Yuling Wu
MedImmune
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Yuling Wu.
Journal of Clinical Microbiology | 2015
Batu K. Sharma-Kuinkel; Yuling Wu; David E. Tabor; Hoyin Mok; Bret R. Sellman; Amy S. Jenkins; Li Yu; Hasan S. Jafri; Thomas H. Rude; Felicia Ruffin; Wiley A. Schell; Lawrence P. Park; Qin Yan; Joshua T. Thaden; Julia A. Messina; Vance G. Fowler; Mark T. Esser
ABSTRACT Alpha-toxin is a major Staphylococcus aureus virulence factor. This study evaluated potential relationships between in vitro alpha-toxin expression of S. aureus bloodstream isolates, anti-alpha-toxin antibody in serum of patients with S. aureus bacteremia (SAB), and clinical outcomes in 100 hemodialysis and 100 postsurgical SAB patients. Isolates underwent spa typing and hla sequencing. Serum anti-alpha-toxin IgG and neutralizing antibody levels were measured by using an enzyme-linked immunosorbent assay and a red blood cell (RBC)-based hemolysis neutralization assay. Neutralization of alpha-toxin by an anti-alpha-toxin monoclonal antibody (MAb MEDI4893) was tested in an RBC-based lysis assay. Most isolates encoded hla (197/200; 98.5%) and expressed alpha-toxin (173/200; 86.5%). In vitro alpha-toxin levels were inversely associated with survival (cure, 2.19 μg/ml, versus failure, 1.09 μg/ml; P < 0.01). Both neutralizing (hemodialysis, 1.26 IU/ml, versus postsurgical, 0.95; P < 0.05) and IgG (hemodialysis, 1.94 IU/ml, versus postsurgical, 1.27; P < 0.05) antibody levels were higher in the hemodialysis population. Antibody levels were also significantly higher in patients infected with alpha-toxin-expressing S. aureus isolates (P < 0.05). Levels of both neutralizing antibodies and IgG were similar among patients who were cured and those not cured (failures). Sequence analysis of hla revealed 12 distinct hla genotypes, and all genotypic variants were susceptible to a neutralizing monoclonal antibody in clinical development (MEDI4893). These data demonstrate that alpha-toxin is highly conserved in clinical S. aureus isolates. Higher in vitro alpha-toxin levels were associated with a positive clinical outcome. Although patients infected with alpha-toxin-producing S. aureus exhibited higher anti-alpha-toxin antibody levels, these levels were not associated with a better clinical outcome in this study.
Antimicrobial Agents and Chemotherapy | 2017
Xiang-Qing Yu; Gabriel Robbie; Yuling Wu; Mark T. Esser; Kathryn Jensen; Howard I. Schwartz; Terramika Bellamy; Martha Hernandez-Illas; Hasan S. Jafri
ABSTRACT MEDI4893 is an investigational immunoglobulin G1(κ) monoclonal antibody that specifically binds to and neutralizes alpha-toxin, a key Staphylococcus aureus virulence factor. A triple-amino-acid substitution, M252Y/S254T/T256E, was engineered into the MEDI4893 Fc region to extend its serum half-life. A phase 1, double-blind, dose escalation study was designed to evaluate the safety, tolerability, pharmacokinetics, anti-alpha-toxin-neutralizing activity, and antidrug antibody (ADA) response of MEDI4893 following a single intravenous infusion in healthy adults 18 to 65 years of age. Thirty-three subjects were randomly assigned to receive MEDI4893 at 225 mg (n = 3), 750 mg (n = 3), 2,250 mg (n = 8), or 5,000 mg (n = 12) or placebo (n = 7) and were followed for 360 days. Adverse events were mild or moderate in severity; none were serious. The MEDI4893 peak serum concentration increased dose proportionally from 77.2 μg/ml (225-mg dose) to 1,784 μg/ml (5,000-mg dose). The area under the concentration-time curve from 0 to 360 days also increased dose proportionally, from 4,840 μg · day/ml (225-mg dose) to 91,493 μg · day/ml (5,000-mg dose), indicating linear pharmacokinetics. MEDI4893s terminal half-life was estimated to be 80 to 112 days, which is approximately 4-fold longer than the half-lives of other human immunoglobulin G antibodies. The alpha-toxin-neutralizing activity in serum correlated highly with the MEDI4893 concentrations in serum. Three adults transiently tested positive for ADA on day 151, but this did not have an impact on MEDI4893 serum concentrations or the MEDI4893 safety profile; no subjects exhibited serum ADA at the study end. These data support the continued development of MEDI4893 for the prevention of S. aureus-mediated pneumonia. (This study has been registered at ClinicalTrials.gov under identifier NCT02296320.)
Journal of Pharmaceutical and Biomedical Analysis | 2013
Robert J. Kubiak; Lanju Zhang; Jianchun Zhang; Yuan Zhu; Nancy Lee; Frank F. Weichold; Harry Yang; Varghese Abraham; Peter F. Akufongwe; Lisa Hewitt; Susan Robinson; Weiyi Liu; Xu Liu; Mun Mun Patnaik; Susan Spitz; Yuling Wu; Lorin Roskos
Biotherapeutic proteins induce undesired immune responses that can affect drug efficacy and safety. For this reason, immunogenicity assessment is an integral part of drug development and is mandated by the regulatory authorities. Immunogenicity is typically evaluated by a tiered approach consisting of a screening assay followed by a competitive inhibition with unlabeled drug serving as confirmatory assay and additional characterization of the immune response. The confirmatory assay is intended to reduce the number of false positive responses generated in the screening tier and ensure that all samples are correctly classified as positive or negative. The positive-negative sample decisions are based on screening and confirmatory assay cut points that are statistically derived through evaluation of drug-naive samples. In this paper, we describe the analysis of cut point data for the presence of statistical correlation between the screening and confirmatory results. Data were obtained from validations of solution-phase bridging assays for detection of anti-drug antibodies against monoclonal antibody therapeutics. All data sets showed moderate to strong positive correlation, indicating that the screening and confirmatory assays were not independent and were likely to generate similar information. We present theoretical evidence that correlated results may be a general feature of the tiered approach when the same test platform is used for both screening and confirmatory assays. The competitive inhibition test, therefore, may be of limited value beyond reduction of the overall false positive rate. Our results indicate that similar sample results could be obtained by using just the screening assay with the false positive rate set to 1%.
Journal of Pharmaceutical and Biomedical Analysis | 2013
Yuling Wu; Xu Liu; Yan Chen; Robert M. Woods; Nancy Lee; Harry Yang; Partha S. Chowdhury; Lorin Roskos; Wendy I. White
To address a possible linkage between the occurrence of the hypersensitivity reactions and the induction of IgE anti-drug-antibodies (ADA), a drug specific IgE ADA assay was developed using electrochemiluminescence (ECL) technology. In the assay a drug-specific IgE isotype chimeric antibody was generated and used as an ADA positive control. The biotinylated drug X (an antibody) and ruthenium-labeled omalizumab (an anti-human IgE antibody) were used as capture and detection reagents, respectively. The binding affinities of the chimeric IgE isotype positive control have been shown to be highly comparable to drug X and drug Y (drug X is the 2nd generation of drug Y), indicating that it could serve as a highly useful control to compare and contrast the relative ability of the two generations of drug to elicit IgE ADA responses. The assay cut point factor (CPF) was estimated to be 1.13. The cut point factor derived from normal human serum samples was statistically equivalent to the cut point factor determined from targeted population samples. The assay could detect less than 250ng/mL of IgE antibodies in the presence of 300μg/mL drug X. The assay sensitivity was <0.2ng/mL. A minimal prozone was observed at 100μg/mL IgE ADA, but the sample remained highly detectable. The inter-assay precision was within 12%. The assay was not susceptible to non-specific matrix effects. The performance specifications ensured that the assay was suitable for validation. The combination of the chimeric IgE positive control and the detection antibody (ruthenium-labeled omalizumab) used for the assay could potentially provide a general bioanalytical approach for other biopharmaceuticals for the detection of IgE ADA responses.
International Immunopharmacology | 2016
Sandra Gallagher; Sean Turman; Isharat Yusuf; Ahmad Akhgar; Yuling Wu; Lorin Roskos; Ronald Herbst; Yue Wang
B cell depletion therapy is beneficial for patients with B cell malignancies and autoimmune diseases. CD19, a transmembrane protein, is expressed on a vast majority of normal and neoplastic B cells, making it a suitable target for monoclonal antibody (MAb) mediated immunotherapy. We have developed MEDI-551, an affinity optimized and afucosylated IgG1 MAb targeting human CD19 for B cell depletion. MEDI-551 is currently under investigation in multiple clinical trials. Because MEDI-551 does not cross react with rodent and non-human primate CD19, the pharmacological characteristics of the MAb were evaluated in human CD19 transgenic mice (hCD19 Tg). Here we show that MEDI-551 potently depletes tissue and circulating B cells in hCD19 Tg mice and is more efficacious than the anti-CD19 MAb with intact fucose. The length of B cell depletion depends on MEDI-551 dose; and, B cell recovery in the circulation follows stepwise phenotypic maturation. Furthermore, intravenous (IV) and subcutaneous (SC) administration of MEDI-551 results in comparable efficacy. Lastly, the combination of MEDI-551 with the anti-CD20 MAb, rituximab, further prolongs the duration of B cell depletion. In summary, the pharmacological profile of MEDI-551 presented in hCD19 Tg mice supports further testing of MEDI-551 in clinical trials involving B cell malignancies and autoimmune diseases.
Cancer Research | 2017
Shilpa Gupta; Juneko E. Grilley-Olson; David S. Hong; Aurelien Marabelle; Pamela N. Munster; Rahul Aggarwal; Sandrine Aspeslagh; Robert G. Dixon; Manish R. Patel; Vivek Subbiah; Chris Morehouse; Yuling Wu; Jiping Zha; Leo Tseng; Zachary Cooper; Shannon R. Morris; Joshua Brody
MEDI9197 (formerly 3M-052), is a novel TLR7/8 dual agonist formulated for intratumoral (IT) injection and optimized for retention within the tumor after IT injection. IT administration aims to focus antigen-presentation in the tumor thus minimizing systemic inflammatory toxicities. We report the safety and pharmacodynamic (PD) results from a phase I, FTIH study of MEDI9197 in patients (pts) with solid tumors (NCT02556463). As of 28 Nov 2016, this dose escalation trial of MEDI9197 (0.005 to 0.055 mg; Q4W) has enrolled 20 pts with subcutaneous/cutaneous tumors. The maximum-tolerated dose is 0.037 mg. Two dose-limiting-toxicities were observed: Grade 3 cytokine release syndrome (CRS) at 0.037 mg and Grade 4 CRS at 0.055 mg. The most common (> 4 pts)drug related adverse events (AEs) were pyrexia, fatigue, chills, decreased lymphocyte count, nausea, and injection site pain. One pt discontinued MEDI9197 as a result of a drug-related AE (Grade 4 CRS) and there were no Grade 5 drug-related AEs. Five out of 20 pts (25%) received ≥ 4 injections and one pt received 10 injections with treatment ongoing. Tumoral and peripheral PD effects of MEDI9197 were assessed from pts treated with 0.037 (n=6), 0.012 (n=5), and 0.005 (n=6) mg. Local PD effects were assessed by immunohistochemistry (IHC) in longitudinal biopsies with the majority of pts treated with 0.037 mg demonstrating an increase in CD8 (T cells), CD40 (myeloid and B cells), CD56 (NK cells) or PD-L1 (tumor and immune cells) 3-weeks after treatment initiation based on quantitative image analysis. RNAseq analysis of paired tumor biopsies showed an increase in innate and adaptive immune activation signatures consistent with IHC and indicating increased inflammation. Increased peripheral levels of IFNγ, CXCL10, and CXCL11 were observed in 5/6 pts with median peak values of 236, 9286, and 558 pg/mL respectively, within 24 hours of the 1st IT administration of 0.037 mg of MEDI9197. The fold change increase ranged from 4.5-43, 30-132, and 3.7-52 for IFNγ, CXCL10, and CXCL11 respectively. Interestingly, a second IT injection into the same lesion resulted in a blunted peak in IFNγ, CXCL10, and CXCL11 levels in 4/4 pts by 51.1%, 67.2%, and 58.2% vs. post-first dose suggesting tolerance or tachyphylaxis effect. Peak plasma levels of MEDI9197 were extremely low (≤0.1 ng/mL), ~600-fold below the minimum effective concentration (59 ng/mL) that induced cytokines in vitro in human peripheral blood mononuclear cells. Analysis of whole blood microarray data demonstrated increases (>2 fold) in TH1 and Type 1 IFN gene expression signatures with a transient decrease (>2 fold) in CD8A transcript expression suggesting trafficking of T cells in at least 4/6 pts treated with 0.037 mg of MEDI9197. In conclusion, IT administration of MEDI9197 was feasible and had AEs as well as local and systemic PD effects consistent with its expected mechanism of action. Citation Format: Shilpa Gupta, Juneko Grilley-Olson, David Hong, Aurelien Marabelle, Pamela Munster, Rahul Aggarwal, Sandrine Aspeslagh, Robert G. Dixon, Manish Patel, Vivek Subbiah, Chris Morehouse, Yuling Wu, Jiping Zha, Leo Tseng, Zachary A. Cooper, Shannon Morris, Joshua Brody. Safety and pharmacodynamic activity of MEDI9197, a TLR 7/8 agonist, administered intratumorally in subjects with solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr CT091. doi:10.1158/1538-7445.AM2017-CT091
Clinical And Translational Immunology | 2018
Alexey Ruzin; Yuling Wu; Li Yu; Xiang-Qing Yu; David E. Tabor; Hoyin Mok; Christine Tkaczyk; Kathryn Jensen; Terramika Bellamy; Lorin Roskos; Mark T. Esser; Hasan S. Jafri
MEDI4893 is a novel, long‐acting human monoclonal antibody targeting Staphylococcus aureus (SA) alpha toxin (AT). This report presents the results of the exploratory analyses from a randomised phase 1 dose‐escalation study in healthy human subjects receiving single intravenous MEDI4893 doses or placebo.
Infection and Immunity | 2017
Yuling Wu; Xu Liu; Ahmad Akhgar; Jia J. Li; Hoyin Mok; Bret R. Sellman; Li Yu; Lorin Roskos; Mark T. Esser; Alexey Ruzin
ABSTRACT Staphylococcus aureus causes an array of serious infections resulting in high morbidity and mortality worldwide. This study evaluated naturally occurring serum anti-alpha-toxin (anti-AT) antibody levels in human subjects from various age groups, individuals with S. aureus dialysis and surgical-site infections, and S. aureus-colonized versus noncolonized subjects. Anti-AT immunoglobulin G (IgG) and neutralizing antibody (NAb) levels in infants (aged ≤1 year) were significantly lower than those in other populations. In comparison to adolescent, adult, and elderly populations, young children (aged 2 to 10 years) had equivalent anti-AT IgG levels but significantly lower anti-AT NAb levels. Therefore, the development of anti-AT NAbs appears to occur later than that of AT-specific IgG, suggesting a maturation of the immune response to AT. Anti-AT IgG levels were slightly higher in S. aureus-colonized subjects than in noncolonized subjects. The methicillin susceptibility status of colonizing isolates had no effect on anti-AT antibody levels in S. aureus-colonized subjects. The highest anti-AT IgG and NAb levels were observed in dialysis patients with acute S. aureus infection. Anti-AT IgG and NAb levels were well correlated in subjects aged >10 years, regardless of colonization or infection status. These data demonstrate that AT elicits a robust IgG humoral response in infants and young children that becomes stable prior to adolescence, matures into higher levels of NAbs in healthy adolescents, and becomes elevated during S. aureus infection. These findings may assist in identifying subjects and patient populations that could benefit from vaccination or immunoprophylaxis with anti-AT monoclonal antibodies.
Aaps Journal | 2017
Zhandong Don Zhong; Adrienne Clements-Egan; Boris Gorovits; Mauricio Maia; Giane Sumner; Valerie Theobald; Yuling Wu; Manoj Rajadhyaksha
Sensitive and specific methodology is required for the detection and characterization of anti-drug antibodies (ADAs). High-quality ADA data enables the evaluation of potential impact of ADAs on the drug pharmacokinetic profile, patient safety, and efficacious response to the drug. Immunogenicity assessments are typically initiated at early stages in preclinical studies and continue throughout the drug development program. One of the potential bioanalytical challenges encountered with ADA testing is the need to identify and mitigate the interference mediated by the presence of soluble drug target. A drug target, when present at sufficiently high circulating concentrations, can potentially interfere with the performance of ADA and neutralizing antibody (NAb) assays, leading to either false-positive or, in some cases, false-negative ADA and NAb assay results. This publication describes various mechanisms of assay interference by soluble drug target, as well as strategies to recognize and mitigate such target interference. Pertinent examples are presented to illustrate the impact of target interference on ADA and NAb assays as well as several mitigation strategies, including the use of anti-target antibodies, soluble versions of the receptors, target-binding proteins, lectins, and solid-phase removal of targets. Furthermore, recommendations for detection and mitigation of such interference in different formats of ADA and NAb assays are provided.
Rheumatology | 2018
Xiang Guo; Brandon W. Higgs; A.-C. Bay-Jensen; Yuling Wu; Morten A. Karsdal; Michael Kuziora; A. Godwood; D. Close; Patricia C. Ryan; Lorin Roskos; Wendy I. White
Objectives Targeting the granulocyte-macrophage colony-stimulating factor (GM-CSF) pathway holds great potential in the treatment of inflammatory diseases. Mavrilimumab, a human monoclonal GM-CSF receptor-α antibody, has demonstrated clinical efficacy in RA. Our current study aimed to elucidate mechanisms of action and identify peripheral biomarkers associated with therapeutic responses of GM-CSF antagonism in RA. Methods A 24-week placebo (PBO)-controlled trial was conducted in 305 RA patients who received mavrilimumab (30, 100 or 150 mg) or PBO once every 2 weeks. Serum biomarkers and whole blood gene expression profiles were measured by protein immunoassay and whole genome microarray. Results Mavrilimumab treatment induced significant down-regulation of type IV collagen formation marker (P4NP 7S), macrophage-derived chemokine (CCL22), IL-2 receptor α and IL-6 compared with PBO. Both early and sustained reduction of P4NP 7S was associated with clinical response to 150 mg mavrilimumab treatment. Gene expression analyses demonstrated reduced expression of transcripts enriched in macrophage and IL-22/IL-17 signalling pathways after GM-CSF blockade therapy. Myeloid and T cell-associated transcripts were suppressed in mavrilimumab-treated ACR20 responders but not non-responders. While CCL22 and IL-6 down-regulation may reflect a direct effect of GM-CSFR blockade on the production of pro-inflammatory mediators by myeloid cells, the suppression of IL-2 receptor α and IL-17/IL-22 associated transcripts suggests an indirect suppressive effect of mavrilimumab on T cell activation. Conclusion Our results demonstrated association of peripheral biomarker changes with therapeutic response to mavrilimumab in RA patients. The sustained efficacy of mavrilimumab in RA may result from both direct effects on myeloid cells and indirect effects on T cell activation after GM-CSFR blockade.