Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where John G. Matthews is active.

Publication


Featured researches published by John G. Matthews.


The New England Journal of Medicine | 2011

Lebrikizumab Treatment in Adults with Asthma

Jonathan Corren; Robert F. Lemanske; Nicola A. Hanania; Phillip E. Korenblat; Merdad V. Parsey; Joseph R. Arron; Jeffrey M. Harris; Heleen Scheerens; Lawren C. Wu; Zheng Su; Sofia Mosesova; Mark D. Eisner; Sean P. Bohen; John G. Matthews

BACKGROUND Many patients with asthma have uncontrolled disease despite treatment with inhaled glucocorticoids. One potential cause of the variability in response to treatment is heterogeneity in the role of interleukin-13 expression in the clinical asthma phenotype. We hypothesized that anti-interleukin-13 therapy would benefit patients with asthma who had a pretreatment profile consistent with interleukin-13 activity. METHODS We conducted a randomized, double-blind, placebo-controlled study of lebrikizumab, a monoclonal antibody to interleukin-13, in 219 adults who had asthma that was inadequately controlled despite inhaled glucocorticoid therapy. The primary efficacy outcome was the relative change in prebronchodilator forced expiratory volume in 1 second (FEV(1)) from baseline to week 12. Among the secondary outcomes was the rate of asthma exacerbations through 24 weeks. Patient subgroups were prespecified according to baseline type 2 helper T-cell (Th2) status (assessed on the basis of total IgE level and blood eosinophil count) and serum periostin level. RESULTS At baseline, patients had a mean FEV(1) that was 65% of the predicted value and were taking a mean dose of inhaled glucocorticoids of 580 μg per day; 80% were also taking a long-acting beta-agonist. At week 12, the mean increase in FEV(1) was 5.5 percentage points higher in the lebrikizumab group than in the placebo group (P = 0.02). Among patients in the high-periostin subgroup, the increase from baseline FEV(1) was 8.2 percentage points higher in the lebrikizumab group than in the placebo group (P = 0.03). Among patients in the low-periostin subgroup, the increase from baseline FEV(1) was 1.6 percentage points higher in the lebrikizumab group than in the placebo group (P = 0.61). Musculoskeletal side effects were more common with lebrikizumab than with placebo (13.2% vs. 5.4%, P = 0.045). CONCLUSIONS Lebrikizumab treatment was associated with improved lung function. Patients with high pretreatment levels of serum periostin had greater improvement in lung function with lebrikizumab than did patients with low periostin levels. (Funded by Genentech; ClinicalTrials.gov number, NCT00930163 .).


Thorax | 2015

Lebrikizumab in moderate-to-severe asthma: pooled data from two randomised placebo-controlled studies

Nicola A. Hanania; Michael Noonan; Jonathan Corren; Phillip E. Korenblat; Yanan Zheng; Saloumeh Kadkhodayan Fischer; Melissa Cheu; Wendy S. Putnam; Elaine Murray; Heleen Scheerens; Cecile Holweg; Romeo Maciuca; Sarah Gray; Ramona Doyle; Dana McClintock; Julie Olsson; John G. Matthews; Karl Yen

Introduction In a subset of patients with asthma, standard-of-care treatment does not achieve disease control, highlighting the need for novel therapeutic approaches. Lebrikizumab is a humanised, monoclonal antibody that binds to and blocks interleukin-13 activity. Methods LUTE and VERSE were replicate, randomised, double-blind, placebo-controlled studies, evaluating multiple doses of lebrikizumab in patients with uncontrolled asthma despite the use of medium-to-high-dose inhaled corticosteroid and a second controller. Patients received lebrikizumab 37.5, 125, 250 mg or placebo subcutaneously every four weeks. The primary endpoint was the rate of asthma exacerbations during the placebo-controlled period. Analyses were performed on prespecified subgroups based on baseline serum periostin levels. Following the discovery of a host-cell impurity in the study drug material, protocols were amended to convert from phase III to phase IIb. Subsequently, dosing of study medication was discontinued early as a precautionary measure. The data collected for analysis were from a placebo-controlled period of variable duration and pooled across both studies. Results The median duration of treatment was approximately 24 weeks. Treatment with lebrikizumab reduced the rate of asthma exacerbations, which was more pronounced in the periostin-high patients (all doses: 60% reduction) than in the periostin-low patients (all doses: 5% reduction); no dose–response was evident. Lung function also improved following lebrikizumab treatment, with greatest increase in FEV1 in periostin-high patients (all doses: 9.1% placebo-adjusted improvement) compared with periostin-low patients (all doses: 2.6% placebo-adjusted improvement). Lebrikizumab was well tolerated and no clinically important safety signals were observed. Conclusions These data are consistent with, and extend, previously published results demonstrating the efficacy of lebrikizumab in improving rate of asthma exacerbations and lung function in patients with moderate-to-severe asthma who remain uncontrolled despite current standard-of-care treatment. Trial registration numbers The LUTE study was registered under NCT01545440 and the VERSE study under NCT01545453 at http://www.clinicaltrials.gov


The Lancet Respiratory Medicine | 2016

Efficacy and safety of lebrikizumab in patients with uncontrolled asthma (LAVOLTA I and LAVOLTA II): replicate, phase 3, randomised, double-blind, placebo-controlled trials

Nicola A. Hanania; Phillip E. Korenblat; Kenneth R. Chapman; Eric D. Bateman; Petr Kopecky; Pierluigi Paggiaro; Akihito Yokoyama; Julie Olsson; Sarah Gray; Cecile Holweg; Mark D. Eisner; Charles Asare; Saloumeh Kadkhodayan Fischer; Kun Peng; Wendy S. Putnam; John G. Matthews

BACKGROUND In phase 2 trials, lebrikizumab, an anti-interleukin-13 monoclonal antibody, reduced exacerbation rates and improved FEV1 in patients with uncontrolled asthma, particularly in those with high concentrations of type 2 biomarkers (eg, periostin or blood eosinophils). We undertook replicate phase 3 studies to assess the efficacy and safety of lebrikizumab in patients with uncontrolled asthma despite inhaled corticosteroids and at least one second controller medication. METHODS Adult patients with uncontrolled asthma, pre-bronchodilator FEV1 40-80% predicted, and stable background therapy were randomly assigned (1:1:1) with an interactive voice-web-based response system to receive lebrikizumab 37·5 mg or 125 mg, or placebo subcutaneously, once every 4 weeks. Randomisation was stratified by screening serum periostin concentration, history of asthma exacerbations within the last 12 months, baseline asthma medications, and country. The primary efficacy endpoint was the rate of asthma exacerbations over 52 weeks in biomarker-high patients (periostin ≥50 ng/mL or blood eosinophils ≥300 cells per μL), analysed with a Poisson regression model corrected for overdispersion with Pearson χ2 that included terms for treatment group, number of asthma exacerbations within the 12 months before study entry, baseline asthma medications, geographic region, screening periostin concentration, and blood eosinophil counts as covariates. Both trials are registered at ClinicalTrials.gov, LAVOLTA I, number NCT01867125, and LAVOLTA II, number NCT01868061. FINDINGS 1081 patients were treated in LAVOLTA I and 1067 patients in LAVOLTA II. Over 52 weeks, lebrikizumab reduced exacerbation rates in biomarker-high patients in the 37·5 mg dose group (rate ratio [RR] 0·49 [95% CI 0·34-0·69], p<0·0001) and in the 125 mg dose group (RR 0·70 [0·51-0·95], p=0·0232) versus placebo in LAVOLTA I. Exacerbation rates were also reduced in biomarker-high patients in both dose groups versus placebo in LAVOLTA II (37·5 mg: RR 0·74 [95% CI 0·54-1·01], p=0·0609; 125 mg: RR 0·74 [0·54-1·02], p=0·0626). Pooling both studies, the proportion of patients who experienced treatment-emergent adverse events (79% [1125 of 1432 patients] for both lebrikizumab doses vs 80% [576 of 716 patients] for placebo), serious adverse events (8% [115 patients] for both lebrikizumab doses vs 9% [65 patients] for placebo), and adverse events leading to study drug discontinuation (3% [49 patients] for both lebrikizumab doses vs 4% [31 patients] for placebo) were similar between lebrikizumab and placebo. The following serious adverse events were reported in the placebo-controlled period: one event of aplastic anaemia and five serious adverse events related to raised concentrations of eosinophils in patients treated with lebrikizumab and one event of eosinophilic pneumonia in the placebo group. INTERPRETATION Lebrikizumab did not consistently show significant reduction in asthma exacerbations in biomarker-high patients. However, it blocked interleukin-13 as evidenced by the effect on interleukin-13-related pharmacodynamic biomarkers, and clinically relevant changes could not be ruled out. FUNDING F Hoffmann-La Roche.


European Respiratory Journal | 2015

Clinical and inflammatory characteristics of the European U-BIOPRED adult severe asthma cohort

Dominick Shaw; Ana R. Sousa; Stephen J. Fowler; Louise Fleming; Graham Roberts; Julie Corfield; Ioannis Pandis; Aruna T. Bansal; Elisabeth H. Bel; Charles Auffray; Chris Compton; Hans Bisgaard; Enrica Bucchioni; Massimo Caruso; Pascal Chanez; Barbro Dahlén; Sven Erik Dahlén; Kerry Dyson; Urs Frey; Thomas Geiser; Maria Gerhardsson de Verdier; David Gibeon; Yike Guo; Simone Hashimoto; Gunilla Hedlin; Elizabeth Jeyasingham; Pieter Paul W Hekking; Tim Higenbottam; Ildiko Horvath; Alan J. Knox

U-BIOPRED is a European Union consortium of 20 academic institutions, 11 pharmaceutical companies and six patient organisations with the objective of improving the understanding of asthma disease mechanisms using a systems biology approach. This cross-sectional assessment of adults with severe asthma, mild/moderate asthma and healthy controls from 11 European countries consisted of analyses of patient-reported outcomes, lung function, blood and airway inflammatory measurements. Patients with severe asthma (nonsmokers, n=311; smokers/ex-smokers, n=110) had more symptoms and exacerbations compared to patients with mild/moderate disease (n=88) (2.5 exacerbations versus 0.4 in the preceding 12 months; p<0.001), with worse quality of life, and higher levels of anxiety and depression. They also had a higher incidence of nasal polyps and gastro-oesophageal reflux with lower lung function. Sputum eosinophil count was higher in severe asthma compared to mild/moderate asthma (median count 2.99% versus 1.05%; p=0.004) despite treatment with higher doses of inhaled and/or oral corticosteroids. Consistent with other severe asthma cohorts, U-BIOPRED is characterised by poor symptom control, increased comorbidity and airway inflammation, despite high levels of treatment. It is well suited to identify asthma phenotypes using the array of “omic” datasets that are at the core of this systems medicine approach. Severe asthma results in more airway inflammation, worse symptoms and lower lung function, despite increased therapy http://ow.ly/QznR3


The Journal of Allergy and Clinical Immunology | 2013

Dose-ranging study of lebrikizumab in asthmatic patients not receiving inhaled steroids

Michael Noonan; Phillip E. Korenblat; Sofia Mosesova; Heleen Scheerens; Joseph R. Arron; Yanan Zheng; Wendy S. Putnam; Merdad V. Parsey; Sean P. Bohen; John G. Matthews

BACKGROUND Asthma is a disease with marked heterogeneity in its clinical course and response to treatment. IL-13 is central to type 2 inflammation, which contributes to many key features of asthma. Lebrikizumab is an anti-IL-13 mAb previously reported to significantly improve lung function in patients with inadequately controlled asthma despite inhaled corticosteroid therapy, especially in periostin-high patients. OBJECTIVE This phase II study investigated the efficacy and safety of IL-13 blockade with different doses of lebrikizumab in asthmatic patients not receiving inhaled corticosteroids. METHODS Patients were randomized to receive 125, 250, or 500 mg of lebrikizumab or placebo subcutaneously monthly for 12 weeks with an 8-week follow-up period. The primary efficacy end point was the relative change in prebronchodilator FEV1 from baseline to week 12. RESULTS A total of 212 patients were randomized. The mean relative change in FEV1 was numerically higher in all lebrikizumab dose groups versus the placebo group, although the difference was neither statistically nor clinically significant. There were no meaningful differences in changes in FEV1 between the dose groups and the placebo group by the periostin subgroup. Lebrikizumab treatment was associated with a reduced risk of treatment failure at all doses versus placebo (P < .001), and results were similar by the periostin subgroup, with no apparent differences between doses of lebrikizumab. Lebrikizumab was generally well tolerated. CONCLUSION Blocking IL-13, a single cytokine, in this population of asthmatic patients is insufficient to improve lung function. There is evidence that IL-13 blockade may improve disease control, as measured by prevention of protocol-defined treatment failure in these patients.


Clinical & Experimental Allergy | 2014

The effects of lebrikizumab in patients with mild asthma following whole lung allergen challenge.

Heleen Scheerens; Joseph R. Arron; Yanan Zheng; Wendy S. Putnam; Richard W. Erickson; David F. Choy; Jeffrey M. Harris; June H. Lee; Nizar N. Jarjour; John G. Matthews

Interleukin 13 (IL13) is a T‐helper type 2 (Th2) cytokine associated with inflammation and pathology in allergic diseases such as bronchial asthma. We have shown that treatment with lebrikizumab, an anti‐IL13 monoclonal antibody, significantly improves prebronchodilator forced expiratory volume in 1 s (FEV1) in a subset of subjects with uncontrolled asthma.


Science Translational Medicine | 2014

Targeting membrane-expressed IgE B cell receptor with an antibody to the M1 prime epitope reduces IgE production

Gail M. Gauvreau; Jeffrey M. Harris; Louis Philippe Boulet; Heleen Scheerens; J. M. FitzGerald; W. Putnam; Donald W. Cockcroft; Beth E. Davis; Richard Leigh; Zheng Y; Barbro Dahlén; Wang Y; Romeo Maciuca; Irvin Mayers; Liao Xc; Lawren C. Wu; John G. Matthews; Paul M. O'Byrne

A humanized monoclonal antibody that targets the membrane IgE B cell receptor was associated with improvements in asthmatic airway responses. Asthma Antibody Clears the Air A humanized antibody may be the key for treating allergic asthma. Gauvreau et al. report that quilizumab—a humanized monoclonal antibody that targets the membrane immunoglobulin E (IgE) B cell receptor—led to reductions in total and allergen-specific serum IgE that were sustained for 6 months after dosing and were associated with improvements in allergen-induced early and late asthmatic airway responses. A monoclonal antibody that neutralizes IgE can effectively treat asthma in the clinic; however, use is limited by dosing restrictions and it does not prevent new IgE production. Inhibiting IgE production by targeting membrane IgE–expressing cells may enable more subjects to be treated and less frequent dosing, and has the potential for sustained effects upon the cessation of therapy. Elevated serum levels of both total and allergen-specific immunoglobulin E (IgE) correlate with atopic diseases such as allergic rhinitis and allergic asthma. Neutralization of IgE by anti-IgE antibodies can effectively treat allergic asthma. Preclinical studies indicate that targeting membrane IgE–positive cells with antibodies against M1 prime can inhibit the production of new IgE and significantly reduce the levels of serum IgE. We report results from two trials that investigated the safety, pharmacokinetics, and activity of quilizumab, a humanized monoclonal antibody targeting specifically the M1 prime epitope of membrane IgE, in subjects with allergic rhinitis (NCT01160861) or mild allergic asthma (NCT01196039). In both studies, quilizumab treatment was well tolerated and led to reductions in total and allergen-specific serum IgE that lasted for at least 6 months after the cessation of dosing. In subjects with allergic asthma who were subjected to an allergen challenge, quilizumab treatment blocked the generation of new IgE, reduced allergen-induced early and late asthmatic airway responses by 26 and 36%, respectively, and reduced allergen-induced increases in sputum eosinophils by ~50% compared with placebo. These studies indicate that targeting of membrane IgE–expressing cells with anti-M1 prime antibodies can prevent IgE production in humans.


Clinical & Experimental Allergy | 2014

OX40L blockade and allergen‐induced airway responses in subjects with mild asthma

Gail M. Gauvreau; Louis Philippe Boulet; Donald W. Cockcroft; J. M. FitzGerald; Irvin Mayers; Chris Carlsten; M. Laviolette; Kieran J. Killian; Beth E. Davis; Mark Larche; C. Kipling; Benny Dua; S. Mosesova; W. Putnam; Zheng Y; Heleen Scheerens; D. McClintock; John G. Matthews; Paul M. O'Byrne

The OX40/OX40L interaction contributes to an optimal T cell response following allergic stimuli and plays an important role in the maintenance and reactivation of memory T effector cells.


American Journal of Respiratory and Critical Care Medicine | 2016

Roles of Periostin in Respiratory Disorders

Kenji Izuhara; Simon J. Conway; Bethany B. Moore; Hisako Matsumoto; Cecile Holweg; John G. Matthews; Joseph R. Arron

Periostin is a matricellular protein that has been implicated in many disease states. It interacts with multiple signaling cascades to modulate the expression of downstream genes that regulate cellular interactions within the extracellular matrix. This review focuses on the role of periostin in respiratory diseases, including asthma and idiopathic pulmonary fibrosis, and its potential to help guide treatment or assess prognosis. Epithelial injury is a common feature of many respiratory diseases, resulting in the secretion, among others, of periostin, which is subsequently involved in airway remodeling and other aspects of pulmonary pathophysiology. In asthma, periostin is recognized as a biomarker of type 2 inflammation; POSTN gene expression is up-regulated in bronchial epithelial cells by IL-13 and IL-4. Serum periostin has been evaluated for the identification of patients with increased clinical benefit from treatment with anti-IL-13 (lebrikizumab, tralokinumab) and anti-IgE (omalizumab) therapy and may be prognostic for increased risk of asthma exacerbations and progressive lung function decline. Furthermore, in asthma, periostin may regulate subepithelial fibrosis and mucus production and may serve as a systemic biomarker of eosinophilic airway inflammation. Periostin is also highly expressed in the lungs of patients with idiopathic pulmonary fibrosis, and its serum levels may predict clinical progression. Overall, periostin contributes to multiple pathogenic processes across respiratory diseases, and peripheral blood levels of periostin may have utility as a biomarker of treatment response and disease progression.


Thorax | 2016

Research in progress: Medical Research Council United Kingdom Refractory Asthma Stratification Programme (RASP-UK)

Liam Heaney; Ratko Djukanovic; Ashley Woodcock; Samantha Walker; John G. Matthews; Ian D. Pavord; Peter Bradding; Robert Niven; Christopher E. Brightling; Rekha Chaudhuri; Joseph R. Arron; David F. Choy; Douglas C. Cowan; Adel Mansur; Andrew Menzies-Gow; Ian M. Adcock; Kian Fan Chung; Christopher Corrigan; Peter Coyle; Sebastian L. Johnston; Peter H. Howarth; James Lordan; Ian Sabroe; Jeannette Bigler; Dirk E. Smith; Matthew Catley; Richard May; Lisa Pierre; Christopher S. Stevenson; Glenn Crater

The UK Refractory Asthma Stratification Programme (RASP-UK) will explore novel biomarker stratification strategies in severe asthma to improve clinical management and accelerate development of new therapies. Prior asthma mechanistic studies have not stratified on inflammatory phenotype and the understanding of pathophysiological mechanisms in asthma without Type 2 cytokine inflammation is limited. RASP-UK will objectively assess adherence to corticosteroids (CS) and examine a novel composite biomarker strategy to optimise CS dose; this will also address what proportion of patients with severe asthma have persistent symptoms without eosinophilic airways inflammation after progressive CS withdrawal. There will be interactive partnership with the pharmaceutical industry to facilitate access to stratified populations for novel therapeutic studies.

Collaboration


Dive into the John G. Matthews's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicola A. Hanania

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian M. Adcock

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge