans, Muscle, Skeletal, Nerve Tissue Proteins, Regeneration, Weight Loss genetics, Genome-Wide Association Study, Pulmonary Disease, Chronic Obstructive genetics
- Abstract
Background: Chronic obstructive pulmonary disease (COPD) is the third leading cause of death globally. COPD patients with cachexia or weight loss have increased risk of death independent of body mass index (BMI) and lung function. We tested the hypothesis genetic variation is associated with weight loss in COPD using a genome-wide association study approach., Methods: Participants with COPD (N = 4308) from three studies (COPDGene, ECLIPSE, and SPIROMICS) were analysed. Discovery analyses were performed in COPDGene with replication in SPIROMICS and ECLIPSE. In COPDGene, weight loss was defined as self-reported unintentional weight loss > 5% in the past year or low BMI (BMI < 20 kg/m
2 ). In ECLIPSE and SPIROMICS, weight loss was calculated using available longitudinal visits. Stratified analyses were performed among African American (AA) and Non-Hispanic White (NHW) participants with COPD. Single variant and gene-based analyses were performed adjusting for confounders. Fine mapping was performed using a Bayesian approach integrating genetic association results with linkage disequilibrium and functional annotation. Significant gene networks were identified by integrating genetic regions associated with weight loss with skeletal muscle protein-protein interaction (PPI) data., Results: At the single variant level, only the rs35368512 variant, intergenic to GRXCR1 and LINC02383, was associated with weight loss (odds ratio = 3.6, 95% confidence interval = 2.3-5.6, P = 3.2 × 10-8 ) among AA COPD participants in COPDGene. At the gene level in COPDGene, EFNA2 and BAIAP2 were significantly associated with weight loss in AA and NHW COPD participants, respectively. The EFNA2 association replicated among AA from SPIROMICS (P = 0.0014), whereas the BAIAP2 association replicated in NHW from ECLIPSE (P = 0.025). The EFNA2 gene encodes the membrane-bound protein ephrin-A2 involved in the regulation of developmental processes and adult tissue homeostasis such as skeletal muscle. The BAIAP2 gene encodes the insulin-responsive protein of mass 53 kD (IRSp53), a negative regulator of myogenic differentiation. Integration of the gene-based findings participants with PPI data revealed networks of genes involved in pathways such as Rho and synapse signalling., Conclusions: The EFNA2 and BAIAP2 genes were significantly associated with weight loss in COPD participants. Collectively, the integrative network analyses indicated genetic variation associated with weight loss in COPD may influence skeletal muscle regeneration and tissue remodelling., (© 2021 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.)- Published
- 2021
- Full Text
- View/download PDF
15. A single blood eosinophil count measurement is as good as two for prediction of ICS treatment response in the IMPACT trial.
- Author
-
Bafadhel M, Barnes N, Bourke SC, Compton C, Criner GJ, Dransfield MT, Halpin DMG, Han MK, Hartley B, Jones CE, Lange P, Lettis S, Lipson DA, Lomas DA, Martin N, Martinez FJ, Wise R, and Singh D
- Subjects
- Administration, Inhalation, Adrenal Cortex Hormones therapeutic use, Bronchodilator Agents therapeutic use, Clinical Trials as Topic, Humans, Leukocyte Count, Eosinophils, Pulmonary Disease, Chronic Obstructive drug therapy
- Abstract
Competing Interests: Conflict of interest: M. Bafadhel reports grants from AstraZeneca; advisory board attendance for AstraZeneca, Chiesi, Boehringer Ingelheim and GSK (in the last 3 years); attendance at educational meetings facilitated by AstraZeneca, Chiesi and Boehringer Ingelheim (in the last 3 years); and scientific advisor for ProAxsis and AlbusHealth. Conflict of interest: N. Barnes is an employee of GSK and holds stocks and shares in GSK. Conflict of interest: S.C. Bourke reports research grants from GSK, Philips, ResMed and Pfizer Open Air, support to attend scientific meetings from Boehringer Ingelheim, Chiesi, GSK and AstraZeneca and personal fees from Novartis, Chiesi and ResMed. Conflict of interest: C. Compton is an employee of GSK and holds stocks and shares in GSK. Conflict of interest: G.J. Criner reports personal fees from Almirall, Amgen, AstraZeneca, Boehringer Ingelheim, Broncus Medical, Chiesi, CSA Medical, Eolo, Gala Therapeutics, GSK, Helios Medical, Medtronic, Merck, Mereo BioPharma, NGM Pharmaceuticals, Novartis, Nuvaira, Olympus, Philips Respironics, Pulmonx, Respivant Sciences, The Implementation Group and Verona; and has ownership interest in HGE Technologies. Conflict of interest: M.T. Dransfield reports personal fees from AstraZeneca, Boehringer Ingelheim, PneumRx/BTG, Quark Pharmaceuticals and GSK, grant support from the American Lung Association, Department of Defense, Department of Veterans Affairs and NIH, and contracted clinical trial support from Boehringer Ingelheim, Novartis, AstraZeneca, Yungjin, PneumRx/BTG, Pulmonx, Boston Scientific, Gala, Nuvaira and GSK. Conflict of interest: D.M.G. Halpin reports personal fees from AstraZeneca, Boehringer Ingelheim, Chiesi, GSK, Novartis, Pfizer and Sanofi, and non-financial support from Boehringer Ingelheim and Novartis. Conflict of interest: M.K. Han reports personal fees from AstraZeneca, GSK, Mylan, Merck and Boehringer Ingelheim, and research support from Novartis and Sunovion. Conflict of interest: B. Hartley is a contingent worker with a contract research organisation working on behalf of GSK and holds shares in GSK. Conflict of interest: C.E. Jones is an employee of GSK and holds stocks and shares in GSK. Conflict of interest: P. Lange reports personal fees from GSK, AstraZeneca and Boehringer Ingelheim, and grant support from Boehringer Ingelheim and GSK. Conflict of interest: S. Lettis is an employee of GSK and holds stocks and shares in GSK. Conflict of interest: D.A. Lipson is an employee of GSK and holds stocks and shares in GSK. Conflict of interest: D.A. Lomas reports grant income, honoraria, and consultancy fees from GSK, and personal fees from Grifols, and chaired the GSK Respiratory Therapy Area Board 2012–2015. Conflict of interest: N. Martin is an employee of GSK and holds stocks and shares in GSK. Conflict of interest: F.J. Martinez reports personal fees and non-financial support from the American College of Chest Physicians, AstraZeneca, Boehringer Ingelheim, Continuing Education, ConCert, Genentech, GSK, Inova Fairfax Health System, Miller Communications, National Society for Continuing Education, Novartis, Pearl Pharmaceuticals, PeerView Communications, Prime Communications, Puerto Rico Respiratory Society, Chiesi, Roche, Sunovion, Theravance, Potomac, University of Alabama Birmingham, Physicians Education Resource, Canadian Respiratory Network and Teva, non-financial support from ProterrixBio, Gilead, Nitto and Zambon, and personal fees from Columbia University, Integritas, MD Magazine, Methodist Hospital Brooklyn, New York University, Unity, UpToDate, WedMD/MedScape, Western Connecticut Health Network, Academic CME, Patara, PlatformIQ, American Thoracic Society, Rockpointe and France Foundation, grant support from NIH, Rare Disease Health Communications and ProMedior, and is a member of steering committees for Afferent/Merck, Biogen, Veracyte, Prometic, Bayer and Bridge Biotherapeutics. Conflict of interest: R. Wise reports personal fees from AstraZeneca/MedImmune, Boehringer Ingelheim, ContraFect, Pulmonx, Roche, Spiration, Sunovion, Merck, Circassia, Pneuma, Verona, Bonti, Denali, Aradigm, Mylan/Theravance, Propeller Health, AbbVie and GSK, and grant support from AstraZeneca/MedImmune, Boehringer Ingelheim, Pearl Therapeutics, GSK and Sanofi-Aventis. Conflict of interest: D. Singh reports personal fees from GSK, AstraZeneca, Boehringer Ingelheim, Chiesi, Cipla, Genentech, Glenmark, Menarini, Mundipharma, Novartis, Peptinnovate, Pfizer, Pulmatrix, Theravance and Verona, and grant support from AstraZeneca, Boehringer Ingelheim, Chiesi, Glenmark, Menarini, Mundipharma, Novartis, Pfizer, Pulmatrix, Theravance and Verona.
- Published
- 2021
- Full Text
- View/download PDF
16. Quantitative 18 F-fluorodeoxyglucose positron emission tomography/computed tomography to assess pulmonary inflammation in COPD.
- Author
-
Vass L, Fisk M, Cheriyan J, Mohan D, Forman J, Oseni A, Devaraj A, Mäki-Petäjä KM, McEniery CM, Fuld J, Hopkinson NS, Lomas DA, Cockcroft JR, Tal-Singer R, Polkey MI, and Wilkinson IB
- Abstract
Rationale: COPD and smoking are characterised by pulmonary inflammation.
18 F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) imaging may improve knowledge of pulmonary inflammation in COPD patients and aid early development of novel therapies as an imaging biomarker., Objectives: To evaluate pulmonary inflammation, assessed by FDG uptake, in whole and regional lung in "usual" (smoking-related) COPD patients, alpha-1 antitrypsin deficiency (α1 ATD) COPD patients, smokers without COPD and never-smokers using FDG PET/CT. Secondly, to explore cross-sectional associations between FDG PET/CT and systemic inflammatory markers in COPD patients and repeatability of the technique in COPD patients., Methods: Data from two imaging studies were evaluated. Pulmonary FDG uptake (normalised Ki ; nKi ) was measured by Patlak graphical analysis in four subject groups: 84 COPD patients, 11 α1 ATD-COPD patients, 12 smokers and 10 never-smokers. Within the COPD group, associations between nKi and systemic markers of inflammation were assessed. Repeatability was evaluated in 32 COPD patients comparing nKi values at baseline and at 4-month follow-up., Results: COPD patients, α1 ATD-COPD patients and smokers had increased whole lung FDG uptake (nKi ) compared with never-smokers (0.0037±0.001, 0.0040±0.001, 0.0040±0.001 versus 0.0028±0.001 mL·cm-3 ·min-1 , respectively, p<0.05 for all). Similar results were observed in upper and middle lung regions. In COPD participants, plasma fibrinogen was associated with whole lung nKi (β=0.30, p=0.02) in multivariate analysis adjusted for current smoking, forced expiratory volume in 1 s % predicted, systemic neutrophils and C-reactive protein levels. Mean percentage difference in nKi between the baseline and follow-up was 3.2%, and the within subject coefficient of variability was 7.7%., Conclusions: FDG PET/CT has potential as a noninvasive tool to enable whole lung and regional quantification of FDG uptake to assess smoking- and COPD-related pulmonary inflammation., Competing Interests: Conflict of interest: L. Vass reports a PhD studentship grant from GlaxoSmithKline during the conduct of the study. Conflict of interest: M. Fisk has nothing to disclose. Conflict of interest: J. Cheriyan reports grants from GlaxoSmithKline (GSK) during the conduct of the study and as a Cambridge University Hospital National Health Service (NHS) Trust employee was obligated to spend 50% of his NHS time to do clinical trial work for GSK. However, he received no employee benefits from GSK. The payment for this time was made directly by GSK to Cambridge University Hospitals NHS Trust. Conflict of interest: D. Mohan is an employee of and shareholder in GSK. Conflict of interest: J. Forman has nothing to disclose. Conflict of interest: A. Oseni has nothing to disclose. Conflict of interest: A. Devaraj has nothing to disclose. Conflict of interest: K.M. Mäki-Petäjä has nothing to disclose. Conflict of interest: C.M. McEniery has nothing to disclose. Conflict of interest: J. Fuld has nothing to disclose. Conflict of interest: N.S. Hopkinson has nothing to disclose. Conflict of interest: D.A. Lomas reports grants, personal fees and nonfinancial support from GlaxoSmithKline, during the conduct of the study; and personal fees and grants from GlaxoSmithKline, and personal fees from Griffols, outside the submitted work. Conflict of interest: J.R. Cockcroft received a grant from GSK outside this submitted work. Conflict of interest: R. Tal-Singer was an employee and shareholder of GSK during the conduct of the study, and has received personal fees from Immunomet, Vocalis Health and Ena Respiratory. Conflict of interest: M.I. Polkey reports personal fees for an advisory board on this topic from GSK and grants from Innovate UK paid to his institution to conduct this work, during the conduct of the study. Conflict of interest: I.B. Wilkinson received a grant from Innovate UK and an educational imaging award from GSK during the conduct of the study., (Copyright ©The authors 2021.)- Published
- 2021
- Full Text
- View/download PDF
17. Limitations to Contingency Measures: Reflections from COVID-19 Surges in the UK.
- Author
-
Gordon C, Yardley S, Lomas DA, and Edwards SJL
- Subjects
- Humans, SARS-CoV-2, United Kingdom, COVID-19
- Published
- 2021
- Full Text
- View/download PDF
18. The development of highly potent and selective small molecule correctors of Z α 1 -antitrypsin misfolding.
- Author
-
Liddle J, Pearce AC, Arico-Muendel C, Belyanskaya S, Brewster A, Brown M, Chung CW, Denis A, Dodic N, Dossang A, Eddershaw P, Klimaszewska D, Haq I, Holmes DS, Jagger A, Jakhria T, Jigorel E, Lind K, Messer J, Neu M, Olszewski A, Ronzoni R, Rowedder J, Rüdiger M, Skinner S, Smith KJ, Trottet L, Uings I, Zhu Z, Irving JA, and Lomas DA
- Subjects
- Crystallization, Drug Development methods, Drug Evaluation, Preclinical, Endoplasmic Reticulum metabolism, Gene Library, Hepatocytes metabolism, Humans, Models, Molecular, Protein Conformation, alpha 1-Antitrypsin genetics, Drug Design, Protein Folding, alpha 1-Antitrypsin metabolism
- Abstract
α1-antitrypsin deficiency is characterised by the misfolding and intracellular polymerisation of mutant α1-antitrypsin protein within the endoplasmic reticulum (ER) of hepatocytes. Small molecules that bind and stabilise Z α
1 -antitrypsin were identified via a DNA-encoded library screen. A subsequent structure based optimisation led to a series of highly potent, selective and cellular active α1-antitrypsin correctors., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
19. Triple Versus Dual Combination Therapy in Chronic Obstructive Pulmonary Disease in Asian Countries: Analysis of the IMPACT Trial.
- Author
-
Halpin DMG, Criner GJ, Dransfield MT, Han MK, Hartley B, Harvey C, Jones CE, Kato M, Lange P, Lettis S, Lomas DA, Martinez FJ, Martin N, Singh D, Wise R, Zheng J, and Lipson DA
- Abstract
Introduction: In the IMPACT trial, single-inhaler triple therapy fluticasone furoate/umeclidinium/vilanterol (FF/UMEC/VI) reduced moderate/severe exacerbation rates versus FF/VI or UMEC/VI dual therapy in patients with chronic obstructive pulmonary disease (COPD); however, pneumonia incidence was higher in FF-containing arms. As COPD is a growing problem in Asia, we compared the efficacy and safety of FF/UMEC/VI in Asia versus non-Asia regions., Methods: IMPACT was a double-blind, 52-week trial in symptomatic COPD patients with ≥ 1 moderate/severe exacerbation in the prior year. This pre-specified analysis evaluated the annual rate of moderate/severe exacerbations, change from baseline in trough forced expiratory volume in 1 s, and St George's Respiratory Questionnaire total score, mortality, and safety (including pneumonia) in Asia versus non-Asia regions., Results: The intent-to-treat population comprised 10,355 patients (Asia n = 1644 [16%]). Rate ratios (95% confidence intervals) for moderate/severe exacerbations with FF/UMEC/VI were 0.89 (0.76-1.05) versus FF/VI and 0.86 (0.71-1.04) versus UMEC/VI in Asia, and 0.84 (0.79-0.90) and 0.74 (0.68-0.80) in non-Asia. Efficacy of FF/UMEC/VI on other endpoints was similar in both regions. There was an increased incidence of investigator-reported pneumonia in patients in Asia (FF/UMEC/VI: 13%; FF/VI: 14%; UMEC/VI: 6%) compared with non-Asia (FF/UMEC/VI: 6%; FF/VI: 5%; UMEC/VI: 4%). The increased risk of pneumonia in patients in Asia was most marked in patients with lower body mass index, lower lung function, and taking inhaled corticosteroids. In post hoc analysis of adjudicated on-treatment all-cause mortality, probabilities of death were numerically lower in both regions with FF/UMEC/VI (Asia: 1.16%; non-Asia: 1.35%) and FF/VI (Asia: 1.77%; non-Asia: 1.21%) versus UMEC/VI (Asia: 1.91%; non-Asia: 2.23%)., Conclusions: FF/UMEC/VI provides similar benefits in COPD patients in Asia and non-Asia regions. Clinical benefits of treatment, including reduction in mortality risk, should be weighed against risk of pneumonia, taking account of all known risk factors., Trial Registration: ClinicalTrials.gov identification, NCT02164513.
- Published
- 2021
- Full Text
- View/download PDF
20. The Importance of N186 in the Alpha-1-Antitrypsin Shutter Region Is Revealed by the Novel Bologna Deficiency Variant.
- Author
-
Ronzoni R, Ferrarotti I, D'Acunto E, Balderacchi AM, Ottaviani S, Lomas DA, Irving JA, Miranda E, and Fra A
- Subjects
- Amino Acid Substitution, HEK293 Cells, Humans, Protein Domains, alpha 1-Antitrypsin Deficiency genetics, alpha 1-Antitrypsin Deficiency pathology, Mutation, Missense, alpha 1-Antitrypsin biosynthesis, alpha 1-Antitrypsin chemistry, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin Deficiency metabolism
- Abstract
Alpha-1-antitrypsin (AAT) deficiency causes pulmonary disease due to decreased levels of circulating AAT and consequently unbalanced protease activity in the lungs. Deposition of specific AAT variants, such as the common Z AAT, within hepatocytes may also result in liver disease. These deposits are comprised of ordered polymers of AAT formed by an inter-molecular domain swap. The discovery and characterization of rare variants of AAT and other serpins have historically played a crucial role in the dissection of the structural mechanisms leading to AAT polymer formation. Here, we report a severely deficient shutter region variant, Bologna AAT (N186Y), which was identified in five unrelated subjects with different geographical origins. We characterized the new variant by expression in cellular models in comparison with known polymerogenic AAT variants. Bologna AAT showed secretion deficiency and intracellular accumulation as detergent-insoluble polymers. Extracellular polymers were detected in both the culture media of cells expressing Bologna AAT and in the plasma of a patient homozygous for this variant. Structural modelling revealed that the mutation disrupts the hydrogen bonding network in the AAT shutter region. These data support a crucial coordinating role for asparagine 186 and the importance of this network in promoting formation of the native structure.
- Published
- 2021
- Full Text
- View/download PDF
21. Scaling Concepts in Serpin Polymer Physics.
- Author
-
Raccosta S, Librizzi F, Jagger AM, Noto R, Martorana V, Lomas DA, Irving JA, and Manno M
- Abstract
α1-Antitrypsin is a protease inhibitor belonging to the serpin family. Serpin polymerisation is at the core of a class of genetic conformational diseases called serpinopathies. These polymers are known to be unbranched, flexible, and heterogeneous in size with a beads-on-a-string appearance viewed by negative stain electron microscopy. Here, we use atomic force microscopy and time-lapse dynamic light scattering to measure polymer size and shape for wild-type (M) and Glu342→Lys (Z) α1-antitrypsin, the most common variant that leads to severe pathological deficiency. Our data for small polymers deposited onto mica and in solution reveal a power law relation between the polymer size, namely the end-to-end distance or the hydrodynamic radius, and the polymer mass, proportional to the contour length. We use the scaling concepts of polymer physics to assess that α1-antitrypsin polymers are random linear chains with a low persistence length.
- Published
- 2021
- Full Text
- View/download PDF
22. Risk of Exacerbation and Pneumonia with Single-Inhaler Triple versus Dual Therapy in IMPACT.
- Author
-
Dransfield MT, Crim C, Criner GJ, Day NC, Halpin DMG, Han MK, Jones CE, Kilbride S, LaFon D, Lipson DA, Lomas DA, Martin N, Martinez FJ, Singh D, Wise RA, and Lange P
- Subjects
- Administration, Inhalation, Androstadienes therapeutic use, Benzyl Alcohols therapeutic use, Bronchodilator Agents therapeutic use, Chlorobenzenes therapeutic use, Double-Blind Method, Humans, Nebulizers and Vaporizers, Treatment Outcome, Pneumonia drug therapy, Pneumonia epidemiology, Pulmonary Disease, Chronic Obstructive drug therapy
- Abstract
Rationale: In the IMPACT (Informing the Pathway of COPD Treatment) trial, single-inhaler fluticasone furoate/umeclidinium/vilanterol (FF/UMEC/VI) triple therapy reduced exacerbation risk versus FF/VI and UMEC/VI and mortality risk versus UMEC/VI. However, pneumonia incidence was higher in the inhaled corticosteroid (FF)-containing arms, raising questions about the relative benefit of exacerbation reduction compared with the increased risk of pneumonia. Objectives: Determine benefit-risk of the three treatments by evaluating time-to-first and rates of composite exacerbation or pneumonia outcomes. Methods: We evaluated time-to-first (prespecified) and rates ( post hoc ) of investigator-reported pneumonia, serious pneumonia leading to hospitalization or death, and the composite endpoints of 1 ) moderate (required antibiotics/corticosteroids)/severe (hospitalized) exacerbation or pneumonia and 2 ) severe exacerbation or serious (hospitalized) pneumonia. Analyses were repeated for radiographically confirmed pneumonia ( post hoc ). Results: Moderate/severe exacerbations occurred in 47%, 49%, and 50% of patients randomized to FF/UMEC/VI, FF/VI and UMEC/VI, and pneumonias in 8%, 7%, and 5%, respectively. FF/UMEC/VI reduced the risk of combined moderate/severe exacerbation or pneumonia (time-to-first) versus FF/VI (hazard ratio, 0.87 [95% confidence interval (CI), 0.82-0.92]) and UMEC/VI (0.87 [0.81-0.94]), as well as the risk of combined severe exacerbation or serious pneumonia versus UMEC/VI (0.83 [0.72-0.96]). FF/UMEC/VI reduced the rate of combined moderate/severe exacerbation or pneumonia (rate ratio, 0.78 [0.72-0.84]) and combined severe exacerbation or serious pneumonia (rate ratio, 0.76 [0.65-0.89]) versus UMEC/VI. Results were similar for radiographically confirmed pneumonia endpoints. Conclusions: Despite higher incidence of pneumonia in FF-containing arms, these composite exacerbation/pneumonia outcomes support a favorable benefit-risk profile of FF/UMEC/VI versus FF/VI and UMEC/VI in patients with symptomatic chronic obstructive pulmonary disease and a history of exacerbations.
- Published
- 2021
- Full Text
- View/download PDF
23. Lessons and risks of medical device deployment in a global pandemic.
- Author
-
Shipley RJ, Brealey D, Haniffa R, Elwell C, Baker T, Lomas DA, and Singer M
- Subjects
- COVID-19 epidemiology, Global Health legislation & jurisprudence, Humans, International Cooperation, Manufacturing Industry economics, Manufacturing Industry legislation & jurisprudence, Policy, Respiration, Artificial instrumentation, COVID-19 therapy, Delivery of Health Care economics, Global Health economics, Pandemics prevention & control, Ventilators, Mechanical economics
- Published
- 2021
- Full Text
- View/download PDF
24. The molecular species responsible for α 1 -antitrypsin deficiency are suppressed by a small molecule chaperone.
- Author
-
Ronzoni R, Heyer-Chauhan N, Fra A, Pearce AC, Rüdiger M, Miranda E, Irving JA, and Lomas DA
- Subjects
- Antibodies, Monoclonal pharmacology, Hepatocytes drug effects, Humans, Molecular Chaperones antagonists & inhibitors, Molecular Chaperones chemistry, Molecular Chaperones ultrastructure, Polymers chemistry, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, alpha 1-Antitrypsin chemistry, alpha 1-Antitrypsin drug effects, alpha 1-Antitrypsin ultrastructure, alpha 1-Antitrypsin Deficiency genetics, Molecular Chaperones genetics, Protein Conformation drug effects, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin Deficiency drug therapy
- Abstract
The formation of ordered Z (Glu342Lys) α
1 -antitrypsin polymers in hepatocytes is central to liver disease in α1 -antitrypsin deficiency. In vitro experiments have identified an intermediate conformational state (M*) that precedes polymer formation, but this has yet to be identified in vivo. Moreover, the mechanism of polymer formation and their fate in cells have been incompletely characterised. We have used cell models of disease in conjunction with conformation-selective monoclonal antibodies and a small molecule inhibitor of polymerisation to define the dynamics of polymer formation, accumulation and secretion. Pulse-chase experiments demonstrate that Z α1 -antitrypsin accumulates as short-chain polymers that partition with soluble cellular components and are partially secreted by cells. These precede the formation of larger, insoluble polymers with a longer half-life (10.9 ± 1.7 h and 20.9 ± 7.4 h for soluble and insoluble polymers, respectively). The M* intermediate (or a by-product thereof) was identified in the cells by a conformation-specific monoclonal antibody. This was completely abrogated by treatment with the small molecule, which also blocked the formation of intracellular polymers. These data allow us to conclude that the M* conformation is central to polymerisation of Z α1 -antitrypsin in vivo; preventing its accumulation represents a tractable approach for pharmacological treatment of this condition; polymers are partially secreted; and polymers exist as two distinct populations in cells whose different dynamics have likely consequences for the aetiology of the disease., (© 2020 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2021
- Full Text
- View/download PDF
25. Markers of disease activity in COPD: an 8-year mortality study in the ECLIPSE cohort.
- Author
-
Celli B, Locantore N, Yates JC, Bakke P, Calverley PMA, Crim C, Coxson HO, Lomas DA, MacNee W, Miller BE, Mullerova H, Rennard SI, Silverman EK, Wouters E, Tal-Singer R, Agusti A, and Vestbo J
- Subjects
- Biomarkers, Cross-Sectional Studies, Forced Expiratory Volume, Humans, Quality of Life, Respiratory Function Tests, Severity of Illness Index, Surveys and Questionnaires, Pulmonary Disease, Chronic Obstructive
- Abstract
Rationale: There are no validated measures of disease activity in COPD. Since "active" disease is expected to have worse outcomes ( e.g. mortality), we explored potential markers of disease activity in patients enrolled in the ECLIPSE cohort in relation to 8-year all-cause mortality., Methods: We investigated 1) how changes in relevant clinical variables over time (1 or 3 years) relate to 8-year mortality; 2) whether these variables inter-relate; and 3) if any clinical, imaging and/or biological marker measured cross-sectionally at baseline relates to any activity component., Results: Results showed that 1) after 1 year, hospitalisation for COPD, exacerbation frequency, worsening of body mass index, airflow obstruction, dyspnoea and exercise (BODE) index or health status (St George's Respiratory Questionnaire (SGRQ)) and persistence of systemic inflammation were significantly associated with 8-year mortality; 2) at 3 years, the same markers, plus forced expiratory volume in 1 s (FEV
1 ) decline and to a lesser degree computed tomography (CT) emphysema, showed association, thus qualifying as markers of disease activity; 3) changes in FEV1 , inflammatory cytokines and CT emphysema were not inter-related, while the multidimensional indices (BODE and SGRQ) showed modest correlations; and 4) changes in these markers could not be predicted by any baseline cross-sectional measure., Conclusions: In COPD, 1- and 3-year changes in exacerbation frequency, systemic inflammation, BODE and SGRQ scores and FEV1 decline are independent markers of disease activity associated with 8-year all-cause mortality. These disease activity markers are generally independent and not predictable from baseline measurements., Competing Interests: Conflict of interest: B. Celli reports grants and personal fees for scientific committee work from GlaxoSmithKline, during the conduct of the study; grants and provision of research facilities from AstraZeneca, personal fees for consultancy and scientific committee work from GlaxoSmithKline, personal fees for consultancy from Boehringer Ingelheim, Sanofi-Aventis, Menarini, Chiesi and Pulmonx, outside the submitted work. Conflict of interest: N. Locantore is an employee and shareholder of GSK. Conflict of interest: J.C. Yates is an employee of and owns shares in GSK. Conflict of interest: P. Bakke reports personal fees for advisory board work and lectures from GlaxoSmithKline, AstraZeneca and Boehringer Ingelheim, personal fees for advisory board work from Chiesi, outside the submitted work. Conflict of interest: P.M.A. Calverley reports personal fees from GSK, Boehringer Ingelheim, Novartis, Zambon, Respironics and Recipharm, outside the submitted work. Conflict of interest: C. Crim is an employee and shareholder of GlaxoSmithKline. Conflict of interest: H.O. Coxson reports grants and personal fees for scientific committee work from GSK, during the conduct of the study. Conflict of interest: D.A. Lomas reports grants and personal fees from GlaxoSmithKline, during the conduct of the study; grants and personal fees for advisory board work and lectures from GlaxoSmithKline, personal fees for advisory board work from Griffols, outside the submitted work. Conflict of interest: W. MacNee reports personal fees for scientific committee work from GSK, during the conduct of the study; personal fees from GSK and AstraZeneca, grants and personal fees from Pfizer, outside the submitted work. Conflict of interest: B.E. Miller is an employee and shareholder of GSK. Conflict of interest: H. Mullerova is a former employee of GlaxoSmithKline. Conflict of interest: S.I. Rennard is a former employee of AstraZeneca, has provided consultancy to GSK, Verona, Bergenbio and NovoVentures, and currently holds shares in AstraZeneca, outside of the submitted work. Prior to 2007, S.I. Rennard received funding from the tobacco industry for studies relating to harm reduction and to the impact of tobacco smoke on stem cells, and also consulted with RJ Reynolds (without personal fee) on the topic of harm reduction: funding from RJ Reynolds to evaluate the effect of a harm reduction product in normal smokers (1996) and in subjects with chronic bronchitis (1999) and to assess the effect of smoking cessation on lower respiratory tract inflammation (2000); participation in a Philip Morris multicentre study to assess biomarkers of smoke exposure (2002); funding for a clinical trial from the Institute for Science and Health (2005), which receives support from the tobacco industry, to evaluate biomarkers in exhaled breath associated with smoking cessation and reduction (this study was supplemented with funding from Lorillard and RJ Reynolds); grants from the Philip Morris External Research Program (2005) to assess the impact of cigarette smoking on circulating stem cells in the mouse; consultancy for RJ Reynolds on the topic of harm reduction until 2007 (no personal remuneration). There are no active tobacco-industry funded projects. All ties with tobacco industry companies and entities supported by tobacco companies were terminated in 2007. Conflict of interest: E.K. Silverman reports grants, personal fees and travel expenses from GlaxoSmithKline, grants from NIH, during the conduct of the study. Conflict of interest: E. Wouters reports personal fees for advisory board work from Nycomed and Boehringer Ingelheim BV, grants and personal fees for lectures from AstraZeneca and GSK, personal fees for lectures from Novartis and Chiesi, outside the submitted work. Conflict of interest: R. Tal-Singer reports is an employee and shareholder of GlaxoSmithKline. Conflict of interest: A. Agusti reports personal fees for scientific committee work from GSK, during the conduct of the study; personal fees from AstraZeneca, Chiesi and Nuvaira, grants and personal fees from Menarini and GSK, outside the submitted work. Conflict of interest: J. Vestbo reports personal fees for steering committee work from GSK, during the conduct of the study; personal fees for lectures and consultancy from AstraZeneca, Chiesi and Novartis, grants and personal fees for lectures and consultancy from Boehringer Ingelheim, grants and personal fees for consultancy from GSK, outside the submitted work; and has a family member employed by Chiesi (Denmark)., (Copyright ©ERS 2021.)- Published
- 2021
- Full Text
- View/download PDF
26. Conversion of the death inhibitor ARC to a killer activates pancreatic β cell death in diabetes.
- Author
-
McKimpson WM, Chen Y, Irving JA, Zheng M, Weinberger J, Tan WLW, Tiang Z, Jagger AM, Chua SC Jr, Pessin JE, Foo RS, Lomas DA, and Kitsis RN
- Subjects
- Animals, Apoptosis Regulatory Proteins physiology, Cytoplasm metabolism, Cytoskeletal Proteins genetics, Diabetes Mellitus, Experimental etiology, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Type 2 etiology, Diabetes Mellitus, Type 2 metabolism, Female, Humans, Insulin-Secreting Cells metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle Proteins physiology, Nerve Tissue Proteins genetics, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin metabolism, Apoptosis, Cell Nucleus metabolism, Cytoskeletal Proteins metabolism, Diabetes Mellitus, Experimental pathology, Diabetes Mellitus, Type 2 pathology, Insulin-Secreting Cells pathology, Nerve Tissue Proteins metabolism, alpha 1-Antitrypsin chemistry
- Abstract
Loss of insulin-secreting pancreatic β cells through apoptosis contributes to the progression of type 2 diabetes, but underlying mechanisms remain elusive. Here, we identify a pathway in which the cell death inhibitor ARC paradoxically becomes a killer during diabetes. While cytoplasmic ARC maintains β cell viability and pancreatic architecture, a pool of ARC relocates to the nucleus to induce β cell apoptosis in humans with diabetes and several pathophysiologically distinct mouse models. β cell death results through the coordinate downregulation of serpins (serine protease inhibitors) not previously known to be synthesized and secreted by β cells. Loss of the serpin α
1 -antitrypsin from the extracellular space unleashes elastase, triggering the disruption of β cell anchorage and subsequent cell death. Administration of α1 -antitrypsin to mice with diabetes prevents β cell death and metabolic abnormalities. These data uncover a pathway for β cell loss in type 2 diabetes and identify an FDA-approved drug that may impede progression of this syndrome., Competing Interests: Declaration of interests David Lomas is an inventor on the patent PCT/GB2019/051761 for a small molecule treatment of antitrypsin deficiency. This small molecule has not been used in this work., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
27. Prognostic value of clinically important deterioration in COPD: IMPACT trial analysis.
- Author
-
Han MK, Criner GJ, Dransfield MT, Halpin DMG, Jones CE, Kilbride S, Lange P, Lettis S, Lipson DA, Lomas DA, Martin N, Martinez FJ, Wise RA, Naya IP, and Singh D
- Abstract
Introduction: Clinically important deterioration (CID) is a multicomponent measure for assessing disease worsening in chronic obstructive pulmonary disease (COPD). This analysis investigated the prognostic value of a CID event on future clinical outcomes and the effect of single-inhaler triple versus dual therapy on reducing CID risk in patients in the IMPACT trial., Methods: IMPACT was a phase III, double-blind, 52-week, multicentre trial. Patients with symptomatic COPD and at least one moderate/severe exacerbation in the prior year were randomised 2:2:1 to fluticasone furoate/umeclidinium/vilanterol (FF/UMEC/VI) 100/62.5/25 µg, FF/VI 100/25 µg or UMEC/VI 62.5/25 µg. CID at the time-point of interest was defined as a moderate/severe exacerbation, ≥100 mL decrease in trough forced expiratory volume in 1 s or deterioration in health status (increase of ≥4.0 units in St George's Respiratory Questionnaire total score or increase of ≥2.0 units in COPD Assessment Test score) from baseline. A treatment-independent post hoc prognostic analysis compared clinical outcomes up to week 52 in patients with/without a CID by week 28. A prospective analysis evaluated time to first CID with each treatment., Results: Patients with a CID by week 28 had significantly increased exacerbation rates after week 28, smaller improvements in lung function and health status at week 52 (all p<0.001), and increased risk of all-cause mortality after week 28 versus patients who were CID-free. FF/UMEC/VI significantly reduced CID risk versus dual therapies (all p<0.001)., Conclusions: Prevention of short-term disease worsening was associated with better long-term clinical outcomes. FF/UMEC/VI reduced CID risk versus dual therapies; this effect may improve long-term prognosis in this population., Competing Interests: Conflict of interest: M.K. Han personal fees from AstraZeneca, Boehringer Ingelheim and GlaxoSmithKline, research support from Novartis and Sunovion, and personal fees from Mylan, Merck and Verona, outside the submitted work. Conflict of interest: G.J. Criner personal fees from Almirall, AstraZeneca, Boehringer Ingelheim, Chiesi, CSA Medical, Eolo Medical and GlaxoSmithKline, and ownership interest in HGE Health Care Solutions, and personal fees from Novartis, Nuvaira, Olympus, Pulmonx, Verona, Amgen, Broncus Medical, Gala Therapeutics, Helios Medical, Merck, Medtronic, Mereo BioPharma, NGM Biopharmaceuticals, Philips Respironics, Respivant Sciences and The Implementation Group, outside the submitted work. Conflict of interest: M.T. Dransfield grants from the Dept of Defense, personal fees and other support from Boehringer Ingelheim and GlaxoSmithKline, other support from Novartis, personal fees and other support from AstraZeneca, other support from Yungjin, personal fees and other support from PneumRx/BTG, other support from Pulmonx, personal fees from Genentech, other support from Boston Scientific, personal fees from Quark Pharmaceuticals, grants from NIH, personal fees from Mereo, and grants from the American Lung Association and the NIH, outside the submitted work. Conflict of interest: D.M.G. Halpin reports personal fees from AstraZeneca, personal fees and nonfinancial support from Boehringer Ingelheim, personal fees from Chiesi, personal fees and nonfinancial support from GlaxoSmithKline, and personal fees from Novartis, Pfizer and Sanofi, outside the submitted work. Conflict of interest: C.E. Jones is an employee of and holds shares/options in GlaxoSmithKline, outside the submitted work. Conflict of interest: S. Kilbride is an employee of and holds shares/options in GlaxoSmithKline, outside the submitted work. Conflict of interest: P. Lange personal fees from AstraZeneca, Boehringer Ingelheim and Chiesi, and grants and personal fees from GlaxoSmithKline, outside the submitted work. Conflict of interest: S. Lettis is an employee of and holds shares/options in GlaxoSmithKline outside the submitted work. Conflict of interest: D.A. Lipson is an employee of and holds shares/options in GlaxoSmithKline outside the submitted work. Conflict of interest: D.A. Lomas reports grants from GlaxoSmithKline and personal fees from Grifols, outside the submitted work. Conflict of interest: N. Martin is an employee of and holds shares/options in GlaxoSmithKline, outside the submitted work. Conflict of interest: F.J. Martinez reports personal fees, nonfinancial support and other support from AstraZeneca and Boehringer Ingelheim, nonfinancial support and other support from ProterrixBio, personal fees from Columbia University, personal fees and nonfinancial support from Genentech, GlaxoSmithKline and Inova Fairfax Health System, personal fees from MD Magazine and Methodist Hospital Brooklyn, personal fees and nonfinancial support from Miller Communications, the National Association for Continuing Education and Novartis, personal fees from New York University, personal fees and nonfinancial support from Pearl Pharmaceuticals, PeerView Communications, Prime Communications, the Puerto Rican Respiratory Society, Chiesi, Sunovion and Theravance, personal fees from UpToDate and WebMD/MedScape, other support from Afferent/Merck, nonfinancial support from Gilead and Nitto, personal fees and other support from Patara/Respivant, personal fees and nonfinancial support from Potomac, other support from Biogen, personal fees and nonfinancial support from the University of Alabama Birmingham, other support from Veracyte, nonfinancial support from Zambon, personal fees from the American Thoracic Society, grants from the NIH, personal fees and nonfinancial support from the Physicians Education Resource, personal fees from Rockpointe, other support from Prometic, personal fees from Rare Disease Healthcare Communications, other support from Bayer and Bridge Biotherapeutics, personal fees and nonfinancial support from the Canadian Respiratory Network, other support from ProMedior, personal fees and nonfinancial support from Teva, personal fees from the France Foundation, personal fees and nonfinancial support from Dartmouth, other support from Gala, and personal fees from the Physicians Education Resource, outside the submitted work. Conflict of interest: R.A. Wise reports grants and personal fees from AstraZeneca/Medimmune/Pearl and Boehringer Ingelheim, personal fees from Contrafect, Pulmonx, Roche, Spiration and Sunovion, grants from Pearl Therapeutics, personal fees from Merck, Circassia, Pneuma, Verona, Mylan/Theravance and Propeller Health, grants from Sanofi-Aventis, personal fees from AbbVie, and grants and personal fees from GSK, outside the submitted work. Conflict of interest: I.P. Naya holds shares/options in GlaxoSmithKline, outside the submitted work. Conflict of interest: D. Singh reports grants and personal fees from AstraZeneca, Boehringer Ingelheim and Chiesi, personal fees from Cipla and Genentech, grants and personal fees from Glenmark, personal fees from GlaxoSmithKline, grants and personal fees from Menarini and Mundipharma, personal fees from Peptinnovate, and grants and personal fees from Pfizer, Pulmatrix, Theravance and Verona, outside the submitted work., (Copyright ©ERS 2021.)
- Published
- 2021
- Full Text
- View/download PDF
28. Development of a small molecule that corrects misfolding and increases secretion of Z α 1 -antitrypsin.
- Author
-
Lomas DA, Irving JA, Arico-Muendel C, Belyanskaya S, Brewster A, Brown M, Chung CW, Dave H, Denis A, Dodic N, Dossang A, Eddershaw P, Klimaszewska D, Haq I, Holmes DS, Hutchinson JP, Jagger AM, Jakhria T, Jigorel E, Liddle J, Lind K, Marciniak SJ, Messer J, Neu M, Olszewski A, Ordonez A, Ronzoni R, Rowedder J, Rüdiger M, Skinner S, Smith KJ, Terry R, Trottet L, Uings I, Wilson S, Zhu Z, and Pearce AC
- Subjects
- Animals, Endoplasmic Reticulum, Hepatocytes, Mice, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin Deficiency
- Abstract
Severe α
1 -antitrypsin deficiency results from the Z allele (Glu342Lys) that causes the accumulation of homopolymers of mutant α1 -antitrypsin within the endoplasmic reticulum of hepatocytes in association with liver disease. We have used a DNA-encoded chemical library to undertake a high-throughput screen to identify small molecules that bind to, and stabilise Z α1 -antitrypsin. The lead compound blocks Z α1 -antitrypsin polymerisation in vitro, reduces intracellular polymerisation and increases the secretion of Z α1 -antitrypsin threefold in an iPSC model of disease. Crystallographic and biophysical analyses demonstrate that GSK716 and related molecules bind to a cryptic binding pocket, negate the local effects of the Z mutation and stabilise the bound state against progression along the polymerisation pathway. Oral dosing of transgenic mice at 100 mg/kg three times a day for 20 days increased the secretion of Z α1 -antitrypsin into the plasma by sevenfold. There was no observable clearance of hepatic inclusions with respect to controls over the same time period. This study provides proof of principle that "mutation ameliorating" small molecules can block the aberrant polymerisation that underlies Z α1 -antitrypsin deficiency., (© 2021 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2021
- Full Text
- View/download PDF
29. Effect of Age on the Efficacy and Safety of Once-Daily Single-Inhaler Triple-Therapy Fluticasone Furoate/Umeclidinium/Vilanterol in Patients With COPD: A Post Hoc Analysis of the Informing the Pathway of COPD Treatment Trial.
- Author
-
Hanania NA, Mannino DM, Criner GJ, Dransfield MT, Han MK, Jones CE, Kilbride S, Lomas DA, Martin N, Martinez FJ, Singh D, Wise RA, Halpin DMG, Lima R, and Lipson DA
- Subjects
- Administration, Inhalation, Aged, Androstadienes administration & dosage, Androstadienes adverse effects, Bronchodilator Agents administration & dosage, Bronchodilator Agents adverse effects, Double-Blind Method, Drug Combinations, Drug Monitoring methods, Drug Monitoring statistics & numerical data, Female, Humans, Male, Nebulizers and Vaporizers, Respiratory System Agents administration & dosage, Respiratory System Agents adverse effects, Benzyl Alcohols administration & dosage, Benzyl Alcohols adverse effects, Chlorobenzenes administration & dosage, Chlorobenzenes adverse effects, Health Status Disparities, Pulmonary Disease, Chronic Obstructive diagnosis, Pulmonary Disease, Chronic Obstructive drug therapy, Pulmonary Disease, Chronic Obstructive physiopathology, Quinuclidines administration & dosage, Quinuclidines adverse effects, Respiratory Function Tests methods, Symptom Flare Up
- Abstract
Background: In the Informing the Pathway of COPD Treatment (IMPACT) trial, single-inhaler triple-therapy fluticasone furoate (FF), umeclidinium (UMEC), and vilanterol (VI) reduced moderate/severe exacerbation rates vs FF/VI and UMEC/VI in patients with symptomatic COPD and a history of exacerbations, with a similar safety profile., Research Question: Are trial outcomes with single-inhaler triple-therapy FF/UMEC/VI vs FF/VI and UMEC/VI affected by age in patients with symptomatic COPD and a history of exacerbations?, Study Design and Methods: IMPACT was a phase III, double-blind, 52-week trial. Patients ≥ 40 years of age with symptomatic COPD and ≥ 1 moderate/severe exacerbation in the previous year were randomly assigned 2:2:1 to FF/UMEC/VI 100/62.5/25 μg, FF/VI 100/25 μg, or UMEC/VI 62.5/25 μg. End points assessed by age included annual rate of moderate/severe exacerbations, change from baseline (CFB) in trough FEV
1 , proportion of St. George's Respiratory Questionnaire (SGRQ) responders (≥ 4 units decrease from baseline in SGRQ total score), and safety., Results: The intention-to-treat population comprised 10,355 patients; 4,724 (46%), 4,225 (41%), and 1,406 (14%) were ≤ 64, 65 to 74, and ≥ 75 years of age, respectively. FF/UMEC/VI reduced on-treatment moderate/severe exacerbation rates vs FF/VI (% reduction [95% CI]: ≤ 64 years, 8% [-1 to 16]; P = .070; 65-74 years, 22% [14-29]; P < .001; ≥ 75 years, 18% [3-31]; P = .021) and vs UMEC/VI (≤ 64 years, 16% [7-25]; P = .002; 65-74 years, 33% [25-41]; P < .001; ≥ 75 years, 24% [6-38]; P = .012), with greatest rate reduction seen in the 65 to 74 and ≥ 75 years subgroups. Post hoc analyses of CFB in trough FEV1 and proportion of SGRQ responders at week 52 were significantly greater with FF/UMEC/VI than with FF/VI or UMEC/VI in all subgroups. No new safety signals were identified., Interpretation: FF/UMEC/VI reduced the rate of moderate/severe exacerbations and improved lung function and health status vs FF/VI and UMEC/VI irrespective of age for most end points, with a similar safety profile., Clinical Trial Registration: ClinicalTrials.gov; No.: NCT02164513; URL: www.clinicaltrials.govCTT116855., (Copyright © 2020. Published by Elsevier Inc.)- Published
- 2021
- Full Text
- View/download PDF
30. InforMing the PAthway of COPD Treatment (IMPACT Trial) Single-Inhaler Triple Therapy (Fluticasone Furoate/Umeclidinium/Vilanterol) Versus Fluticasone Furoate/Vilanterol and Umeclidinium/Vilanterol in Patients With COPD: Analysis of the Western Europe and North America Regions.
- Author
-
Bourdin A, Criner G, Devouassoux G, Dransfield M, Halpin DMG, Han MK, Jones CE, Kalhan R, Lange P, Lettis S, Lipson DA, Lomas DA, Echave-Sustaeta María-Tomé JM, Martin N, Martinez FJ, Quasny H, Sail L, Siler TM, Singh D, Thomashow B, Watz H, Wise R, and Hanania NA
- Abstract
Background: The InforMing the Pathway of COPD Treatment (IMPACT) trial demonstrated lower moderate/severe exacerbation rates with fluticasone furoate/umeclidinium/vilanterol (FF/UMEC/VI) versus FF/VI or UMEC/VI in patients with chronic obstructive pulmonary disease (COPD) and a history of exacerbations. Since IMPACT was a global study, post-hoc analyses were conducted by geographic region to investigate potential differences in overall findings., Methods: IMPACT was a 52-week, randomized, double-blind trial. Patients with symptomatic COPD and ≥1 moderate/severe exacerbation in the prior year were randomized 2:2:1 to once-daily FF/UMEC/VI 100/62.5/25µg, FF/VI 100/25µg, or UMEC/VI 62.5/25µg. Endpoints assessed in the overall, Western Europe and North America populations included on-treatment moderate/severe exacerbation (rates and time-to-first), trough forced expiratory volume in 1 second and St George's Respiratory Questionnaire (SGRQ) total score. Safety was assessed., Results: Overall, 10,355 patients were enrolled, 3164 from Western Europe, 2639 from North America. FF/UMEC/VI significantly reduced on-treatment moderate/severe exacerbation rates versus FF/VI and UMEC/VI in Western Europe (rate ratios 0.82 [95%CI 0.74-0.91], P <.001 and 0.76 [0.67-0.87], P <.001) and in North America (0.87 [0.77-0.97], P =.014 and 0.69 [0.60-0.80], P <.001). FF/UMEC/VI reduced time-to-first moderate/severe exacerbation and improved lung function versus FF/VI and UMEC/VI in both regions, and improved SGRQ total score in Western Europe, but not North America. Safety profiles were generally similar between treatment groups/regions; the inhaled corticosteroid class effect of increased pneumonia incidence was seen in North America but not Western Europe., Conclusion: Consistent with intent-to-treat results, FF/UMEC/VI reduced moderate/severe exacerbation rate and risk and improved lung function in Western Europe and North America; however, between-regions differences were seen for SGRQ total score and pneumonia incidence., Clinical Trial Registration: NCT02164513., Competing Interests: Editorial support (in the form of writing assistance, assembling figures, collating author comments, grammatical editing and referencing) was provided by Chrystelle Rasamison, Fishawack Indicia Ltd, United Kingdom, and was funded by GSK. A Bourdin was an investigator in the IMPACT trial and has received personal fees and a grant from Boehringer Ingelheim, personal fees from AstraZeneca, Chiesi, GSK, Novartis and Sanofi-Regeneron, and is an investigator in clinical trials from Nuvaira, Pulmonx, Actelion, MSD, United Therapeutic, Vertex, Acceleron, and Galapagos. G Criner has received personal fees from Almirall, AstraZeneca, Boehringer Ingelheim, Chiesi, CSA Medical, Eolo, GSK, HGE Technologies, Novartis, Nuvaira, Olympus, Pulmonx, and Verona. G Devouassoux has received personal fees from AstraZeneca, Novartis Pharma, GSK, Chiesi, and Boehringer Ingelheim, and contracted clinical trial support from AstraZeneca, GSK, Novartis Pharma, ALK, Chiesi, and Boehringer Ingelheim. M Dransfield has received personal fees from Boehringer Ingelheim, PneumRx/BTG, Genentech, Quark Pharmaceuticals, Mereo, AstraZeneca and GSK, a grant from the Department of Defense, and contracted clinical trial support from PneumRx/BTG, Novartis, AstraZeneca, Yungjin, Pulmonx, Boston Scientific, Boehringer Ingelheim, and GSK. DMG Halpin reports personal fees from AstraZeneca, Boehringer Ingelheim, Chiesi, GSK, Novartis, and Pfizer, and non-financial support Boehringer Ingelheim and Novartis. MK Han has received personal fees from AstraZeneca, Boehringer Ingelheim, Merck, Mylan and GSK and research support from Novartis and Sunovion. CE Jones, S Lettis, DA Lipson, N Martin, H Quasny, and L Sail are GSK employees and hold stock/shares in GSK. R Kalhan reports grants from the National Heart, Lung, and Blood Institute during the conduct of the study, grants and personal fees from Boehringer Ingelheim, grants from PneumRx (BTG), grants from Spiration, grants and personal fees from AstraZeneca, personal fees from CVS Caremark, personal fees from Aptus Health, grants and personal fees from GSK, personal fees from Boston Scientific, and personal fees from Boston Consulting Group. P Lange reports personal fees from GSK, AstraZeneca, Chiesi and Boehringer Ingelheim, and grant support from Boehringer Ingelheim. DA Lomas reports personal fees from GSK and Grifols; he chaired the GSK Respiratory Therapy Area Board in 2012–2015. JM Echave-Sustaeta María-Tomé has received personal fees from GSK and was an investigator in the IMPACT trial. FJ Martinez has received personal fees and non-financial support from, AstraZeneca, Boehringer Ingelheim, Genentech, GSK, Inova Fairfax Health System, Miller Communications, National Society for Continuing Education, Novartis, Pearl Pharmaceuticals, PeerView Communications, Prime Communications, Puerto Rico Respiratory Society, Chiesi, Sunovion, Theravance, Potomac, University of Alabama-Birmingham, Physicians Education Resource, Canadian Respiratory Network, Teva and Dartmouth; and personal fees from Columbia University, MD Magazine, Methodist Hospital Brooklyn, New York University, UpToDate, WebMD/MedScape, Patara/Respivant, the American Thoracic Society, Rockpointe, Rare Disease Healthcare Communications and the France Foundation, and has taken part in advisory boards for AstraZeneca, Boehringer Ingelheim, ProterrixBio, Genentech, Novartis, Pearl Pharmaceuticals, Theravance, Zambon, Gala, Chiesi, GSK, Sunovion, and Teva, steering committees for AstraZeneca, Pearl Pharmaceuticals, Afferent/Merck, Gilead, Nitto, Patara/Respivant, Biogen, Veracyte, Prometic, Bayer, ProMedior and GSK, and been an advisor for Bridge Biotherapeutics. TM Siler has received research grants from AstraZeneca, Boehringer Ingelheim, Chiesi Farmaceutici, Compleware, Evidera (PPD), Forest Research Institute (now AstraZeneca), GSK, Novartis, Pearl Therapeutics, Proterix BioPharma, Oncocyte, Sanofi, Seer, Sunovion, Teva, Theravance BioPharma, Vapotherm, Restorbio and Westward, and personal fees from GSK, Sunovion, Theravance Biopharma and Vapotherm. D Singh reports personal fees from GSK, AstraZeneca, Boehringer Ingelheim, Chiesi, Cipla, Genentech, Glenmark, Menarini, Mundipharma, Novartis, Peptinnovate, Pfizer, Pulmatrix, Theravance, and Verona, and grant support from AstraZeneca, Boehringer Ingelheim, Chiesi, Glenmark, Menarini, Mundipharma, Novartis, Pfizer, Pulmatrix, Theravance and Verona. B Thomashow has taken part in advisory boards for AstraZeneca and GSK. H Watz reports personal fees from AstraZeneca, Boehringer Ingelheim, BerlinChemie, Chiesi, GSK, Novartis, Takeda, and Verona Pharma. R Wise reports personal fees from AstraZeneca/Medimmune/Pearl, Boehringer Ingelheim, Contrafect, Pulmonx, Roche, Spiration, Sunovion, Circassia, Pneuma, Verona, Mylan/Theravance, Propeller Health, AbbVie GSK, Merck, Kiniksa and Galderma, and has received research grants from AstraZeneca/MedImmune/Pearl, Boehringer Ingelheim, Pearl Therapeutics, Sanofi-Aventis and GSK. NA Hanania was an investigator on the IMPACT trial and reports receiving personal fees from GSK, AstraZeneca, Boehringer Ingelheim, Sanofi Genzyme, Novartis, Regeneron, Genentech, Sunovion, and Mylan. He also received research support from GSK, Boehringer Ingelheim, Sanofi Genzyme, Novartis and Astra Zeneca. ELLIPTA is owned by or licensed to the GSK Group of Companies., (JCOPDF © 2021.)
- Published
- 2021
- Full Text
- View/download PDF
31. High-resolution ex vivo NMR spectroscopy of human Z α 1 -antitrypsin.
- Author
-
Jagger AM, Waudby CA, Irving JA, Christodoulou J, and Lomas DA
- Subjects
- Genetic Predisposition to Disease genetics, Glycoproteins, Humans, Models, Molecular, Molecular Medicine, Mutation, Protein Aggregation, Pathological, Protein Conformation, Recombinant Proteins, Serine Proteinase Inhibitors chemistry, Magnetic Resonance Spectroscopy methods, alpha 1-Antitrypsin chemistry, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin metabolism, alpha 1-Antitrypsin Deficiency genetics, alpha 1-Antitrypsin Deficiency metabolism
- Abstract
Genetic mutations predispose the serine protease inhibitor α
1 -antitrypsin to misfolding and polymerisation within hepatocytes, causing liver disease and chronic obstructive pulmonary disease. This misfolding occurs via a transiently populated intermediate state, but our structural understanding of this process is limited by the instability of recombinant α1 -antitrypsin variants in solution. Here we apply NMR spectroscopy to patient-derived samples of α1 -antitrypsin at natural isotopic abundance to investigate the consequences of disease-causing mutations, and observe widespread chemical shift perturbations for methyl groups in Z AAT (E342K). By comparison with perturbations induced by binding of a small-molecule inhibitor of misfolding we conclude that they arise from rapid exchange between the native conformation and a well-populated intermediate state. The observation that this intermediate is stabilised by inhibitor binding suggests a paradoxical approach to the targeted treatment of protein misfolding disorders, wherein the stabilisation of disease-associated states provides selectivity while inhibiting further transitions along misfolding pathways.- Published
- 2020
- Full Text
- View/download PDF
32. Non-invasive testing for liver pathology in alpha-1 antitrypsin deficiency.
- Author
-
Abbas SH, Pickett E, Lomas DA, Thorburn D, Gooptu B, Hurst JR, and Marshall A
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Female, Humans, Middle Aged, Predictive Value of Tests, Sensitivity and Specificity, Young Adult, Elasticity Imaging Techniques, Liver Cirrhosis diagnostic imaging, Liver Cirrhosis pathology, alpha 1-Antitrypsin Deficiency diagnostic imaging, alpha 1-Antitrypsin Deficiency pathology
- Abstract
Background: Many patients with alpha-1 antitrypsin deficiency (A1ATD) receive care in respiratory clinics without access to specialist hepatology expertise. Liver disease can develop asymptomatically, and non-invasive markers of fibrosis may help identify patients who require definitive assessment with liver biopsy. We evaluated the utility of non-invasive markers of liver fibrosis in A1ATD to guide testing in settings without ready access to hepatology expertise., Methods: Patients attending the London A1ATD service undergo assessment using blood tests to calculate the 'APRI' and 'FIB-4' score, liver ultrasound and Fibroscan. Liver biopsy is offered to patients who have abnormal liver function tests with abnormal liver ultrasound and/or liver stiffness >6 kPa on Fibroscan. Liver biopsies were assessed for the presence of A1AT, steatosis, fibrosis and inflammation., Results: 75 patients with A1ATD had results for analysis, 56% were female, age 16-82 years. 75% of patients had Fibroscan <6 kPa, 19% had Fibroscan 6-7.9 kPa and 6%>8 kPa. There was a significant correlation between FIB-4 and Fibroscan (r=0.244, p=0.035). Fibroscan >6 kPa corresponded to a FIB-4 score of >1.26. However, FIB-4 >1.26 had poor sensitivity (47%), specificity (32%) and positive-predictive value (PPV; 36%) to identify Fibroscan >6 kPa. The negative-predictive value (NPV) was stronger at 81%. APRI data were similar. Twelve patients underwent liver biopsy, with 11 reports available for analysis. Six had FIB-4 scores<1.26 and five had Fibroscan of <6 kPa. A1AT was present in 64% of biopsies, steatosis in 82%, mild fibrosis in 36%, moderate fibrosis in 9% and severe fibrosis in 9%., Conclusion: A combination of liver ultrasound and non-invasive fibrosis tests can help identify patients with A1ATD liver injury. However, APRI and FIB-4 scores alone had poor sensitivity and specificity to justify use as an independent tool for liver pathology in A1ATD., Competing Interests: Competing interests: No other authors have any conflicts of interest to declare., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
33. The Effect of Inhaled Corticosteroid Withdrawal and Baseline Inhaled Treatment on Exacerbations in the IMPACT Study. A Randomized, Double-Blind, Multicenter Clinical Trial.
- Author
-
Han MK, Criner GJ, Dransfield MT, Halpin DMG, Jones CE, Kilbride S, Lange P, Lettis S, Lipson DA, Lomas DA, Martin N, Wise RA, Singh D, and Martinez FJ
- Subjects
- Administration, Inhalation, Aged, Double-Blind Method, Female, Humans, Male, Middle Aged, Benzyl Alcohols administration & dosage, Bronchodilator Agents administration & dosage, Chlorobenzenes administration & dosage, Fluticasone administration & dosage, Pulmonary Disease, Chronic Obstructive drug therapy, Pulmonary Disease, Chronic Obstructive physiopathology, Quinuclidines administration & dosage, Substance Withdrawal Syndrome physiopathology
- Abstract
Rationale: In the IMPACT (Informing the Pathway of Chronic Obstructive Pulmonary Disease Treatment) trial, fluticasone furoate (FF)/umeclidinium (UMEC)/vilanterol (VI) significantly reduced exacerbations compared with FF/VI or UMEC/VI in patients with symptomatic chronic obstructive pulmonary disease and a history of exacerbations. Objectives: To understand whether inhaled corticosteroid (ICS) withdrawal affected IMPACT results, given direct transition from prior maintenance medication to study medication at randomization. Methods: Exacerbations and change from baseline in trough FEV
1 and St. George's Respiratory Questionnaire results were analyzed by prior ICS use. Exacerbations were also analyzed while excluding data from the first 30 days. Measurements and Main Results: FF/UMEC/VI significantly reduced the annual moderate/severe exacerbation rate compared with UMEC/VI in prior ICS users (29% reduction; P < 0.001), but only a numerical reduction was seen among prior ICS nonusers (12% reduction; P = 0.115). To minimize impact from ICS withdrawal, in an analysis excluding the first 30 days, FF/UMEC/VI continued to significantly reduce the annual on-treatment moderate/severe exacerbation rate (19%; P < 0.001) compared with UMEC/VI. The benefit of FF/UMEC/VI compared with UMEC/VI was seen for severe exacerbation rates, regardless of prior ICS use (prior ICS users, 35% reduction; P < 0.001; non-ICS users, 35% reduction; P = 0.018), and overall when excluding the first 30 days (29%; P < 0.001). Improvements from baseline with FF/UMEC/VI compared with UMEC/VI were also maintained throughout the study for both trough FEV1 and St. George's Respiratory Questionnaire, regardless of prior ICS use. Conclusions: These data support the important treatment effects of FF/UMEC/VI combination therapy on exacerbation reduction, lung function, and quality of life that do not appear to be related to abrupt ICS withdrawal.Clinical trial registered with www.clinicaltrials.gov (NCT02164513).- Published
- 2020
- Full Text
- View/download PDF
34. The structural basis for Z α 1 -antitrypsin polymerization in the liver.
- Author
-
Faull SV, Elliston ELK, Gooptu B, Jagger AM, Aldobiyan I, Redzej A, Badaoui M, Heyer-Chauhan N, Rashid ST, Reynolds GM, Adams DH, Miranda E, Orlova EV, Irving JA, and Lomas DA
- Abstract
The serpinopathies are among a diverse set of conformational diseases that involve the aberrant self-association of proteins into ordered aggregates. α
1 -Antitrypsin deficiency is the archetypal serpinopathy and results from the formation and deposition of mutant forms of α1 -antitrypsin as "polymer" chains in liver tissue. No detailed structural analysis has been performed of this material. Moreover, there is little information on the relevance of well-studied artificially induced polymers to these disease-associated molecules. We have isolated polymers from the liver tissue of Z α1 -antitrypsin homozygotes (E342K) who have undergone transplantation, labeled them using a Fab fragment, and performed single-particle analysis of negative-stain electron micrographs. The data show structural equivalence between heat-induced and ex vivo polymers and that the intersubunit linkage is best explained by a carboxyl-terminal domain swap between molecules of α1 -antitrypsin., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution License 4.0 (CC BY).)- Published
- 2020
- Full Text
- View/download PDF
35. Single-Inhaler Triple Therapy and Health-Related Quality of Life in COPD: The IMPACT Study.
- Author
-
Tabberer M, Jones CE, Kilbride S, Halpin DMG, Lomas DA, Pascoe S, Singh D, Wise RA, Criner GJ, Lange P, Dransfield MT, Han MK, Martinez FJ, Kaisermann MC, and Lipson DA
- Subjects
- Administration, Inhalation, Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Nebulizers and Vaporizers, Surveys and Questionnaires, Androstadienes therapeutic use, Benzyl Alcohols therapeutic use, Bronchodilator Agents therapeutic use, Chlorobenzenes therapeutic use, Pulmonary Disease, Chronic Obstructive drug therapy, Pulmonary Disease, Chronic Obstructive psychology, Quality of Life psychology, Quinuclidines therapeutic use
- Abstract
Introduction: The phase 3 InforMing the PAthway of COPD (chronic obstructive pulmonary disease) Treatment (IMPACT) trial, single-inhaler therapy with fluticasone furoate (FF) 100 μg, umeclidinium (UMEC) 62.5 μg, and vilanterol (VI) 25 μg demonstrated a reduction in the rate of moderate or severe exacerbations compared with FF/VI or UMEC/VI in patients with symptomatic COPD at risk of exacerbations. This article reports additional evidence of improvements in symptoms and health-related quality of life (HRQoL) with FF/UMEC/VI compared with either FF/VI or UMEC/VI from the IMPACT study., Methods: Patient-reported HRQoL assessments and symptom measures included as pre-specified IMPACT end points were the St George's Respiratory Questionnaire (SGRQ), COPD Assessment Test (CAT), and Baseline Dyspnea Index (BDI) as the anchor for the Transitional Dyspnea Index (TDI) focal score (BDI/TDI) in a subset of patients enrolled at study sites in North America and Europe. Change from baseline was assessed at weeks 4, 28, and 52., Results: The intent-to-treat population included 10,355 patients (TDI population: 5058 patients). Clinically meaningful improvements in SGRQ total score between baseline and week 52 favored FF/UMEC/VI over FF/VI (- 1.8 units, p < 0.001) and UMEC/VI (- 1.8 units, p < 0.001). Similar improvements in the CAT and TDI focal score were also observed with FF/UMEC/VI versus FF/VI or UMEC/VI., Conclusions: This study demonstrates that in patients with symptomatic COPD at risk of exacerbations, once-daily FF/UMEC/VI, compared with FF/VI or UMEC/VI, improves patient-perceived HRQoL and symptoms., Trial Registration Number: NCT02164513.
- Published
- 2020
- Full Text
- View/download PDF
36. Intrahepatic heteropolymerization of M and Z alpha-1-antitrypsin.
- Author
-
Laffranchi M, Elliston EL, Miranda E, Perez J, Ronzoni R, Jagger AM, Heyer-Chauhan N, Brantly ML, Fra A, Lomas DA, and Irving JA
- Subjects
- Alleles, Catalytic Domain drug effects, Crystallography, Endoplasmic Reticulum genetics, Endoplasmic Reticulum pathology, Epitopes genetics, Epitopes immunology, Genetic Variation genetics, Hepatocytes metabolism, Hepatocytes pathology, Humans, Liver metabolism, Liver pathology, Liver Cirrhosis metabolism, Liver Cirrhosis pathology, Protein Aggregates immunology, Protein Conformation, alpha 1-Antitrypsin chemistry, alpha 1-Antitrypsin ultrastructure, alpha 1-Antitrypsin Deficiency immunology, alpha 1-Antitrypsin Deficiency pathology, Liver Cirrhosis genetics, Protein Aggregates genetics, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin Deficiency genetics
- Abstract
The α-1-antitrypsin (or alpha-1-antitrypsin, A1AT) Z variant is the primary cause of severe A1AT deficiency and forms polymeric chains that aggregate in the endoplasmic reticulum of hepatocytes. Around 2%-5% of Europeans are heterozygous for the Z and WT M allele, and there is evidence of increased risk of liver disease when compared with MM A1AT individuals. We have shown that Z and M A1AT can copolymerize in cell models, but there has been no direct observation of heteropolymer formation in vivo. To this end, we developed a monoclonal antibody (mAb2H2) that specifically binds to M in preference to Z A1AT, localized its epitope using crystallography to a region perturbed by the Z (Glu342Lys) substitution, and used Fab fragments to label polymers isolated from an MZ heterozygote liver explant. Glu342 is critical to the affinity of mAb2H2, since it also recognized the mild S-deficiency variant (Glu264Val) present in circulating polymers from SZ heterozygotes. Negative-stain electron microscopy of the Fab2H2-labeled liver polymers revealed that M comprises around 6% of the polymer subunits in the MZ liver sample. These data demonstrate that Z A1AT can form heteropolymers with polymerization-inert variants in vivo with implications for liver disease in heterozygous individuals.
- Published
- 2020
- Full Text
- View/download PDF
37. Reduction in All-Cause Mortality with Fluticasone Furoate/Umeclidinium/Vilanterol in Patients with Chronic Obstructive Pulmonary Disease.
- Author
-
Lipson DA, Crim C, Criner GJ, Day NC, Dransfield MT, Halpin DMG, Han MK, Jones CE, Kilbride S, Lange P, Lomas DA, Lettis S, Manchester P, Martin N, Midwinter D, Morris A, Pascoe SJ, Singh D, Wise RA, and Martinez FJ
- Subjects
- Administration, Inhalation, Aged, Cause of Death, Drug Therapy, Combination, Female, Forced Expiratory Volume, Humans, Male, Middle Aged, Proportional Hazards Models, Pulmonary Disease, Chronic Obstructive physiopathology, Severity of Illness Index, Adrenergic beta-2 Receptor Agonists therapeutic use, Androstadienes therapeutic use, Benzyl Alcohols therapeutic use, Chlorobenzenes therapeutic use, Glucocorticoids therapeutic use, Mortality, Muscarinic Antagonists therapeutic use, Pulmonary Disease, Chronic Obstructive drug therapy, Quinuclidines therapeutic use
- Abstract
Rationale: The IMPACT (Informing the Pathway of Chronic Obstructive Pulmonary Disease Treatment) trial demonstrated a significant reduction in all-cause mortality (ACM) risk with fluticasone furoate/umeclidinium/vilanterol (FF/UMEC/VI) versus UMEC/VI in patients with chronic obstructive pulmonary disease (COPD) at risk of future exacerbations. Five hundred seventy-four patients were censored in the original analysis owing to incomplete vital status information. Objectives: Report ACM and impact of stepping down therapy, following collection of additional vital status data. Methods: Patients were randomized 2:2:1 to FF/UMEC/VI 100/62.5/25 μg, FF/VI 100/25 μg, or UMEC/VI 62.5/25 μg following a run-in on their COPD therapies. Time to ACM was prespecified. Additional vital status data collection and subsequent analyses were performed post hoc . Measurements and Main Results: We report vital status data for 99.6% of the intention-to-treat population ( n = 10,355), documenting 98 (2.36%) deaths on FF/UMEC/VI, 109 (2.64%) on FF/VI, and 66 (3.19%) on UMEC/VI. For FF/UMEC/VI, the hazard ratio for death was 0.72 (95% confidence interval, 0.53-0.99; P = 0.042) versus UMEC/VI and 0.89 (95% confidence interval, 0.67-1.16; P = 0.387) versus FF/VI. Independent adjudication confirmed lower rates of cardiovascular and respiratory death and death associated with the patient's COPD. Conclusions: In this secondary analysis of an efficacy outcome from the IMPACT trial, once-daily single-inhaler FF/UMEC/VI triple therapy reduced the risk of ACM versus UMEC/VI in patients with symptomatic COPD and a history of exacerbations.
- Published
- 2020
- Full Text
- View/download PDF
38. Single-inhaler triple therapy fluticasone furoate/umeclidinium/vilanterol versus fluticasone furoate/vilanterol and umeclidinium/vilanterol in patients with COPD: results on cardiovascular safety from the IMPACT trial.
- Author
-
Day NC, Kumar S, Criner G, Dransfield M, Halpin DMG, Han MK, Jones CE, Kaisermann MC, Kilbride S, Lange P, Lomas DA, Martin N, Martinez FJ, Singh D, Wise R, and Lipson DA
- Subjects
- Aged, Androstadienes adverse effects, Benzyl Alcohols adverse effects, Cardiovascular Diseases chemically induced, Cardiovascular Diseases diagnosis, Chlorobenzenes adverse effects, Double-Blind Method, Drug Combinations, Female, Humans, Male, Middle Aged, Quinuclidines adverse effects, Androstadienes administration & dosage, Benzyl Alcohols administration & dosage, Chlorobenzenes administration & dosage, Nebulizers and Vaporizers trends, Pulmonary Disease, Chronic Obstructive diagnosis, Pulmonary Disease, Chronic Obstructive drug therapy, Quinuclidines administration & dosage
- Abstract
Background: This analysis of the IMPACT study assessed the cardiovascular (CV) safety of single-inhaler triple therapy with fluticasone furoate/umeclidinium/vilanterol (FF/UMEC/VI) versus FF/VI and UMEC/VI dual therapy., Methods: IMPACT was a 52-week, randomized, double-blind, multicenter Phase III study comparing the efficacy and safety of FF/UMEC/VI 100/62.5/25 mcg with FF/VI 100/25 mcg or UMEC/VI 62.5/25 mcg in patients ≥40 years of age with symptomatic chronic obstructive pulmonary disease (COPD) and ≥1 moderate/severe exacerbation in the previous year. The inclusion criteria for the study were intentionally designed to permit the enrollment of patients with significant concurrent CV disease/risk. CV safety assessments included proportion of patients with and exposure-adjusted rates of on-treatment CV adverse events of special interest (CVAESI) and major adverse cardiac events (MACE), as well as time-to-first (TTF) CVAESI, and TTF CVAESI resulting in hospitalization/prolonged hospitalization or death., Results: Baseline CV risk factors were similar across treatment groups. Overall, 68% of patients (n = 7012) had ≥1 CV risk factor and 40% (n = 4127) had ≥2. At baseline, 29% of patients reported a current/past cardiac disorder and 58% reported a current/past vascular disorder. The proportion of patients with on-treatment CVAESI was 11% for both FF/UMEC/VI and UMEC/VI, and 10% for FF/VI. There was no statistical difference for FF/UMEC/VI versus FF/VI or UMEC/VI in TTF CVAESI (hazard ratio [HR]: 0.98, 95% confidence interval [CI]: 0.85, 1.11; p = 0.711 and HR: 0.92, 95% CI: 0.78, 1.08; p = 0.317, respectively) nor TTF CVAESI leading to hospitalization/prolonged hospitalization or death (HR: 1.19, 95% CI: 0.93, 1.51; p = 0.167 and HR: 0.96, 95% CI: 0.72, 1.27; p = 0.760, respectively). On-treatment MACE occurred in ≤3% of patients across treatment groups, with similar prevalence and rates between treatments., Conclusions: In a symptomatic COPD population with a history of exacerbations and a high rate of CV disease/risk, the proportion of patients with CVAESI and MACE was 10-11% and 1-3%, respectively, across treatment arms, and the risk of CVAESI was low and similar across treatment arms. There was no statistically significant increased CV risk associated with the use of FF/UMEC/VI versus FF/VI or UMEC/VI, and UMEC/VI versus FF/VI., Trial Registration: NCT02164513 (GSK study number CTT116855).
- Published
- 2020
- Full Text
- View/download PDF
39. The effect of exacerbation history on outcomes in the IMPACT trial.
- Author
-
Halpin DMG, Dransfield MT, Han MK, Jones CE, Kilbride S, Lange P, Lipson DA, Lomas DA, Martinez FJ, Pascoe S, Singh D, Wise R, and Criner GJ
- Subjects
- Administration, Inhalation, Adrenergic beta-2 Receptor Agonists administration & dosage, Adrenergic beta-2 Receptor Agonists adverse effects, Aged, Androstadienes adverse effects, Benzyl Alcohols adverse effects, Bronchodilator Agents administration & dosage, Bronchodilator Agents adverse effects, Chlorobenzenes adverse effects, Disease Progression, Double-Blind Method, Drug Combinations, Female, Health Status, Humans, Lung physiopathology, Male, Middle Aged, Muscarinic Antagonists administration & dosage, Muscarinic Antagonists adverse effects, Nebulizers and Vaporizers, Pulmonary Disease, Chronic Obstructive diagnosis, Pulmonary Disease, Chronic Obstructive physiopathology, Quinuclidines adverse effects, Recovery of Function, Time Factors, Treatment Outcome, Androstadienes administration & dosage, Benzyl Alcohols administration & dosage, Chlorobenzenes administration & dosage, Lung drug effects, Pulmonary Disease, Chronic Obstructive drug therapy, Quinuclidines administration & dosage
- Abstract
IMPACT, a 52-week, randomised, double-blind trial, assessed the efficacy and safety of fluticasone furoate/umeclidinium/vilanterol (FF/UMEC/VI) triple therapy versus FF/VI or UMEC/VI in patients with symptomatic COPD and a history of exacerbations.Subgroup analyses assessed whether the efficacy of FF/UMEC/VI versus FF/VI or UMEC/VI and UMEC/VI versus FF/VI varies according to prior exacerbation history, and the combined effects of exacerbation history and blood eosinophil counts. Three subgroups were defined: single moderate (1 moderate/no severe; n=3056 (30%)), frequent moderate (≥2 moderate/no severe; n=4628 (45%)) and severe (≥1 severe/any moderate; n=2671 (26%)). End-points included annual on-treatment moderate/severe exacerbation rate (pre-specified), lung function and health status (both post-hoc).Moderate/severe exacerbation rates (reduction % (95% CI)) were reduced in the FF/UMEC/VI group versus FF/VI (single moderate 20% (10-29), frequent moderate 11% (2-19), severe 17% (7-26)) and versus UMEC/VI (single moderate 18% (5-29), frequent moderate 29% (21-37), severe 26% (14-35)). Moderate/severe exacerbation rates were reduced in the FF/VI group versus UMEC/VI in the frequent moderate subgroup; a numerical reduction was observed in the severe subgroup (single moderate 2% (-12-18), frequent moderate 21% (11-29), severe 11% (-3-22)). Moderate/severe exacerbation rates were lower in the FF/VI group compared with UMEC/VI in patients with higher eosinophil counts. FF/UMEC/VI improved lung function and health status versus both dual therapies irrespective of exacerbation subgroup. UMEC/VI improved lung function versus FF/VI in all subgroups.Triple therapy was more effective than dual regardless of exacerbation history, consistent with results in the intent-to-treat population. Comparisons between dual therapies were influenced by prior exacerbation history and eosinophil counts., Competing Interests: Conflict of interest: D.M.G. Halpin reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; personal fees from AstraZeneca, personal fees and non-financial support from Boehringer Ingelheim, personal fees from Chiesi, personal fees from GlaxoSmithKline, personal fees and non-financial support from Novartis, personal fees from Pfizer, outside the submitted work. Conflict of interest: M.T. Dransfield reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; grants from Department of Defense, personal fees and other from Boehringer Ingelheim, personal fees and other from GlaxoSmithKline, other from Novartis, personal fees and other from AstraZeneca, other from Yungjin, personal fees and other from PneumRx/BTG, other from Pulmonx, personal fees from Genentech, other from Boston Scientific, personal fees from Quark Pharmaceuticals, grants from NIH, personal fees from Mereo, grants from American Lung Association, grants from NIH, outside the submitted work. Conflict of interest: M.K. Han reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; personal fees from AstraZeneca, personal fees from Boehringer Ingelheim, other from Novartis, other from Sunovion, personal fees from Mylan, outside the submitted work. Conflict of interest: E. Jones reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; personal fees from GlaxoSmithKline, other from GlaxoSmithKline, outside the submitted work. Conflict of interest: S. Kilbride reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; personal fees from GlaxoSmithKline, other from GlaxoSmithKline, outside the submitted work. Conflict of interest: P. Lange reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; grants and personal fees from GlaxoSmithKline, personal fees from AstraZeneca, personal fees from Boehringer Ingelheim, personal fees from Chiesi, outside the submitted work. Conflict of interest: D.A. Lipson reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; personal fees from GlaxoSmithKline, other from GlaxoSmithKline, outside the submitted work. Conflict of interest: D.A. Lomas reports personal fees and other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; grants from GlaxoSmithKline, personal fees and other from GlaxoSmithKline, personal fees from Grifols, outside the submitted work. Conflict of interest: F.J. Martinez reports grants, personal fees and non-financial support from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; personal fees and non-financial support from American College of Chest Physicians, personal fees and non-financial support from AstraZeneca, personal fees and non-financial support from Boehringer Ingelheim, non-financial support from ProterrixBio, personal fees from Columbia University, personal fees and non-financial support from ConCert, personal fees and non-financial support from Genentech, personal fees and non-financial support from GlaxoSmithKline, personal fees and non-financial support from Inova Fairfax Health System, personal fees from Integritas, personal fees from MD Magazine, personal fees from Methodist Hospital Brooklyn, personal fees and non-financial support from Miller Communicatinos, personal fees and non-financial support from National Association for Continuing Education, personal fees and non-financial support from Novartis, personal fees from New York University, personal fees and non-financial support from Pearl Pharmaceuticals, personal fees and non-financial support from PeerView Communications, personal fees and non-financial support from Prime Communications, personal fees and non-financial support from Puerto Rican Respiratory Society, personal fees and non-financial support from Chiesi, personal fees and non-financial support from Sunovion, personal fees and non-financial support from Theravance, personal fees from UpToDate, personal fees from WebMD/MedScape, personal fees from Western Connecticut Health Network, other from Afferent/Merck, non-financial support from Gilead, non-financial support from Nitto, personal fees from Patara/Respivant, personal fees from PlatformIQ, personal fees and non-financial support from Potomac, other from Biogen, personal fees and non-financial support from University of Alabama Birmingham, other from Veracyte, non-financial support from Zambon, personal fees from American Thoracic Society, grants from NIH, personal fees and non-financial support from Physicians Education Resource, personal fees from Rockpointe, other from Prometic, personal fees from Rare Disease Healthcare Communications, other from Bayer, other from Bridge Biotherapeutics, personal fees and non-financial support from Canadian Respiratory Network, other from ProMedior, personal fees and non-financial support from Teva, personal fees from France Foundation, personal fees and non-financial support from Dartmouth, outside the submitted work. Conflict of interest: S. Pascoe reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; personal fees from GlaxoSmithKline, other from GlaxoSmithKline, outside the submitted work. Conflict of interest: D. Singh reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; personal fees from GlaxoSmithKline, grants and personal fees from AstraZeneca, grants and personal fees from Boehringer Ingelheim, grants and personal fees from Chiesi, personal fees from Cipla, personal fees from Genentech, grants and personal fees from Glenmark, grants and personal fees from Menarini, grants and personal fees from Mundipharma, grants and personal fees from Novartis, personal fees from Peptinnovate, grants and personal fees from Pfizer, grants and personal fees from Pulmatrix, grants and personal fees from Therevance, grants and personal fees from Verona, outside the submitted work. Conflict of interest: R. Wise reports personal fees, non-financial support and other from GlaxoSmithKline, during the conduct of the study; grants and personal fees from AstraZeneca / Medimmune / Pearl, grants and personal fees from Boehringer Ingelheim, personal fees from Contrafect, personal fees from Pulmonx, personal fees from Roche, personal fees from Spiration, personal fees from Sunovion, grants from Pearl Therapeutics, personal fees from Merck, personal fees from Circassia, personal fees from Pneuma, personal fees from Verona, personal fees from Mylan/Theravance, personal fees from Propeller Health, grants from Sanofi-Aventis, personal fees from AbbVie, grants and personal fees from GSK, outside the submitted work. Conflict of interest: G.J. Criner reports other from GlaxoSmithKline, non-financial support from GlaxoSmithKline, during the conduct of the study; personal fees from Almirall, personal fees from AstraZeneca, personal fees from Boehringer Ingelheim, personal fees from Chiesi, personal fees from CSA Medical, personal fees from Eolo, personal fees from GlaxoSmithKline, personal fees from HGE Technologies, personal fees from Novartis, personal fees from Nuvaira, personal fees from Olympus, personal fees from Pulmonx, personal fees from Verona, personal fees from NGM Bio, outside the submitted work., (Copyright ©ERS 2020.)
- Published
- 2020
- Full Text
- View/download PDF
40. Heme metabolism genes Downregulated in COPD Cachexia.
- Author
-
Wilson AC, Kumar PL, Lee S, Parker MM, Arora I, Morrow JD, Wouters EFM, Casaburi R, Rennard SI, Lomas DA, Agusti A, Tal-Singer R, Dransfield MT, Wells JM, Bhatt SP, Washko G, Thannickal VJ, Tiwari HK, Hersh CP, Castaldi PJ, Silverman EK, and McDonald MN
- Subjects
- Aged, Aged, 80 and over, Cachexia epidemiology, Cohort Studies, Down-Regulation physiology, Female, Follow-Up Studies, Genome-Wide Association Study methods, Humans, Longitudinal Studies, Male, Middle Aged, Pulmonary Disease, Chronic Obstructive epidemiology, Cachexia genetics, Cachexia metabolism, Heme genetics, Heme metabolism, Pulmonary Disease, Chronic Obstructive genetics, Pulmonary Disease, Chronic Obstructive metabolism
- Abstract
Introduction: Cachexia contributes to increased mortality and reduced quality of life in Chronic Obstructive Pulmonary Disease (COPD) and may be associated with underlying gene expression changes. Our goal was to identify differential gene expression signatures associated with COPD cachexia in current and former smokers., Methods: We analyzed whole-blood gene expression data from participants with COPD in a discovery cohort (COPDGene, N = 400) and assessed replication (ECLIPSE, N = 114). To approximate the consensus definition using available criteria, cachexia was defined as weight-loss > 5% in the past 12 months or low body mass index (BMI) (< 20 kg/m
2 ) and 1/3 criteria: decreased muscle strength (six-minute walk distance < 350 m), anemia (hemoglobin < 12 g/dl), and low fat-free mass index (FFMI) (< 15 kg/m2 among women and < 17 kg/m2 among men) in COPDGene. In ECLIPSE, cachexia was defined as weight-loss > 5% in the past 12 months or low BMI and 3/5 criteria: decreased muscle strength, anorexia, abnormal biochemistry (anemia or high c-reactive protein (> 5 mg/l)), fatigue, and low FFMI. Differential gene expression was assessed between cachectic and non-cachectic subjects, adjusting for age, sex, white blood cell counts, and technical covariates. Gene set enrichment analysis was performed using MSigDB., Results: The prevalence of COPD cachexia was 13.7% in COPDGene and 7.9% in ECLIPSE. Fourteen genes were differentially downregulated in cachectic versus non-cachectic COPD patients in COPDGene (FDR < 0.05) and ECLIPSE (FDR < 0.05)., Discussion: Several replicated genes regulating heme metabolism were downregulated among participants with COPD cachexia. Impaired heme biosynthesis may contribute to cachexia development through free-iron buildup and oxidative tissue damage.- Published
- 2020
- Full Text
- View/download PDF
41. Alpha 1 -Antitrypsin Deficiency.
- Author
-
Strnad P, McElvaney NG, and Lomas DA
- Subjects
- Graft vs Host Disease drug therapy, Heterozygote, Humans, Liver Diseases physiopathology, Mutation, Pulmonary Emphysema physiopathology, Liver Diseases etiology, Pulmonary Emphysema etiology, alpha 1-Antitrypsin therapeutic use, alpha 1-Antitrypsin Deficiency complications, alpha 1-Antitrypsin Deficiency diagnosis, alpha 1-Antitrypsin Deficiency genetics, alpha 1-Antitrypsin Deficiency therapy
- Published
- 2020
- Full Text
- View/download PDF
42. C. elegans expressing D76N β 2 -microglobulin: a model for in vivo screening of drug candidates targeting amyloidosis.
- Author
-
Faravelli G, Raimondi S, Marchese L, Partridge FA, Soria C, Mangione PP, Canetti D, Perni M, Aprile FA, Zorzoli I, Di Schiavi E, Lomas DA, Bellotti V, Sattelle DB, and Giorgetti S
- Subjects
- Amyloid chemistry, Amyloid genetics, Amyloid metabolism, Animals, Animals, Genetically Modified, Caenorhabditis elegans genetics, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins metabolism, Disease Models, Animal, Mutation, Missense, Phenotype, Protein Aggregation, Pathological prevention & control, Protein Folding, beta 2-Microglobulin metabolism, Amyloidosis genetics, Drug Evaluation, Preclinical methods, beta 2-Microglobulin genetics
- Abstract
The availability of a genetic model organism with which to study key molecular events underlying amyloidogenesis is crucial for elucidating the mechanism of the disease and the exploration of new therapeutic avenues. The natural human variant of β
2 -microglobulin (D76N β2 -m) is associated with a fatal familial form of systemic amyloidosis. Hitherto, no animal model has been available for studying in vivo the pathogenicity of this protein. We have established a transgenic C. elegans line, expressing the human D76N β2 -m variant. Using the INVertebrate Automated Phenotyping Platform (INVAPP) and the algorithm Paragon, we were able to detect growth and motility impairment in D76N β2 -m expressing worms. We also demonstrated the specificity of the β2 -m variant in determining the pathological phenotype by rescuing the wild type phenotype when β2 -m expression was inhibited by RNA interference (RNAi). Using this model, we have confirmed the efficacy of doxycycline, an inhibitor of the aggregation of amyloidogenic proteins, in rescuing the phenotype. In future, this C. elegans model, in conjunction with the INVAPP/Paragon system, offers the prospect of high-throughput chemical screening in the search for new drug candidates.- Published
- 2019
- Full Text
- View/download PDF
43. Cost-Effectiveness Of Once-Daily Single-Inhaler Triple Therapy In COPD: The IMPACT Trial.
- Author
-
Ismaila AS, Risebrough N, Schroeder M, Shah D, Martin A, Goodall EC, Ndirangu K, Criner G, Dransfield M, Halpin DM, Han MK, and Lomas DA
- Subjects
- Administration, Inhalation, Aged, Androstadienes adverse effects, Benzyl Alcohols adverse effects, Bronchodilator Agents adverse effects, Canada, Chlorobenzenes adverse effects, Clinical Trials, Phase III as Topic, Cost-Benefit Analysis, Disease Progression, Drug Combinations, Female, Humans, Lung physiopathology, Male, Models, Economic, Nebulizers and Vaporizers, Pulmonary Disease, Chronic Obstructive diagnosis, Pulmonary Disease, Chronic Obstructive physiopathology, Quality of Life, Quality-Adjusted Life Years, Quinuclidines adverse effects, Recovery of Function, Time Factors, Treatment Outcome, Androstadienes administration & dosage, Androstadienes economics, Benzyl Alcohols administration & dosage, Benzyl Alcohols economics, Bronchodilator Agents administration & dosage, Bronchodilator Agents economics, Chlorobenzenes administration & dosage, Chlorobenzenes economics, Drug Costs, Lung drug effects, Pulmonary Disease, Chronic Obstructive drug therapy, Pulmonary Disease, Chronic Obstructive economics, Quinuclidines administration & dosage, Quinuclidines economics
- Abstract
Background: We assessed the cost-effectiveness of single-inhaler fluticasone furoate (FF)/umeclidinium (UMEC)/vilanterol (VI) versus FF/VI or UMEC/VI from a Canadian public healthcare perspective, incorporating data from the IMPACT trial in chronic obstructive pulmonary disease (COPD) (NCT02164513)., Methods: Baseline inputs and treatment effects from IMPACT were populated into the validated GALAXY-COPD disease progression model. Canadian unit costs and drug costs (Canadian dollars [C$], 2017) were applied to healthcare resource utilization and treatments. Future costs and health outcomes were discounted at 1.5% annually. Analyses were probabilistic, and outputs included exacerbation rates, costs, and life years (LYs) and quality-adjusted life years (QALYs) gained., Results: Compared with FF/VI and UMEC/VI over a lifetime horizon, the analyses predicted that treatment with FF/UMEC/VI resulted in fewer moderate and severe exacerbations, more LYs and more QALYs gained, with a small incremental cost. The base-case incremental cost-effectiveness ratio (ICER) per QALY gained was C$18,989 (95% confidence interval [CI]: C$14,665, C$25,753) versus FF/VI and C$13,776 (95% CI: C$9787, C$19,448) versus UMEC/VI. FF/UMEC/VI remained cost-effective versus both FF/VI and UMEC/VI in all sensitivity analyses, including in scenario analyses that considered different intervention and comparator discontinuation rates, and treatment effects for subsequent therapy., Conclusion: Treatment with FF/UMEC/VI was predicted to improve outcomes and be a cost-effective treatment option for patients with symptomatic COPD and a history of exacerbations compared with FF/VI or UMEC/VI, in Canada., Competing Interests: The authors declare the following conflicts of interest during the last three years in relation to this article: A.S. Ismaila, M. Schroeder and A. Martin are employees of, and hold shares in, GlaxoSmithKline plc.; A.S. Ismaila is also an unpaid, part-time professor at McMaster University, Canada. E.C. Goodall is an employee of GlaxoSmithKline plc. N. Risebrough, D. Shah and K. Ndirangu are employees of ICON plc., which received funding from GlaxoSmithKline plc. to conduct this study, but were not themselves paid for development of this article. G. Criner reports grants and personal fees from GlaxoSmithKline plc., Boehringer Ingelheim, Chiesi, Mereo, AstraZeneca, Pulmonx, PneumRx, Olympus, Broncus, Lungpacer, Mereo, Nuvaira, ResMed, Respironics and Patara, Pearl, and Sanofi, personal fees from Verona, BTG, EOLO and NGM, and grants from ALung, Fisher Paykel and Galapagos, outside the submitted work. M. Dransfield reports personal and other fees from GlaxoSmithKline plc. during the conduct of this study, and grants from Department of Defense, personal and other fees from Boehringer Ingelheim, GlaxoSmithKline plc., AstraZeneca and PneumRx/BTG, personal fees from Genentech, Quark Pharmaceuticals and Mereo, grants from NIH and American Lung Association and other fees from Novartis, Yungjin, Pulmonx and Boston Scientific, outside the submitted work. D.M.G. Halpin reports personal fees from AstraZeneca, Chiesi, GlaxoSmithKline plc. and Pfizer, and personal fees and non-financial support from Boehringer Ingelheim and Novartis, outside the submitted work. M.L. Han reports personal fees from GlaxoSmithKline plc. during the conduct of this study, and personal fees from GlaxoSmithKline plc., Boehringer Ingelheim, AstraZeneca, Mylan, and other fees from Novartis and Sunovion, outside of the submitted work. D.A. Lomas reports grants, personal fees and non-financial support from GlaxoSmithKline plc. during the conduct of the study, and personal fees and grants from GlaxoSmithKline plc., and personal fees from Griffols, outside of the submitted work. D.A. Lomas also chaired the Respiratory Therapy Area Board at GlaxoSmithKline plc. between 2012 and 2015. The authors report no other conflicts of interest in this work., (© 2019 Ismaila et al.)
- Published
- 2019
- Full Text
- View/download PDF
44. Calcium signalling in mammalian cell lines expressing wild type and mutant human α1-Antitrypsin.
- Author
-
Malintan NT, Buckingham SD, Lomas DA, and Sattelle DB
- Subjects
- Animals, CHO Cells, Cricetulus, Epilepsies, Myoclonic pathology, Heredodegenerative Disorders, Nervous System pathology, Humans, Microscopy, Confocal, Mutation, Optical Imaging, alpha 1-Antitrypsin genetics, Calcium metabolism, Calcium Signaling genetics, Epilepsies, Myoclonic genetics, Heredodegenerative Disorders, Nervous System genetics, alpha 1-Antitrypsin metabolism
- Abstract
A possible role for calcium signalling in the autosomal dominant form of dementia, familial encephalopathy with neuroserpin inclusion bodies (FENIB), has been proposed, which may point towards a mechanism by which cells could sense and respond to the accumulation of mutant serpin polymers in the endoplasmic reticulum (ER). We therefore explored possible defects in Ca
2+ -signalling, which may contribute to the pathology associated with another serpinopathy, α1 -antitrypsin (AAT) deficiency. Using CHO K1 cell lines stably expressing a wild type human AAT (MAAT) and a disease-causing polymer-forming variant (ZAAT) and the truncated variant (NHK AAT), we measured basal intracellular free Ca2+ , its responses to thapsigargin (TG), an ER Ca2+ -ATPase blocker, and store-operated Ca2+ -entry (SOCE). Our fura2 based Ca2+ measurements detected no differences between these 3 parameters in cell lines expressing MAAT and cell lines expressing ZAAT and NHK AAT mutants. Thus, in our cell-based models of α1-antitrypsin (AAT) deficiency, unlike the case for FENIB, we were unable to detect defects in calcium signalling.- Published
- 2019
- Full Text
- View/download PDF
45. Misinterpretation of time-to-first event curves can lead to inappropriate treatment.
- Author
-
Hartley B, Criner GJ, Dransfield MT, Halpin DMG, Han MK, Jones CE, Kilbride S, Lange P, Lipson DA, Lomas DA, Martin N, Martinez FJ, Singh D, Wise RA, and Lettis S
- Subjects
- Humans, Precision Medicine, Pulmonary Disease, Chronic Obstructive
- Abstract
Competing Interests: Conflict of interest: B. Hartley reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; B. Hartley is a contingent worker on assignment at GlaxoSmithKline, and holds shares in GlaxoSmithKline, outside the submitted work. Conflict of interest: G.J. Criner reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; personal fees from Almirall, AstraZeneca, Boehringer Ingelheim, Chiesi, CSA Medical, Eolo, GlaxoSmithKline, HGE Technologies, Novartis, Nuvaira, Olympus, Pulmonx, Verona and NGM Bio, outside the submitted work. Conflict of interest: M.T. Dransfield reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; grants from Department of Defense and NIH, has received personal fees for consultancy from and undertaken clinical trials for Boehringer Ingelheim, AstraZeneca, PneumRx/BTG, Boston Scientific and GlaxoSmithKline, undertaken clinical trials for Novartis, Yungjin and Pulmonx, personal fees for consultancy from Genentech and Quark Pharmaceuticals, outside the submitted work. Conflict of interest: D.M.G. Halpin reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; personal fees from AstraZeneca, Chiesi, GlaxoSmithKline and Pfizer, personal fees and non-financial support from Boehringer Ingelheim and Novartis, outside the submitted work. Conflict of interest: M.K. Han reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; personal fees from AstraZeneca and Boehringer Ingelheim, grant support from Novartis and Sunovion, outside the submitted work. Conflict of interest: C.E. Jones reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; C.E. Jones is an employee of and holds shares/options in GlaxoSmithKline, outside the submitted work. Conflict of interest: S. Kilbride reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; S. Kilbride is an employee of and holds shares/options in GlaxoSmithKline, outside the submitted work. Conflict of interest: P. Lange reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; personal fees from GlaxoSmithKline, AstraZeneca, Boehringer Ingelheim and Chiesi, outside the submitted work. Conflict of interest: D.A. Lipson reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; D.A. Lipson is an employee of and holds shares/options in GlaxoSmithKline, outside the submitted work. Conflict of interest: D.A. Lomas reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; grants, personal fees for consultancy and honoraria from GlaxoSmithKline, personal fees for consultancy from Grifols, outside the submitted work. Conflict of interest: N. Martin reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; N. Martin is an employee of and holds shares/options in GlaxoSmithKline, outside the submitted work. Conflict of interest: F.J. Martinez reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; personal fees and travel support for educational activities from American College of Chest Physicians, Inova Fairfax Health System, MD Magazine, Miller Communications, National Association for Continuing Education, PeerView Communications, Prime Communications, Puerto Rican Respiratory Society, Potomac, University of Alabama Birmingham, Physicians Education Resource, Canadian Respiratory Network and Dartmouth, personal fees for advisory board work, steering committee work and lectures, and travel support from AstraZeneca, personal fees for advisory board work, data monitoring committee work and lectures, and travel support from Boehringer Ingelheim and Genentech, non-financial support for advisory board work from ProterrixBio, personal fees for educational activities from Columbia University, Integritas, Methodist Hospital Brooklyn, New York University, UpToDate, WebMD/MedScape, Western Connecticut Health Network, PlatformIQ, Rockpointe, Rare Disease Healthcare Communications and France Foundation, personal fees for advisory board work and travel support from ConCert, Sunovion, Theravance and Teva, personal fees for advisory board work and non-financial support for travel, lecturing, steering committee work and data monitoring committee work from GlaxoSmithKline, personal fees for advisory board work and lectures, and travel support from Novartis, personal fees for advisory board work and non-financial support for steering committee work from Pearl Pharmaceuticals, personal fees for advisory board work and educational activites, and travel support from Chiesi, non-financial support for steering committee work from Afferent/Merck, Gilead, Nitto, Veracyte, Prometic, Bayer and ProMedior, personal fees for consultancy and steering committee work from Patara/Respivant, non-financial support for data monitoring committee and steering committee work from Biogen, non-financial support for lecturing and advisory board work from Zambon, personal fees for journal editorship from American Thoracic Society, grants from NIH, non-financial support for consultancy from Bridge Biotherapeutics, outside the submitted work. Conflict of interest: D. Singh reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; personal fees from GlaxoSmithKline, Cipla, Genentech and Peptinnovate, grants and personal fees from AstraZeneca, Boehringer Ingleheim, Chiesi, Glenmark, Menarini, Mundipharma, Novartis, Pfizer, Pulmatrix, Therevance and Verona, outside the submitted work. Conflict of interest: R.A. Wise reports grant support and medical writing support funded by GlaxoSmithKline, personal fees for data monitoring committee and advisory board work from GlaxoSmithKline, during the conduct of the study; grants and personal fees for data monitoring committee and consultancy work from AstraZeneca/Medimmune and GSK, grants and personal fees for data monitoring and steering committee work from Boehringer Ingelheim, personal fees for clinical end-point committee work from Contrafect, personal fees for data monitoring committee work from Pulmonx, Roche, Merck and AbbVie, personal fees for steering committee work from Spiration, personal fees for workshops and consultancy from Sunovion, grants from Pearl Therapeutics and Sanofi-Aventis, personal fees for consultancy from Circassia, Pneuma, Verona, Denali, Aradigm, Mylan/Theravance and Propelloer Health, personal fees for safety review committee work from Bonti, outside the submitted work. Conflict of interest: S. Lettis reports grant support and medical writing support funded by GlaxoSmithKline, during the conduct of the study; S. Lettis is an employee of and holds shares/options in GlaxoSmithKline, outside the submitted work.
- Published
- 2019
- Full Text
- View/download PDF
46. Blood eosinophils and treatment response with triple and dual combination therapy in chronic obstructive pulmonary disease: analysis of the IMPACT trial.
- Author
-
Pascoe S, Barnes N, Brusselle G, Compton C, Criner GJ, Dransfield MT, Halpin DMG, Han MK, Hartley B, Lange P, Lettis S, Lipson DA, Lomas DA, Martinez FJ, Papi A, Roche N, van der Valk RJP, Wise R, and Singh D
- Subjects
- Administration, Inhalation, Aged, Androstadienes blood, Benzyl Alcohols blood, Bronchodilator Agents blood, Chlorobenzenes blood, Double-Blind Method, Drug Therapy, Combination, Female, Humans, Male, Pulmonary Disease, Chronic Obstructive blood, Quinuclidines blood, Treatment Outcome, Adrenal Cortex Hormones therapeutic use, Androstadienes therapeutic use, Benzyl Alcohols therapeutic use, Bronchodilator Agents therapeutic use, Chlorobenzenes therapeutic use, Eosinophils, Pulmonary Disease, Chronic Obstructive drug therapy, Quinuclidines therapeutic use
- Abstract
Background: Previous studies have highlighted a relationship between reduction in rate of exacerbations with therapies containing inhaled corticosteroids (ICS) and baseline blood eosinophil count in patients with chronic obstructive pulmonary disease (COPD). The IMPACT trial showed that once-daily single-inhaler triple therapy significantly reduced exacerbations versus dual therapies. Blood eosinophil counts and smoking status could be important modifiers of treatment response to ICS. We aimed to model these relationships and their interactions, including outcomes other than exacerbations., Methods: IMPACT was a phase 3, randomised, double-blind, parallel-group, 52-week global study comparing once-daily single-inhaler triple therapy (fluticasone furoate-umeclidinium-vilanterol) with dual inhaled therapy (fluticasone furoate-vilanterol or umeclidinium-vilanterol). Eligible patients had moderate-to-very-severe COPD and at least one moderate or severe exacerbation in the previous year. We used fractional polynomials to model continuous blood eosinophil counts. We used negative binomial regression for numbers of moderate and severe exacerbations, severe exacerbations, and pneumonia. We modelled differences at week 52 in trough FEV
1 , St George's Respiratory Questionnaire (SGRQ) total score, and Transition Dyspnoea Index using repeated measurements mixed effect models. IMPACT was registered with ClinicalTrials.gov, number NCT02164513., Findings: The magnitude of benefit of regimens containing ICS (fluticasone furoate-umeclidinium-vilanterol n=4151 and fluticasone furoate-vilanterol n=4134) in reducing rates of moderate and severe exacerbations increased in proportion with blood eosinophil count, compared with a non-ICS dual long-acting bronchodilator (umeclidinium-vilanterol n=2070). The moderate and severe exacerbation rate ratio for triple therapy versus umeclidinium-vilanterol was 0·88 (95% CI 0·74 to 1·04) at blood eosinophil count less than 90 cells per μL and 0·56 (0·47 to 0·66) at counts of 310 cells per μL or more; the corresponding rate ratio for fluticasone furoate-vilanterol versus umeclidinium-vilanterol was 1·09 (0·91 to 1·29) and 0·56 (0·47 to 0·66), respectively. Similar results were observed for FEV1 , Transition Dyspnoea Index, and SGRQ total score; however, the relationship with FEV1 was less marked. At blood eosinophil counts less than 90 cells per μL and at counts of 310 cells per μL or more, the triple therapy versus umeclidinium-vilanterol treatment difference was 40 mL (95% CI 10 to 70) and 60 mL (20 to 100) for trough FEV1 , -0·01 (-0·68 to 0·66) and 0·30 (-0·37 to 0·97) for Transition Dyspnoea Index score, and -0·01 (-1·81 to 1·78) and -2·78 (-4·64 to -0·92) for SGRQ total score, respectively. Smoking status modified the relationship between observed efficacy and blood eosinophil count for moderate or severe exacerbations, Transition Dyspnoea Index, and FEV1 , with former smokers being more corticosteroid responsive at any eosinophil count than current smokers., Interpretation: This analysis of the IMPACT trial shows that assessment of blood eosinophil count and smoking status has the potential to optimise ICS use in clinical practice in patients with COPD and a history of exacerbations., Funding: GlaxoSmithKline., (Copyright © 2019 Elsevier Ltd. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
47. It's more than low BMI: prevalence of cachexia and associated mortality in COPD.
- Author
-
McDonald MN, Wouters EFM, Rutten E, Casaburi R, Rennard SI, Lomas DA, Bamman M, Celli B, Agusti A, Tal-Singer R, Hersh CP, Dransfield M, and Silverman EK
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Mortality trends, Prevalence, Weight Loss physiology, Body Mass Index, Cachexia diagnosis, Cachexia mortality, Consensus, Pulmonary Disease, Chronic Obstructive diagnosis, Pulmonary Disease, Chronic Obstructive mortality
- Abstract
Background: Cachexia is associated with increased mortality risk among chronic obstructive pulmonary disease (COPD) patients. However, low body mass index (BMI) as opposed to cachexia is often used, particularly when calculating the BODE (BMI, Obstruction, Dyspnea and Exercise) index. For this reason, we examined mortality using a consensus definition and a weight-loss definition of cachexia among COPD cases and compared two new COPD severity indices with BODE., Methods: In the current report, the consensus definition for cachexia incorporated weight-loss > 5% in 12-months or low BMI in addition to 3/5 of decreased muscle strength, fatigue, anorexia, low FFMI and inflammation. The weight-loss definition incorporated weight-loss > 5% or weight-loss > 2% (if low BMI) in 12-months. The low BMI component in BODE was replaced with the consensus definition to create the CODE (Consensus cachexia, Obstruction, Dyspnea and Exercise) index and the weight-loss definition to create the WODE (Weight loss, Obstruction, Dyspnea and Exercise) index. Mortality was assessed using Kaplan-Meier survival and Cox Regression. Performance of models was compared using C-statistics., Results: Among 1483 COPD cases, the prevalences of cachexia by the consensus and weight-loss definitions were 4.7 and 10.4%, respectively. Cachectic patients had a greater than three-fold increased mortality by either the consensus or the weight-loss definition of cachexia independent of BMI and lung function. The CODE index predicted mortality slightly more accurately than the BODE and WODE indices., Conclusions: Cachexia is associated with increased mortality among COPD patients. Monitoring cachexia using weight-loss criteria is relatively simple and predictive of mortality among COPD cases who may be missed if only low BMI is used.
- Published
- 2019
- Full Text
- View/download PDF
48. Common and Rare Variants Genetic Association Analysis of Cigarettes per Day Among Ever-Smokers in Chronic Obstructive Pulmonary Disease Cases and Controls.
- Author
-
Lutz SM, Frederiksen B, Begum F, McDonald MN, Cho MH, Hobbs BD, Parker MM, DeMeo DL, Hersh CP, Ehringer MA, Young K, Jiang L, Foreman MG, Kinney GL, Make BJ, Lomas DA, Bakke P, Gulsvik A, Crapo JD, Silverman EK, Beaty TH, and Hokanson JE
- Subjects
- Adult, Aged, Aged, 80 and over, Aryl Hydrocarbon Hydroxylases genetics, Case-Control Studies, Cytochrome P-450 CYP2B6 genetics, Cytochrome P450 Family 2 genetics, Europe epidemiology, Female, Genome-Wide Association Study methods, Humans, Longitudinal Studies, Male, Middle Aged, Prevalence, Prognosis, Pulmonary Disease, Chronic Obstructive epidemiology, Pulmonary Disease, Chronic Obstructive pathology, Smoking adverse effects, Smoking epidemiology, United States epidemiology, rab4 GTP-Binding Proteins genetics, Ethnicity genetics, Genetic Markers, Genetic Predisposition to Disease, Polymorphism, Single Nucleotide, Pulmonary Disease, Chronic Obstructive etiology, Smokers statistics & numerical data, Smoking genetics
- Abstract
Introduction: Cigarette smoking is a major environmental risk factor for many diseases, including chronic obstructive pulmonary disease (COPD). There are shared genetic influences on cigarette smoking and COPD. Genetic risk factors for cigarette smoking in cohorts enriched for COPD are largely unknown., Methods: We performed genome-wide association analyses for average cigarettes per day (CPD) across the Genetic Epidemiology of COPD (COPDGene) non-Hispanic white (NHW) (n = 6659) and African American (AA) (n = 3260), GenKOLS (the Genetics of Chronic Obstructive Lung Disease) (n = 1671), and ECLIPSE (the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints) (n = 1942) cohorts. In addition, we performed exome array association analyses across the COPDGene NHW and AA cohorts. We considered analyses across the entire cohort and stratified by COPD case-control status., Results: We identified genome-wide significant associations for CPD on chromosome 15q25 across all cohorts (lowest p = 1.78 × 10-15), except in the COPDGene AA cohort alone. Previously reported associations on chromosome 19 had suggestive and directionally consistent associations (RAB4, p = 1.95 × 10-6; CYP2A7, p = 7.50 × 10-5; CYP2B6, p = 4.04 × 10-4). When we stratified by COPD case-control status, single nucleotide polymorphisms on chromosome 15q25 were nominally associated with both NHW COPD cases (β = 0.11, p = 5.58 × 10-4) and controls (β = 0.12, p = 3.86 × 10-5) For the gene-based exome array association analysis of rare variants, there were no exome-wide significant associations. For these previously replicated associations, the most significant results were among COPDGene NHW subjects for CYP2A7 (p = 5.2 × 10-4)., Conclusions: In a large genome-wide association study of both common variants and a gene-based association of rare coding variants in ever-smokers, we found genome-wide significant associations on chromosome 15q25 with CPD for common variants, but not for rare coding variants. These results were directionally consistent among COPD cases and controls., Implications: We examined both common and rare coding variants associated with CPD in a large population of heavy smokers with and without COPD of NHW and AA descent. We replicated genome-wide significant associations on chromosome 15q25 with CPD for common variants among NHW subjects, but not for rare variants. We demonstrated for the first time that common variants on chromosome 15q25 associated with CPD are similar among COPD cases and controls. Previously reported associations on chromosome 19 showed suggestive and directionally consistent associations among common variants (RAB4, CYP2A7, and CYP2B6) and for rare variants (CYP2A7) among COPDGene NHW subjects. Although the genetic effect sizes for these single nucleotide polymorphisms on chromosome 15q25 are modest, we show that this creates a substantial smoking burden over the lifetime of a smoker., (© The Author(s) 2018. Published by Oxford University Press on behalf of the Society for Research on Nicotine and Tobacco. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
49. Genetic landscape of chronic obstructive pulmonary disease identifies heterogeneous cell-type and phenotype associations.
- Author
-
Sakornsakolpat P, Prokopenko D, Lamontagne M, Reeve NF, Guyatt AL, Jackson VE, Shrine N, Qiao D, Bartz TM, Kim DK, Lee MK, Latourelle JC, Li X, Morrow JD, Obeidat M, Wyss AB, Bakke P, Barr RG, Beaty TH, Belinsky SA, Brusselle GG, Crapo JD, de Jong K, DeMeo DL, Fingerlin TE, Gharib SA, Gulsvik A, Hall IP, Hokanson JE, Kim WJ, Lomas DA, London SJ, Meyers DA, O'Connor GT, Rennard SI, Schwartz DA, Sliwinski P, Sparrow D, Strachan DP, Tal-Singer R, Tesfaigzi Y, Vestbo J, Vonk JM, Yim JJ, Zhou X, Bossé Y, Manichaikul A, Lahousse L, Silverman EK, Boezen HM, Wain LV, Tobin MD, Hobbs BD, and Cho MH
- Subjects
- Adult, Aged, Asthma genetics, Case-Control Studies, Female, Gene Expression genetics, Genetic Loci genetics, Genome-Wide Association Study methods, Humans, Lung physiopathology, Male, Middle Aged, Phenotype, Polymorphism, Single Nucleotide genetics, Pulmonary Fibrosis genetics, Smoking genetics, Genetic Predisposition to Disease genetics, Pulmonary Disease, Chronic Obstructive genetics
- Abstract
Chronic obstructive pulmonary disease (COPD) is the leading cause of respiratory mortality worldwide. Genetic risk loci provide new insights into disease pathogenesis. We performed a genome-wide association study in 35,735 cases and 222,076 controls from the UK Biobank and additional studies from the International COPD Genetics Consortium. We identified 82 loci associated with P < 5 × 10
-8 ; 47 of these were previously described in association with either COPD or population-based measures of lung function. Of the remaining 35 new loci, 13 were associated with lung function in 79,055 individuals from the SpiroMeta consortium. Using gene expression and regulation data, we identified functional enrichment of COPD risk loci in lung tissue, smooth muscle, and several lung cell types. We found 14 COPD loci shared with either asthma or pulmonary fibrosis. COPD genetic risk loci clustered into groups based on associations with quantitative imaging features and comorbidities. Our analyses provide further support for the genetic susceptibility and heterogeneity of COPD.- Published
- 2019
- Full Text
- View/download PDF
50. Characterisation of a type II functionally-deficient variant of alpha-1-antitrypsin discovered in the general population.
- Author
-
Laffranchi M, Elliston ELK, Gangemi F, Berardelli R, Lomas DA, Irving JA, and Fra A
- Subjects
- Amino Acid Sequence, Humans, Models, Biological, Molecular Dynamics Simulation, Mutant Proteins chemistry, Mutant Proteins metabolism, Protein Domains, Protein Structure, Secondary, alpha 1-Antitrypsin chemistry, Genetic Variation, Genetics, Population, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin metabolism
- Abstract
Lung disease in alpha-1-antitrypsin deficiency (AATD) results from dysregulated proteolytic activity, mainly by neutrophil elastase (HNE), in the lung parenchyma. This is the result of a substantial reduction of circulating alpha-1-antitrypsin (AAT) and the presence in the plasma of inactive polymers of AAT. Moreover, some AAT mutants have reduced intrinsic activity toward HNE, as demonstrated for the common Z mutant, as well as for other rarer variants. Here we report the identification and characterisation of the novel AAT reactive centre loop variant Gly349Arg (p.G373R) present in the ExAC database. This AAT variant is secreted at normal levels in cellular models of AATD but shows a severe reduction in anti-HNE activity. Biochemical and molecular dynamics studies suggest it exhibits unfavourable RCL presentation to cognate proteases and compromised insertion of the RCL into β-sheet A. Identification of a fully dysfunctional AAT mutant that does not show a secretory defect underlines the importance of accurate genotyping of patients with pulmonary AATD manifestations regardless of the presence of normal levels of AAT in the circulation. This subtype of disease is reminiscent of dysfunctional phenotypes in anti-thrombin and C1-inibitor deficiencies so, accordingly, we classify this variant as the first pure functionally-deficient (type II) AATD mutant., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.