Network


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

Hotspot


Dive into the research topics where Xiufeng Wu is active.

Publication


Featured researches published by Xiufeng Wu.


mAbs | 2009

Anti-tumor activity of stability-engineered IgG-like bispecific antibodies targeting TRAIL-R2 and LTβR

Jennifer S. Michaelson; Stephen J. Demarest; Brian Robert Miller; Aldo Amatucci; William B. Snyder; Xiufeng Wu; Flora Huang; Samantha Phan; Sharon X. Gao; Adam Doern; Graham K. Farrington; Alexey Lugovskoy; Ingrid Joseph; Veronique Bailly; Xin Wang; Ellen Garber; Jeffrey L. Browning; Scott Glaser

Bispecific antibodies (BsAbs) represent an emerging class of biologics that achieve dual targeting with a single agent. Recombinant DNA technologies have facilitated a variety of creative bispecific designs with many promising therapeutic applications; however, practical methods for producing high quality BsAbs that have good product stability, long serum half-life, straightforward purification, and scalable production have largely been limiting. Here we describe a protein-engineering approach for producing stable, scalable tetravalent IgG-like BsAbs. The stability-engineered IgG-like BsAb was envisioned to target and crosslink two TNF family member receptors, TRAIL-R2 (TNF-Related Apoptosis Inducing Ligand Receptor-2) and LTβR (Lymphotoxin-beta Receptor), expressed on the surface of epithelial tumor cells with the goal of triggering an enhanced anti-tumor effect. Our IgG-like BsAbs consists of a stability-engineered anti- LTβR single chain Fv (scFv) genetically fused to either the N- or C-terminus of the heavy chain of a full-length anti-TRAIL-R2 IgG1 monoclonal antibody. Both N- or C-terminal BsAbs were active in inhibiting tumor cell growth in vitro, and with some cell lines demonstrated enhanced activity relative to the combination of parental Abs. Pharmacokinetic studies in mice revealed long serum half-lives for the BsAbs. In murine tumor xenograft models, therapeutic treatment with the BsAbs resulted in reduction in tumor volume either comparable to or greater than the combination of parental antibodies, indicating that simultaneously targeting and cross-linking receptor pairs is an effective strategy for treating tumor cells. These studies support that stability-engineering is an enabling step for producing scalable IgG-like BsAbs with properties desirable for biopharmaceutical development.


Protein Engineering Design & Selection | 2010

Stability engineering of scFvs for the development of bispecific and multivalent antibodies

Brian Robert Miller; Stephen J. Demarest; Alexey Lugovskoy; Flora Huang; Xiufeng Wu; William B. Snyder; Lisa J. Croner; Norman Wang; Aldo Amatucci; Jennifer S. Michaelson; Scott Glaser

Single-chain Fvs (scFvs) are commonly used building blocks for creating engineered diagnostic and therapeutic antibody molecules. Bispecific antibodies (BsAbs) hold particular interest due to their ability to simultaneously bind and engage two distinct targets. We describe a technology for producing stable, scalable IgG-like bispecific and multivalent antibodies based on methods for rapidly engineering thermally stable scFvs. Focused libraries of mutant scFvs were designed using a combination of sequence-based statistical analyses and structure-, and knowledge-based methods. Libraries encoding these designs were expressed in E. coli and culture supernatants-containing soluble scFvs screened in a high-throughput assay incorporating a thermal challenge prior to an antigen-binding assay. Thermally stable scFvs were identified that retain full antigen-binding affinity. Single mutations were found that increased the measured T(m) of either the V(H) or V(L) domain by as much as 14 degrees C relative to the wild-type scFv. Combinations of mutations further increased the T(m) by as much as an additional 12 degrees C. Introduction of a stability-engineered scFv as part of an IgG-like BsAb enabled scalable production and purification of BsAb with favorable biophysical properties.


mAbs | 2011

A stable IgG-like bispecific antibody targeting the epidermal growth factor receptor and the type I insulin-like growth factor receptor demonstrates superior anti-tumor activity

Jianying Dong; Arlene Sereno; Dikran Aivazian; Emma Langley; Brian Robert Miller; William Snyder; Eric Chan; Matt Cantele; Ronald Morena; Ingrid B.J.K. Joseph; Antonio Boccia; Cyrus Virata; James Gamez; Grace Yco; Michael Favis; Xiufeng Wu; Christilyn Graff; Qin Wang; Ellen Rohde; Rachel Rennard; Lisa Berquist; Flora Huang; Ying Zhang; Sharon X. Gao; Steffan N. Ho; Stephen J. Demarest; Mitchell E Reff; Kandasamy Hariharan; Scott Glaser

The epidermal growth factor receptor (EGFR) and the type I insulin-like growth factor receptor (IGF-1R) are two cell surface receptor tyrosine kinases known to cooperate to promote tumor progression and drug resistance. Combined blockade of EGFR and IGF-1R has shown improved anti-tumor activity in preclinical models. Here, we report the characterization of a stable IgG-like bispecific antibody (BsAb) dual-targeting EGFR and IGF-1R that was developed for cancer therapy. The BsAb molecule (EI-04), constructed with a stability-engineered single chain variable fragment (scFv) against IGF-1R attached to the carboxyl-terminus of an IgG against EGFR, displays favorable biophysical properties for biopharmaceutical development. Biochemically, EI-04 bound to human EGFR and IGF-1R with sub nanomolar affinity, co-engaged the two receptors simultaneously, and blocked the binding of their respective ligands with similar potency compared to the parental monoclonal antibodies (mAbs). In tumor cells, EI-04 effectively inhibited EGFR and IGF-1R phosphorylation, and concurrently blocked downstream AKT and ERK activation, resulting in greater inhibition of tumor cell growth and cell cycle progression than the single mAbs. EI-04, likely due to its tetravalent bispecific format, exhibited high avidity binding to BxPC3 tumor cells co-expressing EGFR and IGF-1R, and consequently improved potency at inhibiting IGF-driven cell growth over the mAb combination. Importantly, EI-04 demonstrated enhanced in vivo anti-tumor efficacy over the parental mAbs in two xenograft models, and even over the mAb combination in the BxPC3 model. Our data support the clinical investigation of EI-04 as a superior cancer therapeutic in treating EGFR and IGF-1R pathway responsive tumors.


Proteins | 2009

Structural understanding of stabilization patterns in engineered bispecific Ig-like antibody molecules

Jacob L. Jordan; Joseph Arndt; Karl Hanf; Guohui Li; Janine Hall; Stephen J. Demarest; Flora Huang; Xiufeng Wu; Brian Robert Miller; Scott Glaser; Erik J. Fernandez; Deping Wang; Alexey Lugovskoy

Bispecific immunoglobulin‐like antibodies capable of engaging multiple antigens represent a promising new class of therapeutic agents. Engineering of these molecules requires optimization of the molecular properties of one of the domain components. Here, we present a detailed crystallographic and computational characterization of the stabilization patterns in the lymphotoxin‐beta receptor (LTβR) binding Fv domain of an anti‐LTβR/anti‐TNF‐related apoptosis inducing ligand receptor‐2 (TRAIL‐R2) bispecific immunoglobulin‐like antibody. We further describe a new hierarchical structure‐guided approach toward engineering of antibody‐like molecules to enhance their thermal and chemical stability. Proteins 2009.


Journal of Biological Chemistry | 2011

Stable IgG-like Bispecific Antibodies Directed toward the Type I Insulin-like Growth Factor Receptor Demonstrate Enhanced Ligand Blockade and Anti-tumor Activity

Jianying Dong; Arlene Sereno; William B. Snyder; Brian Robert Miller; Susan Tamraz; Adam Doern; Michael Favis; Xiufeng Wu; Hon Tran; Emma Langley; Ingrid Joseph; Antonio Boccia; Rebecca Kelly; Kathleen Wortham; Qin Wang; Lisa Berquist; Flora Huang; Sharon X. Gao; Ying Zhang; Alexey Lugovskoy; Shelly Martin; Heather Gouvis; Steven A. Berkowitz; Gisela G. Chiang; Mitchell Reff; Scott Glaser; Kandasamy Hariharan; Stephen J. Demarest

Bispecific antibodies (BsAbs) target multiple epitopes on the same molecular target or different targets. Although interest in BsAbs has persisted for decades, production of stable and active BsAbs has hindered their clinical evaluation. Here, we describe the production and characterization of tetravalent IgG-like BsAbs that combine the activities of allosteric and competitive inhibitors of the type-I insulin-like growth factor receptor (IGF-1R). The BsAbs, which were engineered for thermal stability, express well, demonstrate favorable biophysical properties, and recognize both epitopes on IGF-1R. Only one BsAb with a unique geometry, denoted BIIB4-5scFv, was capable of engaging all four of its binding arms simultaneously. All the BsAbs (especially BIIB4-5scFv) demonstrated enhanced ligand blocking over the single monoclonal antibodies (mAbs), particularly at high ligand concentrations. The pharmacokinetic profiles of two IgG-like BsAbs were tested in nude mice and shown to be comparable with that of the parental mAbs. The BsAbs, especially BIIB4-5scFv, demonstrated an improved ability to reduce the growth of multiple tumor cell lines and to inhibit ligand-induced IGF-1R signaling in tumor cells over the parental mAbs. BIIB4-5scFv also led to superior tumor growth inhibition over its parental mAbs in vivo. In summary, BsAbs that bridge multiple inhibitory mechanisms against a single target may generally represent a more effective strategy for intervention in oncology or other indications compared with traditional mAb therapy.


Journal of Biological Chemistry | 2009

Characterization of inhibitory anti-insulin-like growth factor receptor antibodies with different epitope specificity and ligand-blocking properties: implications for mechanism of action in vivo.

Adam Doern; Xianjun Cao; Arlene Sereno; Christopher L. Reyes; Angelina E. Altshuler; Flora Huang; Cathy Hession; Albert Flavier; Michael Favis; Hon Tran; Eric Ailor; Melissa Levesque; Tracey Murphy; Lisa Berquist; Susan Tamraz; Tracey Snipas; Ellen Garber; William S. Shestowsky; Rachel Rennard; Christilyn Graff; Xiufeng Wu; William Snyder; Lindsay J. Cole; David Gregson; Michael Shields; Steffan N. Ho; Mitchell E Reff; Scott Glaser; Jianying Dong; Stephen J. Demarest

Therapeutic antibodies directed against the type 1 insulin-like growth factor receptor (IGF-1R) have recently gained significant momentum in the clinic because of preliminary data generated in human patients with cancer. These antibodies inhibit ligand-mediated activation of IGF-1R and the resulting down-stream signaling cascade. Here we generated a panel of antibodies against IGF-1R and screened them for their ability to block the binding of both IGF-1 and IGF-2 at escalating ligand concentrations (>1 μm) to investigate allosteric versus competitive blocking mechanisms. Four distinct inhibitory classes were found as follows: 1) allosteric IGF-1 blockers, 2) allosteric IGF-2 blockers, 3) allosteric IGF-1 and IGF-2 blockers, and 4) competitive IGF-1 and IGF-2 blockers. The epitopes of representative antibodies from each of these classes were mapped using a purified IGF-1R library containing 64 mutations. Most of these antibodies bound overlapping surfaces on the cysteine-rich repeat and L2 domains. One class of allosteric IGF-1 and IGF-2 blocker was identified that bound a separate epitope on the outer surface of the FnIII-1 domain. Using various biophysical techniques, we show that the dual IGF blockers inhibit ligand binding using a spectrum of mechanisms ranging from highly allosteric to purely competitive. Binding of IGF-1 or the inhibitory antibodies was associated with conformational changes in IGF-1R, linked to the ordering of dynamic or unstructured regions of the receptor. These results suggest IGF-1R uses disorder/order within its polypeptide sequence to regulate its activity. Interestingly, the activity of representative allosteric and competitive inhibitors on H322M tumor cell growth in vitro was reflective of their individual ligand-blocking properties. Many of the antibodies in the clinic likely adopt one of the inhibitory mechanisms described here, and the outcome of future clinical studies may reveal whether a particular inhibitory mechanism leads to optimal clinical efficacy.


Biotechnology Progress | 2017

Transient and stable CHO expression, purification and characterization of novel hetero-dimeric bispecific IgG antibodies

Yashas Rajendra; Robert B. Peery; Maria D. Hougland; Gavin C. Barnard; Xiufeng Wu; Jonathan R. Fitchett; Michael Bacica; Stephen J. Demarest

IgG bispecific antibodies (BsAbs) represent one of the preferred formats for bispecific antibody therapeutics due to their native‐like IgG properties and their monovalent binding to each target. Most reported studies utilized transient expression in HEK293 cells to produce BsAbs. However, the expression of biotherapeutic molecules using stable CHO cell lines is commonly used for biopharmaceutical manufacturing. Unfortunately, limited information is available in the scientific literature on the expression of BsAbs in CHO cell lines. In this study we describe an alternative approach to express the multiple components of IgG BsAbs using a single plasmid vector (quad vector). This single plasmid vector contains both heavy chain genes and both light chain genes required for the expression and assembly of the IgG BsAb, along with a selectable marker. We expressed, purified, and characterized four different IgG BsAbs or “hetero‐mAbs” using transient CHO expression and stable CHO minipools. Transient CHO titers ranged from 90 to 160 mg/L. Stable CHO titers ranged from 0.4 to 2.3 g/L. Following a simple Protein A purification step, the percentage of correctly paired BsAbs ranged from 74% to 98% as determined by mass spectrometry. We also found that information generated from transient CHO expression was similar to information generated using stable CHO minipools. In conclusion, the quad vector approach represents a simple, but effective, alternative approach for the generation of IgG BsAbs in both transient CHO and stable CHO expression systems.


Methods | 2018

Building blocks for bispecific and trispecific antibodies

Xiufeng Wu; Stephen J. Demarest

Bispecific antibodies (BsAbs), which target two antigens or epitopes, incorporate the specificities and properties of two distinct monoclonal antibodies (mAbs) into a single molecule. As such, BsAbs can elicit synergistic activities and provide the capacity for enhanced therapeutic efficacy and/or safety compared to what can be achieved with conventional monospecific IgGs. There are many building block formats to generate BsAbs and Trispecific antibodies (TsAbs) based on combining the antigen recognition domains of monoclonal antibodies (mAbs). This review describes the many and varied antibody-based building blocks used to achieve multivalency and multispecificity. These diverse building blocks provide opportunities to tailor the design of BsAbs and TsAbs to match the desired applications.


Journal of Immunotherapy | 2004

Generation, and Characterization of Two Tetravalent CH2 Domain-Deleted CC49 Mabs

Xiufeng Wu; Jennifer Hopp; Daniel Perret; Julie Chung; Mitchell Reff; Scott Glaser

of anti-CD3 antibody. Moreover, B7.1/NHS76 fusion protein was found to retain its antigen-binding capacity and good uptake in tumors. To assess their in vivo functions, both fusion proteins were evaluated in murine Colon 26 tumor model. When B7.1/NHS76 (30ug/dose) was administered iv daily3 5 into BALB/c mice bearing 0.5cm sc tumors, a 60% reduction in average tumor size was observed compared to control treated mice at day 19 post-implantation. In contrast, when B7.1/Fc (20ug/dose) was administered in the same protocol, 80% mice underwent complete tumor regression. The therapeutic effects of both fusion proteins were abrogated by depletion of CD8 T cells indicating that this lymphoid subset was instrumental. However, when antisera to CD4 or CD25 Tcell subsets were used to deplete these subsets, the tumors underwent complete regression. Dramatic changes in the infiltration and activation of T-cells could be seen in tumor draining lymph nodes by FACS in the combination therapy groups. In vivo treatment studies using other tumor cell lines and re-challenge studies on previous tumor-free mice are currently in progress. These studies suggest that human B7.1 fusion proteins might be promising antitumor reagents in patients especially when used in combination with methods to delete T-regulatory cells.


Archive | 2007

Stabilized polypeptide compositions

Scott Glaser; Stephen J. Demarest; Brian Robert Miller; William B. Snyder; Xiufeng Wu; Norman Wang; Lisa J. Croner; Alexey Lugovskoy

Collaboration


Dive into the Xiufeng Wu'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge