W. Wen
University of California, San Francisco
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by W. Wen.
Protein Engineering Design & Selection | 2011
Consuelo Garcia-Rodriguez; Isin N. Geren; Jianlong Lou; Fraser Conrad; C.M. Forsyth; W. Wen; S. Chakraborti; H. Zao; G. Manzanarez; Thomas J. Smith; J. Brown; W.H. Tepp; N. Liu; S. Wijesuriya; M.T. Tomic; Eric A. Johnson; Leonard A. Smith; James D. Marks
Botulism, a disease of humans characterized by prolonged paralysis, is caused by botulinum neurotoxins (BoNTs), the most poisonous substances known. There are seven serotypes of BoNT (A-G) which differ from each other by 34-64% at the amino acid level. Each serotype is uniquely recognized by polyclonal antibodies, which originally were used to classify serotypes. To determine if there existed monoclonal antibodies (mAbs) capable of binding two or more serotypes, we evaluated the ability of 35 yeast-displayed single-chain variable fragment antibodies generated from vaccinated humans or mice for their ability to bind multiple BoNT serotypes. Two such clonally related human mAbs (1B18 and 4E17) were identified that bound BoNT serotype A (BoNT/A) and B or BoNT/A, B, E and F, respectively, with high affinity. Using molecular evolution techniques, it proved possible to both increase affinity and maintain cross-serotype reactivity for the 4E17 mAb. Both 1B18 and 4E17 bound to a relatively conserved epitope at the tip of the BoNT translocation domain. Immunoglobulin G constructed from affinity matured variants of 1B18 and 4E17 were evaluated for their ability to neutralize BoNT/B and E, respectively, in vivo. Both antibodies potently neutralized BoNT in vivo demonstrating that this epitope is functionally important in the intoxication pathway. Such cross-serotype binding and neutralizing mAbs should simplify the development of antibody-based BoNT diagnostics and therapeutics.
Science Translational Medicine | 2014
Shunsuke Minagawa; Jianlong Lou; Robert Seed; Anthony Cormier; Shenping Wu; Yifan Cheng; Lynne A. Murray; Ping Tsui; Jane Connor; Ronald Herbst; Cedric Govaerts; Tyren Barker; Stephanie Cambier; Haruhiko Yanagisawa; Amanda Goodsell; Mitsuo Hashimoto; Oliver J. Brand; Ran Cheng; Royce Ma; Kate J. McKnelly; W. Wen; Arthur Hill; David M. Jablons; Paul J. Wolters; Hideya Kitamura; Jun Araya; Andrea J. Barczak; David J. Erle; Louis F. Reichardt; James D. Marks
Therapeutic targeting of an extended-closed conformation of the integrin αvβ8 inhibits TGF-β activation and ameliorates symptoms of experimental airway disease in mice. Breathing Freely Narrowing of the airways through accumulation of scar tissue and inflammation results from chronic injury in common diseases such as chronic obstructive pulmonary disease (COPD) and severe chronic asthma. Such airway narrowing causes the obstruction responsible for the breathlessness that these patients experience, and there are no available treatments that ameliorate fibroinflammatory airway narrowing. In a new study, Minagawa et al. engineered a monoclonal antibody that locks in a specific inactive conformation of a protein named integrin αvβ8. This protein is a crucial receptor required for activation of transforming growth factor–β, a central mediator of pathological inflammation and fibrosis. This antibody, when administered to mice engineered to express only human and not mouse αvβ8, reduced airway inflammation and fibrosis in response to a variety of injurious agents including cigarette smoke and allergens that are involved in the pathogenesis of COPD. Airway remodeling, caused by inflammation and fibrosis, is a major component of chronic obstructive pulmonary disease (COPD) and currently has no effective treatment. Transforming growth factor–β (TGF-β) has been widely implicated in the pathogenesis of airway remodeling in COPD. TGF-β is expressed in a latent form that requires activation. The integrin αvβ8 (encoded by the itgb8 gene) is a receptor for latent TGF-β and is essential for its activation. Expression of integrin αvβ8 is increased in airway fibroblasts in COPD and thus is an attractive therapeutic target for the treatment of airway remodeling in COPD. We demonstrate that an engineered optimized antibody to human αvβ8 (B5) inhibited TGF-β activation in transgenic mice expressing only human and not mouse ITGB8. The B5 engineered antibody blocked fibroinflammatory responses induced by tobacco smoke, cytokines, and allergens by inhibiting TGF-β activation. To clarify the mechanism of action of B5, we used hydrodynamic, mutational, and electron microscopic methods to demonstrate that αvβ8 predominantly adopts a constitutively active, extended-closed headpiece conformation. Epitope mapping and functional characterization of B5 revealed an allosteric mechanism of action due to locking-in of a low-affinity αvβ8 conformation. Collectively, these data demonstrate a new model for integrin function and present a strategy to selectively target the TGF-β pathway to treat fibroinflammatory airway diseases.
Protein Engineering Design & Selection | 2010
Jianlong Lou; Isin N. Geren; Consuelo Garcia-Rodriguez; C.M. Forsyth; W. Wen; K. Knopp; J. Brown; Thomas J. Smith; Leonard A. Smith; James D. Marks
Botulism is caused by the botulinum neurotoxins (BoNTs), the most poisonous substance known. Because of the high potency of BoNT, development of diagnostic and therapeutic antibodies for botulism requires antibodies of very high affinity. Here we report the use of yeast mating to affinity mature BoNT antibodies by light chain shuffling. A library of immunoglobulin light chains was generated in a yeast vector where the light chain is secreted. The heavy chain variable region and the first domain of the constant region (V(H)-C(H)1) from a monoclonal antibody was cloned into a different yeast vector for surface display as a fusion to the Aga2 protein. Through yeast mating of the two haploid yeasts, a library of light chain-shuffled Fab was created. Using this approach, the affinities of one BoNT/A and two BoNT/B scFv antibody fragments were increased from 9- to more than 77-fold. Subcloning the V-genes from the affinity-matured Fab yielded fully human IgG1 with equilibrium binding constants for BoNT/A and BoNT/B of 2.51 x 10(-11) M or lower for all three monoclonal antibodies. This technique provides a rapid route to antibody affinity maturation.
PLOS ONE | 2015
Yongfeng Fan; Isin N. Geren; Jianbo Dong; Jianlong Lou; W. Wen; Fraser Conrad; Theresa J. Smith; Leonard A. Smith; Mengfei Ho; Melissa Pires-Alves; Brenda A. Wilson; James D. Marks
The paralytic disease botulism is caused by botulinum neurotoxins (BoNT), multi-domain proteins containing a zinc endopeptidase that cleaves the cognate SNARE protein, thereby blocking acetylcholine neurotransmitter release. Antitoxins currently used to treat botulism neutralize circulating BoNT but cannot enter, bind to or neutralize BoNT that has already entered the neuron. The light chain endopeptidase domain (LC) of BoNT serotype A (BoNT/A) was targeted for generation of monoclonal antibodies (mAbs) that could reverse paralysis resulting from intoxication by BoNT/A. Single-chain variable fragment (scFv) libraries from immunized humans and mice were displayed on the surface of yeast, and 19 BoNT/A LC-specific mAbs were isolated by using fluorescence-activated cell sorting (FACS). Affinities of the mAbs for BoNT/A LC ranged from a KD value of 9.0×10−11 M to 3.53×10−8 M (mean KD 5.38×10−9 M and median KD 1.53×10−9 M), as determined by flow cytometry analysis. Eleven mAbs inhibited BoNT/A LC catalytic activity with IC50 values ranging from 8.3 ~73×10−9 M. The fine epitopes of selected mAbs were also mapped by alanine-scanning mutagenesis, revealing that the inhibitory mAbs bound the α-exosite region remote from the BoNT/A LC catalytic center. The results provide mAbs that could prove useful for intracellular reversal of paralysis post-intoxication and further define epitopes that could be targeted by small molecule inhibitors.
Toxins | 2015
Yongfeng Fan; Jianbo Dong; Jianlong Lou; W. Wen; Fraser Conrad; Isin N. Geren; Consuelo Garcia-Rodriguez; Theresa J. Smith; Leonard A. Smith; Mengfei Ho; Melissa Pires-Alves; Brenda A. Wilson; James D. Marks
Existing antibodies (Abs) used to treat botulism cannot enter the cytosol of neurons and bind to botulinum neurotoxin (BoNT) at its site of action, and thus cannot reverse paralysis. However, Abs targeting the proteolytic domain of the toxin could inhibit the proteolytic activity of the toxin intracellularly and potentially reverse intoxication, if they could be delivered intracellularly. As such, antibodies that neutralize toxin activity could serve as potent inhibitory cargos for therapeutic antitoxins against botulism. BoNT serotype B (BoNT/B) contains a zinc endopeptidase light chain (LC) domain that cleaves synaoptobrevin-2, a SNARE protein responsible for vesicle fusion and acetylcholine vesicle release. To generate monoclonal Abs (mAbs) that could reverse paralysis, we targeted the protease domain for Ab generation. Single-chain variable fragment (scFv) libraries from immunized mice or humans were displayed on yeast, and 19 unique BoNT/B LC-specific mAbs isolated by fluorescence-activated cell sorting (FACS). The equilibrium dissociation constants (KD) of these mAbs for BoNT/B LC ranged from 0.24 nM to 14.3 nM (mean KD 3.27 nM). Eleven mAbs inhibited BoNT/B LC proteolytic activity. The fine epitopes of selected mAbs were identified by alanine-scanning mutagenesis, revealing that inhibitory mAbs bound near the active site, substrate-binding site or the extended substrate-binding site. The results provide mAbs that could prove useful for intracellular reversal of paralysis and identify epitopes that could be targeted by small molecules inhibitors.
PLOS ONE | 2017
Yongfeng Fan; Consuelo Garcia-Rodriguez; Jianlong Lou; W. Wen; Fraser Conrad; Wenwu Zhai; Theresa J. Smith; Leonard A. Smith; James D. Marks
Human botulism is primarily caused by botulinum neurotoxin (BoNT) serotypes A, B and E, with around 1% caused by serotype F (BoNT/F). BoNT/F comprises at least seven different subtypes with the amino acid sequence difference between subtypes as high as 36%. The sequence differences present a significant challenge for generating monoclonal antibodies (mAbs) that can bind, detect and neutralize all BoNT/F subtypes. We used repertoire cloning of immune mouse antibody variable (V) regions and yeast display to generate a panel of 33 lead single chain Fv (scFv) mAbs that bound one or more BoNT/F subtypes with a median equilibrium dissociation constant (KD) of 4.06 × 10−9 M. By diversifying the V-regions of the lead mAbs and selecting for cross reactivity we generated five mAbs that bound each of the seven subtypes. Three scFv binding non-overlapping epitopes were converted to IgG that had KD for the different BoNT/F subtypes ranging from 2.2×10−8 M to 1.47×10−12 pM. An equimolar combination of the mAbs was able to potently neutralize BoNT/F1, F2, F4 and F7 in the mouse neutralization assay. The mAbs have potential utility as diagnostics capable of recognizing the known BoNT/F subtypes and could be developed as antitoxins to prevent and treat type F botulism.
Toxins | 2018
Consuelo Garcia-Rodriguez; Ali Razai; Isin N. Geren; Jianlong Lou; Fraser Conrad; W. Wen; Shauna Farr-Jones; Theresa J. Smith; Jennifer Brown; Janet Skerry; Leonard A. Smith; James D. Marks
Human botulism is most commonly caused by botulinum neurotoxin (BoNT) serotypes A, B, and E. For this work, we sought to develop a human monoclonal antibody (mAb)-based antitoxin capable of binding and neutralizing multiple subtypes of BoNT/E. Libraries of yeast-displayed single chain Fv (scFv) antibodies were created from the heavy and light chain variable region genes of humans immunized with pentavalent-toxoid- and BoNT/E-binding scFv isolated by Fluorescence-Activated Cell Sorting (FACS). A total of 10 scFv were isolated that bound one or more BoNT/E subtypes with nanomolar-level equilibrium dissociation constants (KD). By diversifying the V-regions of the lead mAbs and selecting for cross-reactivity, we generated three scFv that bound all four BoNT/E subtypes tested at three non-overlapping epitopes. The scFvs were converted to IgG that had KD values for the different BoNT/E subtypes ranging from 9.7 nM to 2.28 pM. An equimolar combination of the three mAbs was able to potently neutralize BoNT/E1, BoNT/E3, and BoNT/E4 in a mouse neutralization assay. The mAbs have potential utility as therapeutics and as diagnostics capable of recognizing multiple BoNT/E subtypes. A derivative of the three-antibody combination (NTM-1633) is in pre-clinical development with an investigational new drug (IND) application filing expected in 2018.
Toxins | 2018
Jianlong Lou; W. Wen; Fraser Conrad; Qi Meng; Jianbo Dong; Zhengda Sun; Consuelo Garcia-Rodriguez; Shauna Farr-Jones; Luisa Cheng; Thomas D. Henderson; Jennifer Brown; Theresa J. Smith; Leonard A. Smith; Anthony Cormier; James D. Marks
The standard of treatment for botulism, equine antitoxin, is a foreign protein with associated safety issues and a short serum half-life which excludes its use as a prophylactic antitoxin and makes it a less-than-optimal therapeutic. Due to these limitations, a recombinant monoclonal antibody (mAb) product is preferable. It has been shown that combining three mAbs that bind non-overlapping epitopes leads to highly potent botulinum neurotoxin (BoNT) neutralization. Recently, a triple human antibody combination for BoNT/A has demonstrated potent toxin neutralization in mouse models with no serious adverse events when tested in a Phase I clinical trial. However, a triple antibody therapeutic poses unique development and manufacturing challenges. Thus, potentially to streamline development of BoNT antitoxins, we sought to achieve the potency of multiple mAb combinations in a single IgG-based molecule that has a long serum half-life. The design, production, and testing of a single tri-epitopic IgG1-based mAb (TeAb) containing the binding sites of each of the three parental BoNT/A mAbs yielded an antibody of nearly equal potency to the combination. The approach taken here could be applied to the design and creation of other multivalent antibodies that could be used for a variety of applications, including toxin elimination.
Protein Engineering Design & Selection | 2011
Consuelo Garcia-Rodriguez; Isin N. Geren; Jianlong Lou; Fraser Conrad; C.M. Forsyth; W. Wen; S. Chakraborti; H. Zao; G. Manzanarez; Thomas J. Smith; J. Brown; W.H. Tepp; N. Liu; S. Wijesuriya; M.T. Tomic; Eric A. Johnson; Leonard A. Smith; James D. Marks
Toxicon | 2015
Jianlong Lou; Consuelo Garcia-Rodriguez; Yongfeng Fan; Jianbo Dong; W. Wen; Fraser Conrad; Gabriel Manzanarez; Margarita Sanchez; James D. Marks
Collaboration
Dive into the W. Wen's collaboration.
United States Army Medical Research Institute of Infectious Diseases
View shared research outputsUnited States Army Medical Research Institute of Infectious Diseases
View shared research outputs