Network


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

Hotspot


Dive into the research topics where Wei-Ching Liang is active.

Publication


Featured researches published by Wei-Ching Liang.


Journal of Biological Chemistry | 2006

Cross-species Vascular Endothelial Growth Factor (VEGF)-blocking Antibodies Completely Inhibit the Growth of Human Tumor Xenografts and Measure the Contribution of Stromal VEGF

Wei-Ching Liang; Xiumin Wu; Franklin Peale; Chingwei V. Lee; Y. Gloria Meng; Johnny Gutierrez; Ling Fu; Ajay K. Malik; Hans-Peter Gerber; Napoleone Ferrara; Germaine Fuh

To fully assess the role of VEGF-A in tumor angiogenesis, antibodies that can block all sources of vascular endothelial growth factor (VEGF) are desired. Selectively targeting tumor-derived VEGF overlooks the contribution of host stromal VEGF. Other strategies, such as targeting VEGF receptors directly or using receptor decoys, result in inhibiting not only VEGF-A but also VEGF homologues (e.g. placental growth factor, VEGF-B, and VEGF-C), which may play a role in angiogenesis. Here we report the identification of novel anti-VEGF antibodies, B20 and G6, from synthetic antibody phage libraries, which block both human and murine VEGF action in vitro. Their affinity-improved variants completely inhibit three human tumor xenografts in mice of skeletal muscle, colorectal, and pancreatic origins (A673, HM-7, and HPAC). Avastin, which only inhibits the tumor-derived human VEGF, is ∼90% effective at inhibiting HM-7 and A673 growth but is <50% effective at inhibiting HPAC growth. Indeed, HPAC tumors contain more host stroma invasion and stroma-derived VEGF than other tumors. Thus, the functional contribution of stromal VEGF varies greatly among tumors, and systemic blockade of both tumor and stroma-derived VEGF is sufficient for inhibiting the growth of tumor xenografts.


The EMBO Journal | 2004

VEGF-null cells require PDGFR α signaling-mediated stromal fibroblast recruitment for tumorigenesis

Jianying Dong; Jeremy Grunstein; Max L. Tejada; Frank Peale; Gretchen Frantz; Wei-Ching Liang; Wei Bai; Lanlan Yu; Joe Kowalski; Xiaohuan Liang; Germaine Fuh; Hans-Peter Gerber; Napoleone Ferrara

We generated VEGF‐null fibrosarcomas from VEGF‐loxP mouse embryonic fibroblasts to investigate the mechanisms of tumor escape after VEGF inactivation. These cells were found to be tumorigenic and angiogenic in vivo in spite of the absence of tumor‐derived VEGF. However, VEGF derived from host stroma was readily detected in the tumor mass and treatment with a newly developed anti‐VEGF monoclonal antibody substantially inhibited tumor growth. The functional significance of stroma‐derived VEGF indicates that the recruitment of stromal cells is critical for the angiogenic and tumorigenic properties of these cells. Here we identified PDGF AA as the major stromal fibroblast chemotactic factor produced by tumor cells, and demonstrated that disrupting the paracrine PDGFR α signaling between tumor cells and stromal fibroblasts by soluble PDGFR α‐IgG significantly reduced tumor growth. Thus, PDGFR α signaling is required for the recruitment of VEGF‐producing stromal fibroblasts for tumor angiogenesis and growth. Our findings highlight a novel aspect of PDGFR α signaling in tumorigenesis.


Cancer Cell | 2008

Blocking Neuropilin-2 Function Inhibits Tumor Cell Metastasis

Maresa Caunt; Judy Mak; Wei-Ching Liang; Scott Stawicki; Qi Pan; Raymond K. Tong; Joe Kowalski; Calvin Ho; Hani Bou Reslan; Jed Ross; Leanne Berry; Ian Kasman; Constance Zlot; Zhiyong Cheng; Jennifer Le Couter; Ellen Filvaroff; Greg Plowman; Franklin Peale; Dorothy French; Richard A. D. Carano; Alexander W. Koch; Yan Wu; Ryan J. Watts; Marc Tessier-Lavigne; Anil Bagri

Metastasis, which commonly uses lymphatics, accounts for much of the mortality associated with cancer. The vascular endothelial growth factor (VEGF)-C coreceptor, neuropilin-2 (Nrp2), modulates but is not necessary for developmental lymphangiogenesis, and its significance for metastasis is unknown. An antibody to Nrp2 that blocks VEGFC binding disrupts VEGFC-induced lymphatic endothelial cell migration, but not proliferation, in part independently of VEGF receptor activation. It does not affect established lymphatics in normal adult mice but reduces tumoral lymphangiogenesis and, importantly, functional lymphatics associated with tumors. It also reduces metastasis to sentinel lymph nodes and distant organs, apparently by delaying the departure of tumor cells from the primary tumor. Our results demonstrate that Nrp2, which was originally identified as an axon-guidance receptor, is an attractive target for modulating metastasis.


Journal of Biological Chemistry | 2006

Structure-function studies of two synthetic anti-vascular endothelial growth factor Fabs and comparison with the Avastin Fab.

Germaine Fuh; Ping Wu; Wei-Ching Liang; Mark Ultsch; Chingwei V. Lee; Barbara Moffat; Christian Wiesmann

In the quest to discover new research tools and to develop better agents in the fight against cancer, two antibodies, G6 and B20-4, were isolated from synthetic antibody phage libraries. Unlike the AVASTIN™ antibody, a recently approved agent for the treatment of patients with colorectal cancer, B20-4 and G6 bind and block both human and murine vascular endothelial growth factor (VEGF). Here we have analyzed and compared the binding epitopes on VEGF for these three antibodies using alanine-scanning mutagenesis and structural analyses. The epitopes recognized by both synthetic antibodies are conserved between human and mouse VEGF, and they match closely to the receptor epitopes both structurally and functionally. In contrast, the Avastin epitope overlaps minimally with the receptor binding surface and centers around a residue that is not conserved in mouse. Our structural and functional analyses elucidate the cross-species reactivity of all three antibodies and emphasize the potential advantages of antibody generation using phage display as the resulting antibodies do not depend on sequence differences across species and preferentially target natural protein-protein interaction surfaces.


The EMBO Journal | 2007

Structural studies of neuropilin/antibody complexes provide insights into semaphorin and VEGF binding

Brent A. Appleton; Ping Wu; Janice Maloney; JianPing Yin; Wei-Ching Liang; Scott Stawicki; Kyle Mortara; Krista K. Bowman; J. Michael Elliott; William Desmarais; J. Fernando Bazan; Anil Bagri; Marc Tessier-Lavigne; Alexander W. Koch; Yan Wu; Ryan J. Watts; Christian Wiesmann

Neuropilins (Nrps) are co‐receptors for class 3 semaphorins and vascular endothelial growth factors and important for the development of the nervous system and the vasculature. The extracellular portion of Nrp is composed of two domains that are essential for semaphorin binding (a1a2), two domains necessary for VEGF binding (b1b2), and one domain critical for receptor dimerization (c). We report several crystal structures of Nrp1 and Nrp2 fragments alone and in complex with antibodies that selectively block either semaphorin or vascular endothelial growth factor (VEGF) binding. In these structures, Nrps adopt an unexpected domain arrangement in which the a2, b1, and b2 domains form a tightly packed core that is only loosely connected to the a1 domain. The locations of the antibody epitopes together with in vitro experiments indicate that VEGF and semaphorin do not directly compete for Nrp binding. Based upon our structural and functional data, we propose possible models for ligand binding to neuropilins.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Structural insight into distinct mechanisms of protease inhibition by antibodies

Yan Wu; Charles Eigenbrot; Wei-Ching Liang; Scott Stawicki; Steven Shia; Bin Fan; Rajkumar Ganesan; Michael T. Lipari; Daniel Kirchhofer

To better understand how the relatively flat antigen-combining sites of antibodies interact with the concave shaped substrate-binding clefts of proteases, we determined the structures of two antibodies in complex with the trypsin-like hepatocyte growth-factor activator (HGFA). The two inhibitory antibodies, Ab58 and Ab75, were generated from a human Fab phage display library with synthetic diversity in the three complementarity determining regions (H1, H2, and H3) of the heavy chain, mimicking the natural diversity of the human Ig repertoire. Biochemical studies and the structures of the Fab58:HGFA (3.5-Å resolution) and the Fab75:HGFA (2.2-Å resolution) complexes revealed that Ab58 obstructed substrate access to the active site, whereas Ab75 allosterically inhibited substrate hydrolysis. In both cases, the antibodies interacted with the same protruding element (99-loop), which forms part of the substrate-binding cleft. Ab58 inserted its H1 and H2 loops in the cleft to occupy important substrate interaction sites (S3 and S2). In contrast, Ab75 bound at the backside of the cleft to a region corresponding to thrombin exosite II, which is known to interact with allosteric effector molecules. In agreement with the structural analysis, binding assays with active site inhibitors and enzymatic assays showed that Ab58 is a competitive inhibitor, and Ab75 is a partial competitive inhibitor. These results provide structural insight into antibody-mediated protease inhibition. They suggest that unlike canonical inhibitors, antibodies may preferentially target protruding loops at the rim of the substrate-binding cleft to interfere with the catalytic machinery of proteases without requiring long insertion loops.


Molecular Cancer Therapeutics | 2008

Antixenograft tumor activity of a humanized anti-insulin-like growth factor-I receptor monoclonal antibody is associated with decreased AKT activation and glucose uptake

Yonglei Shang; Yifan Mao; Jennifer Batson; Suzie J. Scales; Gail Lewis Phillips; Mark R. Lackner; Klara Totpal; Simon C. Williams; Jihong Yang; Zhijun Tang; Zora Modrusan; Christine Tan; Wei-Ching Liang; Siao Ping Tsai; Alexander N. Vanderbilt; Kenji Kozuka; Klaus P. Hoeflich; Janet Tien; Sarajane Ross; Congfen Li; Sang Hoon Lee; An Song; Yan Wu; Jean-Philippe Stephan; Avi Ashkenazi; Jiping Zha

The insulin-like growth factor (IGF) system consists of two ligands (IGF-I and IGF-II), which both signal through IGF-I receptor (IGF-IR) to stimulate proliferation and inhibit apoptosis, with activity contributing to malignant growth of many types of human cancers. We have developed a humanized, affinity-matured anti-human IGF-IR monoclonal antibody (h10H5), which binds with high affinity and specificity to the extracellular domain. h10H5 inhibits IGF-IR-mediated signaling by blocking IGF-I and IGF-II binding and by inducing cell surface receptor down-regulation via internalization and degradation, with the extracellular and intracellular domains of IGF-IR being differentially affected by the proteasomal and lysosomal inhibitors. In vitro, h10H5 exhibits antiproliferative effects on cancer cell lines. In vivo, h10H5 shows single-agent antitumor efficacy in human SK-N-AS neuroblastoma and SW527 breast cancer xenograft models and even greater efficacy in combination with the chemotherapeutic agent docetaxel or an anti–vascular endothelial growth factor antibody. Antitumor activity of h10H5 is associated with decreased AKT activation and glucose uptake and a 316-gene transcription profile with significant changes involving DNA metabolic and cell cycle machineries. These data support the clinical testing of h10H5 as a biotherapeutic for IGF-IR-dependent human tumors and furthermore illustrate a new method of monitoring its activity noninvasively in vivo via 2-fluoro-2-deoxy-d-glucose-positron emission tomography imaging. [Mol Cancer Ther 2008;7(9):2599–608]


Cancer Cell | 2007

Blocking Neuropilin-1 Function Has an Additive Effect with Anti-VEGF to Inhibit Tumor Growth

Qi Pan; Yvan Chanthery; Wei-Ching Liang; Scott Stawicki; Judy Mak; Nisha Rathore; Raymond K. Tong; Joe Kowalski; Sharon Yee; Glenn Pacheco; Sarajane Ross; Zhiyong Cheng; Jennifer Le Couter; Greg Plowman; Franklin Peale; Alexander W. Koch; Yan Wu; Anil Bagri; Marc Tessier-Lavigne; Ryan J. Watts


Journal of Molecular Biology | 2004

High-affinity human antibodies from phage-displayed synthetic Fab libraries with a single framework scaffold.

Chingwei V. Lee; Wei-Ching Liang; Mark S. Dennis; Charles Eigenbrot; Sachdev S. Sidhu; Germaine Fuh


Archive | 2006

Binding polypeptides with diversified and consensus vh/vl hypervariable sequences

Mark S. Dennis; Wei-Ching Liang; Yan Wu

Collaboration


Dive into the Wei-Ching Liang'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge