Network


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

Hotspot


Dive into the research topics where Lawren C. Wu is active.

Publication


Featured researches published by Lawren C. Wu.


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 .).


Journal of Clinical Investigation | 2007

In vivo blockade of OX40 ligand inhibits thymic stromal lymphopoietin driven atopic inflammation

Dhaya Seshasayee; Wyne P. Lee; Meijuan Zhou; Jean Shu; Eric Suto; Juan Zhang; Laurie Diehl; Cary D. Austin; Y. Gloria Meng; Martha Tan; Sherron Bullens; Stefan Seeber; Maria E. Fuentes; Aran Frank Labrijn; Yvo Graus; Lisa A. Miller; Edward S. Schelegle; Dallas M. Hyde; Lawren C. Wu; Sarah G. Hymowitz; Flavius Martin

Thymic stromal lymphopoietin (TSLP) potently induces deregulation of Th2 responses, a hallmark feature of allergic inflammatory diseases such as asthma, atopic dermatitis, and allergic rhinitis. However, direct downstream in vivo mediators in the TSLP-induced atopic immune cascade have not been identified. In our current study, we have shown that OX40 ligand (OX40L) is a critical in vivo mediator of TSLP-mediated Th2 responses. Treating mice with OX40L-blocking antibodies substantially inhibited immune responses induced by TSLP in the lung and skin, including Th2 inflammatory cell infiltration, cytokine secretion, and IgE production. OX40L-blocking antibodies also inhibited antigen-driven Th2 inflammation in mouse and nonhuman primate models of asthma. This treatment resulted in both blockade of the OX40-OX40L receptor-ligand interaction and depletion of OX40L-positive cells. The use of a blocking, OX40L-specific mAb thus presents a promising strategy for the treatment of allergic diseases associated with pathologic Th2 immune responses.


The Journal of Allergy and Clinical Immunology | 2012

Increased expression of immunoreactive thymic stromal lymphopoietin in patients with severe asthma.

Aarti Shikotra; David F. Choy; Chandra M. Ohri; Emma Doran; Claire A. Butler; Beverley Hargadon; Maria Shelley; Alexander R. Abbas; Cary D. Austin; Janet Jackman; Lawren C. Wu; Liam Heaney; Joseph R. Arron; Peter Bradding

BACKGROUND Thymic stromal lymphopoietin (TSLP) is a cytokine implicated in the pathophysiology of asthma through 2 distinct pathways: a TSLP-OX40 ligand (OX40L)-T cell axis and a TSLP-mast cell axis. Whether these pathways are active in human asthma is unknown. OBJECTIVE We sought to investigate whether mucosal TSLP protein expression relates to asthma severity and distinct immunologic pathways. METHODS In healthy subjects and patients with mild-to-severe asthma, we immunostained bronchial biopsy specimens for TSLP, OX40, OX40L, T(H)2 cytokines, and inflammatory cell markers. We examined gene expression using RNA microarrays and quantitative RT-PCR. RESULTS There was considerable heterogeneity in the levels of TSLP, IL-13, and IL-4 immunostaining across the cohort of asthmatic patients examined. Overall, TSLP protein expression was significantly increased in airway epithelium and lamina propria of asthmatic patients, particularly in patients with severe asthma. TSLP immunostaining in both compartments correlated with the severity of airflow obstruction. The majority of leukocytes expressing IL-13 were possibly nuocytes. Accounting for intersubject variability, the 55% of asthmatic patients with increased IL-13 immunostaining in the lamina propria also had increased IL-4 and TSLP expression. This was further substantiated by significant correlations between TSLP gene expression, a T(H)2 gene expression signature, and eosinophilic inflammation in bronchial biopsy specimens. Immunostaining for OX40, OX40L, and CD83 was sparse, with no difference between asthmatic patients and healthy subjects. CONCLUSION TSLP expression is increased in a subset of patients with severe asthma in spite of high-dose inhaled or oral corticosteroid therapy. Targeting TSLP might only be efficacious in the subset of asthma characterized by increased TSLP expression and T(H)2 inflammation.


Science Translational Medicine | 2015

TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma

David F. Choy; Kevin M. Hart; Lee A. Borthwick; Aarti Shikotra; Deepti R. Nagarkar; Salman Siddiqui; Guiquan Jia; Chandra M. Ohri; Emma Doran; Kevin M. Vannella; Claire A. Butler; Beverley Hargadon; Joshua Sciurba; Richard L. Gieseck; Robert W. Thompson; Sandra White; Alexander R. Abbas; Janet Jackman; Lawren C. Wu; Jackson G. Egen; Liam Heaney; Thirumalai R. Ramalingam; Joseph R. Arron; Thomas A. Wynn; Peter Bradding

Concurrent blockade of IL-13 and IL-17A may improve control of asthma. A tale of two asthmas Classifying diseases according to symptoms is rapidly becoming a thing of the past. Targeted therapeutics have shown us that sets of symptoms can be caused by different pathogenic mechanisms. Now, Choy et al. demonstrate that asthma can be divided into three immunological clusters: TH2-high, TH17-high, and TH2/17-low. The TH2-high and TH17-high clusters were inversely correlated in patients. Moreover, neutralizing one signature promoted the other in a mouse model of asthma. These data suggest that combination therapies targeting both pathways may better treat asthmatic individuals. Increasing evidence suggests that asthma is a heterogeneous disorder regulated by distinct molecular mechanisms. In a cross-sectional study of asthmatics of varying severity (n = 51), endobronchial tissue gene expression analysis revealed three major patient clusters: TH2-high, TH17-high, and TH2/17-low. TH2-high and TH17-high patterns were mutually exclusive in individual patient samples, and their gene signatures were inversely correlated and differentially regulated by interleukin-13 (IL-13) and IL-17A. To understand this dichotomous pattern of T helper 2 (TH2) and TH17 signatures, we investigated the potential of type 2 cytokine suppression in promoting TH17 responses in a preclinical model of allergen-induced asthma. Neutralization of IL-4 and/or IL-13 resulted in increased TH17 cells and neutrophilic inflammation in the lung. However, neutralization of IL-13 and IL-17 protected mice from eosinophilia, mucus hyperplasia, and airway hyperreactivity and abolished the neutrophilic inflammation, suggesting that combination therapies targeting both pathways may maximize therapeutic efficacy across a patient population comprising both TH2 and TH17 endotypes.


Nature Reviews Immunology | 2014

The production and regulation of IgE by the immune system

Lawren C. Wu; Ali A. Zarrin

IgE not only provides protective immunity against helminth parasites but can also mediate the type I hypersensitivity reactions that contribute to the pathogenesis of allergic diseases such as asthma, allergic rhinitis and atopic dermatitis. Despite the importance of IgE in immune biology and allergic pathogenesis, the cells and the pathways that produce and regulate IgE are poorly understood. In this Review, we summarize recent advances in our understanding of the production and the regulation of IgE in vivo, as revealed by studies in mice, and we discuss how these findings compare to what is known about human IgE biology.


Journal of Immunology | 2011

Gene Expression Patterns of Th2 Inflammation and Intercellular Communication in Asthmatic Airways

David F. Choy; Barmak Modrek; Alexander R. Abbas; Sarah K. Kummerfeld; Hilary Clark; Lawren C. Wu; Grazyna Fedorowicz; Zora Modrusan; John V. Fahy; Prescott G. Woodruff; Joseph R. Arron

Asthma is canonically thought of as a disorder of excessive Th2-driven inflammation in the airway, although recent studies have described heterogeneity with respect to asthma pathophysiology. We have previously described distinct phenotypes of asthma based on the presence or absence of a three-gene “Th2 signature” in bronchial epithelium, which differ in terms of eosinophilic inflammation, mucin composition, subepithelial fibrosis, and corticosteroid responsiveness. In the present analysis, we sought to describe Th2 inflammation in human asthmatic airways quantitatively with respect to known mediators of inflammation and intercellular communication. Using whole-genome microarray and quantitative real-time PCR analysis of endobronchial biopsies from 27 mild-to-moderate asthmatics and 13 healthy controls with associated clinical and demographic data, we found that asthmatic Th2 inflammation is expressed over a variable continuum, correlating significantly with local and systemic measures of allergy and eosinophilia. We evaluated a composite metric describing 79 coexpressed genes associated with Th2 inflammation against the biological space comprising cytokines, chemokines, and growth factors, identifying distinctive patterns of inflammatory mediators as well as Wnt, TGF-β, and platelet-derived growth factor family members. This integrated description of the factors regulating inflammation, cell migration, and tissue remodeling in asthmatic airways has important consequences for the pathophysiological and clinical impacts of emerging asthma therapeutics targeting Th2 inflammation.


Nature Immunology | 2012

IgE⁺ memory B cells and plasma cells generated through a germinal-center pathway.

Oezcan Talay; Donghong Yan; Hans Brightbill; Elizabeth E M Straney; Meijuan Zhou; Ena Ladi; Wyne P. Lee; Jackson G. Egen; Cary D. Austin; Min Xu; Lawren C. Wu

Immunoglobulin E (IgE) antibodies are pathogenic in asthma and allergic diseases, but the in vivo biology of IgE-producing (IgE+) cells is poorly understood. A model of the differentiation of IgE+ B cells proposes that IgE+ cells develop through a germinal-center IgG1+ intermediate and that IgE memory resides in the compartment of IgG1+ memory B cells. Here we have used a reporter mouse expressing green fluorescent protein associated with membrane IgE transcripts (IgE-GFP) to assess in vivo IgE responses. In contrast to the IgG1-centered model of IgE switching and memory, we found that IgE+ cells developed through a germinal-center IgE+ intermediate to form IgE+ memory B cells and plasma cells. Our studies delineate a new model for the in vivo biology of IgE switching and memory.


Journal of Biological Chemistry | 2010

Development of a two-part strategy to identify a therapeutic human bispecific antibody that inhibits IgE receptor signaling

Janet Jackman; Yongmei Chen; Arthur Jyh-Yen Huang; Barbara Moffat; Justin Scheer; Steven R. Leong; Wyne P. Lee; Juan Zhang; Navneet Sharma; Yanmei Lu; Suhasini Iyer; Robert L. Shields; Nan Chiang; Michele C. Bauer; Diana Wadley; Merone Roose-Girma; Richard Vandlen; Daniel G. Yansura; Yan Wu; Lawren C. Wu

The development of bispecific antibodies as therapeutic agents for human diseases has great clinical potential, but broad application has been hindered by the difficulty of identifying bispecific antibody formats that exhibit favorable pharmacokinetic properties and ease of large-scale manufacturing. Previously, the development of an antibody technology utilizing heavy chain knobs-into-holes mutations and a single common light chain enabled the small-scale generation of human full-length bispecific antibodies. Here we have extended the technology by developing a two-part bispecific antibody discovery strategy that facilitates proof-of-concept studies and clinical candidate antibody generation. Our scheme consists of the efficient small-scale generation of bispecific antibodies lacking a common light chain and the hinge disulfides for proof-of-concept studies coupled with the identification of a common light chain bispecific antibody for large-scale production with high purity and yield. We have applied this technology to generate a bispecific antibody suitable for development as a human therapeutic. This antibody directly inhibits the activation of the high affinity IgE receptor FcϵRI on mast cells and basophils by cross-linking FcϵRI with the inhibitory receptor FcγRIIb, an approach that has strong therapeutic potential for asthma and other allergic diseases. Our approach for producing human bispecific full-length antibodies enables the clinical application of bispecific antibodies to a validated therapeutic pathway in 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.


Journal of Biological Chemistry | 2013

Development of a human IgG4 bispecific antibody for dual targeting of interleukin-4 (IL-4) and interleukin-13 (IL-13) cytokines

Christoph Spiess; Jack Bevers; Janet Jackman; Nan Chiang; Gerald R. Nakamura; Michael Patrick Dillon; Hongbin Liu; Patricia Molina; J. Michael Elliott; Whitney Shatz; Justin Scheer; Glen Giese; Josefine Persson; Yin Zhang; Mark S. Dennis; James Giulianotti; Prateek Gupta; Dorothea Reilly; Enzo Palma; Jianyong Wang; Eric Stefanich; Heleen Scheerens; Germaine Fuh; Lawren C. Wu

Background: Dual neutralization of IL-4 and IL-13 is a promising therapeutic approach for asthma and allergy. Results: Knobs-into-holes IgG1 and IgG4 bispecific antibodies targeting both cytokines were developed. Conclusion: Bispecific antibodies of both isotypes have comparable in vitro potencies, in vivo pharmacokinetics, and lung partitioning. Significance: Further extension of knobs-into-holes technology to human IgG4 isotype as reported here provides greater options for therapeutics. Human bispecific antibodies have great potential for the treatment of human diseases. Although human IgG1 bispecific antibodies have been generated, few attempts have been reported in the scientific literature that extend bispecific antibodies to other human antibody isotypes. In this paper, we report our work expanding the knobs-into-holes bispecific antibody technology to the human IgG4 isotype. We apply this approach to generate a bispecific antibody that targets IL-4 and IL-13, two cytokines that play roles in type 2 inflammation. We show that IgG4 bispecific antibodies can be generated in large quantities with equivalent efficiency and quality and have comparable pharmacokinetic properties and lung partitioning, compared with the IgG1 isotype. This work broadens the range of published therapeutic bispecific antibodies with natural surface architecture and provides additional options for the generation of bispecific antibodies with differing effector functions through the use of different antibody isotypes.

Collaboration


Dive into the Lawren C. Wu's collaboration.

Researchain Logo
Decentralizing Knowledge