Network


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

Hotspot


Dive into the research topics where Jeffrey M. Harris is active.

Publication


Featured researches published by Jeffrey M. Harris.


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 .).


American Journal of Respiratory and Critical Care Medicine | 2013

Exploring the Effects of Omalizumab in Allergic Asthma

Nicola A. Hanania; Sally E. Wenzel; Karin Rosén; Hsin-Ju Hsieh; Sofia Mosesova; David F. Choy; Preeti Lal; Joseph R. Arron; Jeffrey M. Harris; William W. Busse

RATIONALE For many patients with asthma, allergic airway inflammation is primarily a Th2-weighted process; however, heterogeneity in patterns of inflammation suggests phenotypic distinctions exist that influence disease presentation and treatment effects. OBJECTIVES To assess the potential of fractional exhaled nitric oxide (FE(NO)), peripheral blood eosinophil count, and serum periostin as biomarkers of Th2 inflammation and predictors of treatment effects of omalizumab. METHODS The EXTRA omalizumab study enrolled patients (aged 12-75 yr) with uncontrolled severe persistent allergic asthma. Analyses were performed evaluating treatment effects in relation to FE(NO), blood eosinophils, and serum periostin at baseline. Patients were divided into low- and high-biomarker subgroups. Treatment effects were evaluated as number of protocol-defined asthma exacerbations during the 48-week treatment period (primary endpoint). MEASUREMENTS AND MAIN RESULTS A total of 850 patients were enrolled. Data were available from 394 (46.4%), 797 (93.8%), and 534 (62.8%) patients for FE(NO), blood eosinophils, and serum periostin, respectively. After 48 weeks of omalizumab, reductions in protocol-defined exacerbations were greater in high versus low subgroups for all three biomarkers: FE(NO), 53% (95% confidence interval [CI], 37-70; P = 0.001) versus 16% (95% CI, -32 to 46; P = 0.45); eosinophils, 32% (95% CI, 11-48; P = 0.005) versus 9% (95% CI, -24 to 34; P = 0.54); and periostin, 30% (95% CI, -2 to 51; P = 0.07) versus 3% (95% CI, -43 to 32; P = 0.94). CONCLUSIONS The difference in exacerbation frequency between omalizumab and placebo was greatest in the three high-biomarker subgroups, probably associated with the greater risk for exacerbations in high subgroups. Additional studies are required to explore the value of these biomarkers in clinical practice. Clinical trial registered with www.clinicaltrials.gov (NCT00314574).


The Journal of Allergy and Clinical Immunology | 2015

The airway microbiome in patients with severe asthma: Associations with disease features and severity

Yvonne J. Huang; Snehal Nariya; Jeffrey M. Harris; Susan V. Lynch; David F. Choy; Joseph R. Arron; Homer A. Boushey

BACKGROUND Asthma is heterogeneous, and airway dysbiosis is associated with clinical features in patients with mild-to-moderate asthma. Whether similar relationships exist among patients with severe asthma is unknown. OBJECTIVE We sought to evaluate relationships between the bronchial microbiome and features of severe asthma. METHODS Bronchial brushings from 40 participants in the Bronchoscopic Exploratory Research Study of Biomarkers in Corticosteroid-refractory Asthma (BOBCAT) study were evaluated by using 16S ribosomal RNA-based methods. Relationships to clinical and inflammatory features were analyzed among microbiome-profiled subjects. Secondarily, bacterial compositional profiles were compared between patients with severe asthma and previously studied healthy control subjects (n = 7) and patients with mild-to-moderate asthma (n = 41). RESULTS In patients with severe asthma, bronchial bacterial composition was associated with several disease-related features, including body mass index (P < .05, Bray-Curtis distance-based permutational multivariate analysis of variance; PERMANOVA), changes in Asthma Control Questionnaire (ACQ) scores (P < .01), sputum total leukocyte values (P = .06), and bronchial biopsy eosinophil values (per square millimeter, P = .07). Bacterial communities associated with worsening ACQ scores and sputum total leukocyte values (predominantly Proteobacteria) differed markedly from those associated with body mass index (Bacteroidetes/Firmicutes). In contrast, improving/stable ACQ scores and bronchial epithelial gene expression of FK506 binding protein (FKBP5), an indicator of steroid responsiveness, correlated with Actinobacteria. Mostly negative correlations were observed between biopsy eosinophil values and Proteobacteria. No taxa were associated with a TH2-related epithelial gene expression signature, but expression of TH17-related genes was associated with Proteobacteria. Patients with severe asthma compared with healthy control subjects or patients with mild-to-moderate asthma were significantly enriched in Actinobacteria, although the largest differences observed involved a Klebsiella genus member (7.8-fold increase in patients with severe asthma, adjusted P < .001). CONCLUSIONS Specific microbiota are associated with and may modulate inflammatory processes in patients with severe asthma and related phenotypes. Airway dysbiosis in patients with severe asthma appears to differ from that observed in those with milder asthma in the setting of inhaled corticosteroid use.


The Journal of Allergy and Clinical Immunology | 2015

Asthma and lower airway diseaseThe airway microbiome in patients with severe asthma: Associations with disease features and severity

Yvonne J. Huang; Snehal Nariya; Jeffrey M. Harris; Susan V. Lynch; David F. Choy; Joseph R. Arron; Homer A. Boushey

BACKGROUND Asthma is heterogeneous, and airway dysbiosis is associated with clinical features in patients with mild-to-moderate asthma. Whether similar relationships exist among patients with severe asthma is unknown. OBJECTIVE We sought to evaluate relationships between the bronchial microbiome and features of severe asthma. METHODS Bronchial brushings from 40 participants in the Bronchoscopic Exploratory Research Study of Biomarkers in Corticosteroid-refractory Asthma (BOBCAT) study were evaluated by using 16S ribosomal RNA-based methods. Relationships to clinical and inflammatory features were analyzed among microbiome-profiled subjects. Secondarily, bacterial compositional profiles were compared between patients with severe asthma and previously studied healthy control subjects (n = 7) and patients with mild-to-moderate asthma (n = 41). RESULTS In patients with severe asthma, bronchial bacterial composition was associated with several disease-related features, including body mass index (P < .05, Bray-Curtis distance-based permutational multivariate analysis of variance; PERMANOVA), changes in Asthma Control Questionnaire (ACQ) scores (P < .01), sputum total leukocyte values (P = .06), and bronchial biopsy eosinophil values (per square millimeter, P = .07). Bacterial communities associated with worsening ACQ scores and sputum total leukocyte values (predominantly Proteobacteria) differed markedly from those associated with body mass index (Bacteroidetes/Firmicutes). In contrast, improving/stable ACQ scores and bronchial epithelial gene expression of FK506 binding protein (FKBP5), an indicator of steroid responsiveness, correlated with Actinobacteria. Mostly negative correlations were observed between biopsy eosinophil values and Proteobacteria. No taxa were associated with a TH2-related epithelial gene expression signature, but expression of TH17-related genes was associated with Proteobacteria. Patients with severe asthma compared with healthy control subjects or patients with mild-to-moderate asthma were significantly enriched in Actinobacteria, although the largest differences observed involved a Klebsiella genus member (7.8-fold increase in patients with severe asthma, adjusted P < .001). CONCLUSIONS Specific microbiota are associated with and may modulate inflammatory processes in patients with severe asthma and related phenotypes. Airway dysbiosis in patients with severe asthma appears to differ from that observed in those with milder asthma in the setting of inhaled corticosteroid use.


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.


The American Journal of Gastroenterology | 2013

EMerging BiomARKers in Inflammatory Bowel Disease (EMBARK) Study Identifies Fecal Calprotectin, Serum MMP9, and Serum IL-22 as a Novel Combination of Biomarkers for Crohn's Disease Activity: Role of Cross-Sectional Imaging

William A. Faubion; Joel G. Fletcher; Sharon O'Byrne; Brian G. Feagan; Willem J. de Villiers; Bruce Salzberg; Scott E. Plevy; Deborah D. Proctor; John F. Valentine; Peter D. Higgins; Jeffrey M. Harris; Lauri Diehl; Lilyan Wright; Gaik Wei Tew; Diana Luca; Karen Basu; Mary E. Keir

Objectives:In Crohns disease (CD), clinical symptoms correspond poorly to inflammatory disease activity. Biomarkers reflective of mucosal and bowel wall inflammation would be useful to monitor disease activity. The EMBARK study evaluated disease activity in patients with ulcerative colitis (UC) and CD, and used endoscopy with or without cross-sectional imaging for biomarker discovery.Methods:UC (n=107) and CD (n=157) patients were characterized and underwent ileocolonoscopy (ICO). A subset of CD patients (n=66) also underwent computed tomography enterography (CTE). ICO and CTE were scored by a gastroenterologist and radiologist who incorporated findings of inflammation into a single score (ICO-CTE) for patients that underwent both procedures. Serum and fecal biomarkers were evaluated for association with the Mayo Clinic endoscopy score in UC patients and with ICO alone or ICO-CTE in CD patients. Individual biomarkers with a moderate degree of correlation (P≤0.3) were evaluated using multivariate analysis with model selection using a stepwise procedure.Results:In UC, ordinal logistic regression using Mayo Clinic endoscopy subscore selected the combination of fecal calprotectin and serum matrix metalloproteinase 9 (MMP9; pseudo R2=0.353). In CD, we found that use of the ICO-CTE increased specificity of known biomarkers. Using ICO-CTE as the dependent variable for biomarker discovery, the selected biomarkers were the combination of fecal calprotectin, serum MMP9, and serum IL-22 (r=0.699).Conclusions:Incorporation of both ICO and CTE into a single measure increased biomarker performance in CD. Combinations of fecal calprotectin and serum MMP9 for UC, and combinations of fecal calprotectin, serum MMP9, and serum interleukin-22 in CD, demonstrated the strongest association with imaging/endoscopy-defined inflammation.


Analytica Chimica Acta | 1986

Flow injection analysis (FIA) — a personal view

R. Appelqvist; G.R. Beecher; G. Den Boef; J. Emnéus; Zhaolun Fang; L. Gorton; E.H. Hansen; P.E. Hare; Jeffrey M. Harris; J.J. Harrow; N. Ishibashi; J. Janata; G. Johansson; Bo Karlberg; F.J. Krug; W.E. van der Linden; M. D. Luque de Castro; G. Markovarga; James N. Miller; Horacio A. Mottola; H. Müller; G.E. Pacey; Craig Riley; J. Růžička; R.C. Schothorst; K.K. Stewart; Alan Townshend; J. F. Tyson; K. Ueno; Miguel Valcárcel

Abstract 36 authors from 10 countries express their viewpoints on the merits of flow injection analysis. Alison Macdonald has contributed invaluably to the development of FIA through critical evaluation of work of all the above authors, by editing manuscripts, and by carefully resolving differences between authors and referees. Therefore Analytica Chimica Acta, together with its Editor, may be regarded as the Alma Mater of FIA. It is our intention to honour (and humour). Dr. Macdonald by publishing this nonorchestrated and, by us, unedited patchwork of individual papers which reflect a rich mixture of applications, modifications and interpretations of FIA, together with a variety of personal opinions. We are most grateful to our colleagues for endorsing this unconventional way of celebrating the 25-year anniversary of our Editor (J.R. and E.H.H.).


European Respiratory Journal | 2014

Disconnect between sputum neutrophils and other measures of airway inflammation in asthma

Joseph R. Arron; David F. Choy; Michel Laviolette; Steven G. Kelsen; Ammar Hatab; Richard Leigh; Neil C. Thomson; Eugene R. Bleecker; Ron Olivenstein; Pedro C. Avila; Nizar N. Jarjour; Mario Castro; Gail M. Gauvreau; James T. Good; Joel N. Kline; Adel Mansur; Irvin Mayers; Liam Heaney; Qutayba Hamid; Jeffrey M. Harris

To the Editor: Asthma heterogeneity has been described by the nature and intensity of granulocytic infiltration into the airways [1, 2]. Sputum, endobronchial biopsies and bronchoalveolar lavage (BAL) sample different anatomical regions of the airways. Few studies have directly compared the inflammatory cell infiltrates in these regions within a large cohort of moderate–severe asthma patients using standard techniques. In the BOBCAT (Bronchoscopic exploratory research study Of Biomarkers in Corticosteroid-refractory AsThma) study, we sampled multiple airway compartments concurrently, enabling us to evaluate relationships between granulocytic infiltrates within and between compartments in a large cohort of moderate–severe adult asthma patients. This prospective multicentre observational study was conducted in four visits over a 4–6 week period, as described previously [3]. The patients included had moderate–severe persistent asthma (forced expiratory volume in 1 s (FEV1) 40–80% predicted and Asthma Control Questionnaire (ACQ) [4] score >1.5) and, within the past 5 years, evidence of >12% post-bronchodilator reversibility or a provocative concentration of methacholine causing a 20% decline in FEV1 ≤8 mg·mL−1 despite high-dose inhaled corticosteroid (ICS) (≥1000 μg·day−1 fluticasone propionate equivalent) with or without long-acting β2-adrenergic agonist therapy. Key exclusion criteria included initiation or increase in systemic steroid use 30 days prior to screening, chronic or recent (within the past 30 days) use of immunosuppressive therapies, or other active lung disease. Patients had a prior established diagnosis of moderate–severe asthma for ≥6 months prior to screening while receiving a stable dose regimen (>6 weeks) of a high-dose ICS. Allowed concomitant medications included leukotriene receptor antagonists and oral corticosteroids. 78 patients with confirmed moderate–severe asthma were enrolled at 18 sites; 67 (86%) completed the study. All patients had persistently impaired …


Antimicrobial Agents and Chemotherapy | 2017

Phase 2 Randomized Trial of the Safety and Efficacy of MHAA4549A, a Broadly Neutralizing Monoclonal Antibody, in a Human Influenza A Virus Challenge Model

Jacqueline McBride; Jeremy J. Lim; Tracy Burgess; Rong Deng; Michael A. Derby; Mauricio Maia; Priscilla Horn; Omer Siddiqui; Daniel Sheinson; Haiyin Chen-Harris; Elizabeth Newton; Dimitri Fillos; Denise Nazzal; Carrie M. Rosenberger; Maikke B. Ohlson; Rob Lambkin-Williams; Hosnieh Fathi; Jeffrey M. Harris; Jorge A. Tavel

ABSTRACT MHAA4549A, a human monoclonal antibody targeting the hemagglutinin stalk region of influenza A virus (IAV), is being developed as a therapeutic for patients hospitalized with severe IAV infection. The safety and efficacy of MHAA4549A were assessed in a randomized, double-blind, placebo-controlled, dose-ranging study in a human IAV challenge model. One hundred healthy volunteers were inoculated with A/Wisconsin/67/2005 (H3N2) IAV and, 24 to 36 h later, administered a single intravenous dose of either placebo, MHAA4549A (400, 1,200, or 3,600 mg), or a standard oral dose of oseltamivir. Subjects were assessed for safety, pharmacokinetics (PK), and immunogenicity. The intent-to-treat-infected (ITTI) population was assessed for changes in viral load, influenza symptoms, and inflammatory biomarkers. MHAA4549A was well tolerated in all IAV challenge subjects. The 3,600-mg dose of MHAA4549A significantly reduced the viral burden relative to that of the placebo as determined by the area under the curve (AUC) of nasopharyngeal virus infection, quantified using quantitative PCR (98%) and 50% tissue culture infective dose (TCID50) (100%) assays. Peak viral load, duration of viral shedding, influenza symptom scores, mucus weight, and inflammatory biomarkers were also reduced. Serum PK was linear with a half-life of ∼23 days. No MHAA4549A-treated subjects developed anti-drug antibodies. In conclusion, MHAA4549A was well tolerated and demonstrated statistically significant and substantial antiviral activity in an IAV challenge model. (This study has been registered at ClinicalTrials.gov under identifier NCT01980966.)

Collaboration


Dive into the Jeffrey M. Harris's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge