Yen Phung
Laboratory of Molecular Biology
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Yen Phung.
Hepatology | 2014
Wei Gao; Heungnam Kim; Mingqian Feng; Yen Phung; Charles P. Xavier; Jeffrey S. Rubin; Mitchell Ho
Wnt signaling is important for cancer pathogenesis and is often up‐regulated in hepatocellular carcinoma (HCC). Heparan sulfate proteoglycans (HSPGs) function as coreceptors or modulators of Wnt activation. Glypican‐3 (GPC3) is an HSPG that is highly expressed in HCC, where it can attract Wnt proteins to the cell surface and promote cell proliferation. Thus, GPC3 has emerged as a candidate therapeutic target in liver cancer. While monoclonal antibodies to GPC3 are currently being evaluated in preclinical and clinical studies, none have shown an effect on Wnt signaling. Here, we first document the expression of Wnt3a, multiple Wnt receptors, and GPC3 in several HCC cell lines, and demonstrate that GPC3 enhanced the activity of Wnt3a/β‐catenin signaling in these cells. Then we report the identification of HS20, a human monoclonal antibody against GPC3, which preferentially recognized the heparan sulfate chains of GPC3, both the sulfated and nonsulfated portions. HS20 disrupted the interaction of Wnt3a and GPC3 and blocked Wnt3a/β‐catenin signaling. Moreover, HS20 inhibited Wnt3a‐dependent cell proliferation in vitro and HCC xenograft growth in nude mice. In addition, HS20 had no detectable undesired toxicity in mice. Taken together, our results show that a monoclonal antibody primarily targeting the heparin sulfate chains of GPC3 inhibited Wnt/β‐catenin signaling in HCC cells and had potent antitumor activity in vivo. Conclusion: An antibody directed against the heparan sulfate of a proteoglycan shows efficacy in blocking Wnt signaling and HCC growth, suggesting a novel strategy for liver cancer therapy. (Hepatology 2014;60:576–587)
PLOS ONE | 2011
Xinran Xiang; Yen Phung; Mingqian Feng; Kunio Nagashima; Jingli Zhang; V. Courtney Broaddus; Raffit Hassan; David J. FitzGerald; Mitchell Ho
BACKGROUND Tumor microenvironments present significant barriers to penetration by antibodies and immunoconjugates. Tumor microenvironments, however, are difficult to study in vitro. Cells cultured as monolayers exhibit less resistance to therapy than those grown in vivo and an alternative research model more representative of the in vivo tumor is more desirable. SS1P is an immunotoxin composed of the Fv portion of a mesothelin-specific antibody fused to a bacterial toxin that is presently undergoing clinical trials in mesothelioma. METHODOLOGY/PRINCIPAL FINDINGS Here, we examined how the tumor microenvironment affects the penetration and killing activity of SS1P in a new three-dimensional (3D) spheroid model cultured in vitro using the human mesothelioma cell line (NCI-H226) and two primary cell lines isolated from the ascites of malignant mesothelioma patients. Mesothelioma cells grown as monolayers or as spheroids expressed comparable levels of mesothelin; however, spheroids were at least 100 times less affected by SS1P. To understand this disparity in cytotoxicity, we made fluorescence-labeled SS1P molecules and used confocal microscopy to examine the time course of SS1P penetration within spheroids. The penetration was limited after 4 hours. Interestingly, we found a significant increase in the number of tight junctions in the core area of spheroids by electron microscopy. Expression of E-Cadherin, a protein involved in the assembly and sealing of tight junctions and highly expressed in malignant mesothelioma, was found significantly increased in spheroids as compared to monolayers. Moreover, we found that siRNA silencing and antibody inhibition targeting E-Cadherin could enhance SS1P immunotoxin therapy in vitro. CONCLUSION/SIGNIFICANCE This work is one of the first to investigate immunotoxins in 3D tumor spheroids in vitro. This initial description of an in vitro tumor model may offer a simple and more representative model of in vivo tumors and will allow for further investigations of the microenvironmental effects on drug penetration and tumor cell killing. We believe that the methods developed here may apply to the studies of other tumor-targeting antibodies and immunoconjugates in vitro.
Molecular Cancer Therapeutics | 2013
Zhewei Tang; Mingqian Feng; Wei Gao; Yen Phung; Weizao Chen; Amit Chaudhary; Brad St. Croix; Min Qian; Dimiter S. Dimitrov; Mitchell Ho
Monoclonal antibodies against mesothelin are being evaluated for the treatment of mesothelioma and multiple forms of cancers, and show great promise for clinical development for solid cancers. Antibodies against mesothelin have been shown to act via immunotoxin-based inhibition of tumor growth and induction of antibody-dependent cell-mediated cytotoxicity (ADCC). However, complement-dependent cytotoxicity (CDC), considered an important additional mechanism of therapeutic antibodies against tumors, is inactive for such antibodies. Here, we used phage display antibody engineering technology and synthetic peptide screening to identify SD1, a human single-domain antibody to mesothelin. SD1 recognizes a conformational epitope at the C-terminal end (residues 539–588) of mesothelin close to the cell surface. To investigate SD1 as a potential therapeutic agent, we generated a recombinant human Fc (SD1-hFc) fusion protein. Interestingly, the SD1-hFc protein exhibits strong CDC activity, in addition to ADCC, against mesothelin-expressing tumor cells. Furthermore, it causes growth inhibition of human tumor xenografts in nude mice as a single agent. SD1 is the first human single-domain antibody targeting mesothelin-expressing tumors, shows potential as a cancer therapeutic candidate, and may improve current antibody therapy targeting mesothelin-expressing tumors. Mol Cancer Ther; 12(4); 416–26. ©2013 AACR.
mAbs | 2012
Yen Phung; Wei Gao; Yan-Gao Man; Satoshi Nagata; Mitchell Ho
Isolating high-affinity antibodies against native tumor antigens on the cell surface is not straightforward using standard hybridoma procedures. Here, we describe a combination method of synthetic peptide immunization and high-throughput flow cytometry screening to efficiently isolate hybridomas for cell binding. Using this method, we identified high-affinity monoclonal antibodies specific for the native form of glypcian-3 (GPC3), a target heterogeneously expressed in hepatocellular carcinoma (HCC) and other cancers. We isolated a panel of monoclonal antibodies (YP6, YP7, YP8, YP9 and YP9.1) for cell surface binding. The antibodies were used to characterize GPC3 protein expression in human liver cancer cell lines and tissues by flow cytometry, immunoblotting and immunohistochemistry. The best antibody (YP7) bound cell surface-associated GPC3 with equilibrium dissociation constant, KD = 0.3 nmol/L and was highly specific for HCC, not normal tissues or other forms of primary liver cancers (such as cholangiocarcinoma). Interestingly, the new antibody was highly sensitive in that it detected GPC3 in low expression ovarian clear cell carcinoma and melanoma cells. The YP7 antibody exhibited significant HCC xenograft tumor growth inhibition in nude mice. These results describe an improved method for producing high-affinity monoclonal antibodies to cell surface tumor antigens and represent a general approach to isolate therapeutic antibodies against cancer. The new high-affinity antibodies described here have significant potential for GPC3-expressing cancer diagnostics and therapy.
Molecular Pharmaceutics | 2015
Hirofumi Hanaoka; Tadanobu Nagaya; Kazuhide Sato; Yuko Nakamura; Rira Watanabe; Toshiko Harada; Wei Gao; Mingqian Feng; Yen Phung; Insook Kim; Chang H. Paik; Peter L. Choyke; Mitchell Ho; Hisataka Kobayashi
Glypican-3 (GPC3) represents an attractive target for hepatocellular carcinoma (HCC) therapy because it is highly expressed in HCC but not in adult normal tissue. Recently, high affinity anti-GPC3 antibodies have been developed; however, full antibodies may not penetrate evenly into tumor parenchyma, reducing their effectiveness. In this study, we compared a whole IgG antibody, anti-GPC3 YP7, with an anti-GPC3 human heavy chain antibody, HN3, with regard to their relative therapeutic effects. Both YP7 and HN3 bound to GPC3-positive A431/G1 cells and were internalized by the cells by in vitro evaluation with (125)I- and (111)In-radiolabeling antibodies. In vivo biodistribution and tumor accumulation was performed with (111)In-labeled antibodies, and intratumoral microdistribution was evaluated using fluorescently labeled antibodies (IR700). HN3 showed similar high tumor accumulation but superior homogeneity within the tumor compared with YP7. Using the same IR700 conjugated antibodies photoimmunotherapy (PIT) was performed in vitro and in a tumor-bearing mouse model in vivo. PIT with IR700-HN3 and IR700-YP7 demonstrated that comparable results could be achieved despite of low reaccumulation 24 h after the first NIR light exposure. These results indicated that a heavy-chain antibody, HN3, showed more favorable characteristics than YP7, a conventional IgG, as a therapeutic antibody platform for designing molecularly targeted agents against HCC.
PLOS ONE | 2012
Heungnam Kim; Yen Phung; Mitchell Ho
Tumor microenvironments present significant barriers to anti-tumor agents. Molecules involved in multicellular tumor microenvironments, however, are difficult to study ex vivo. Here, we generated a matrix-free tumor spheroid model using the NCI-H226 mesothelioma cell line and compared the gene expression profiles of spheroids and monolayers using microarray analysis. Microarray analysis revealed that 142 probe sets were differentially expressed between tumor spheroids and monolayers. Gene ontology analysis revealed that upregulated genes were primarily related to immune response, wound response, lymphocyte stimulation and response to cytokine stimulation, whereas downregulated genes were primarily associated with apoptosis. Among the 142 genes, 27 are located in the membrane and related to biologic processes of cellular movement, cell-to-cell signaling, cellular growth and proliferation and morphology. Western blot analysis validated elevation of MMP2, BAFF/BLyS/TNFSF13B, RANTES/CCL5 and TNFAIP6/TSG-6 protein expression in spheroids as compared to monolayers. Thus, we have reported the first large scale comparison of the transcriptional profiles using an ex vivo matrix-free spheroid model to identify genes specific to the three-dimensional biological structure of tumors. The method described here can be used for gene expression profiling of tumors other than mesothelioma.
Scientific Reports | 2015
Yi-Fan Zhang; Yen Phung; Wei Gao; Seiji Kawa; Raffit Hassan; Ira Pastan; Mitchell Ho
Mesothelin is an emerging cell surface target in mesothelioma and other solid tumors. Most antibody drug candidates recognize highly immunogenic Region I (296–390) on mesothelin. Here, we report a group of high-affinity non-Region I rabbit monoclonal antibodies. These antibodies do not compete for mesothelin binding with the immunotoxin SS1P that binds Region I of mesothelin. One pair of antibodies (YP218 and YP223) is suitable to detect soluble mesothelin in a sandwich ELISA with high sensitivity. The new assay can also be used to measure serum mesothelin concentration in mesothelioma patients, indicating its potential use for monitoring patients treated with current antibody therapies targeting Region I. The antibodies are highly specific and sensitive in immunostaining of mesothelioma. To explore their use in tumor therapy, we have generated the immunotoxins based on the Fv of these antibodies. One immunotoxin (YP218 Fv-PE38) exhibits potent anti-tumor cytotoxicity towards primary mesothelioma cell lines in vitro and an NCI-H226 xenograft tumor in mice. Furthermore, we have engineered a humanized YP218 Fv that retains full binding affinity for mesothelin-expressing cancer cells. In conclusion, with their unique binding properties, these antibodies may be promising candidates for monitoring and treating mesothelioma and other mesothelin-expressing cancers.
Nanomedicine: Nanotechnology, Biology and Medicine | 2015
Hirofumi Hanaoka; Takahito Nakajima; Kazuhide Sato; Rira Watanabe; Yen Phung; Wei Gao; Toshiko Harada; Insook Kim; Chang H. Paik; Peter L. Choyke; Mitchell Ho; Hisataka Kobayashi
AIM Effectiveness of Glypican-3 (GPC3)-targeted photoimmunotherapy (PIT) combined with the nanoparticle albumin-bound paclitaxel (nab-paclitaxel) for hepatocellular carcinoma was evaluated. MATERIALS & METHODS GPC3 expressing A431/G1 cells were incubated with a phthalocyanine-derivative, IRDye700DX (IR700), conjugated to an anti-GPC3 antibody, IR700-YP7 and exposed to near-infrared light. Therapeutic experiments combining GPC3-targeted PIT with nab-paclitaxel were performed in A431/G1 tumor-bearing mice. RESULTS IR700-YP7 bound to A431/G1 cells and induced rapid target-specific necrotic cell death by near-infrared light exposure in vitro. IR700-YP7 accumulated in A431/G1 tumors. Tumor growth was inhibited by PIT compared with nontreated control. Additionally, PIT dramatically increased nab-paclitaxel delivery and enhanced the therapeutic effect. CONCLUSION PIT targeting GPC3 combined with nab-paclitaxel is a promising method for treating hepatocellular carcinoma.
Cancer Research | 2010
Maria Xiang; Yen Phung; Mitchell Ho
The formation of tumor cells traditionally cultured in vitro as monolayers frequently exhibit less drug resistance than tumors in vivo and are inadequate models for screening anti-cancer agents. Mesothelioma, a highly fatal asbestos-induced cancer of the mesothelium, is notoriously resistant to traditional chemotherapy and radiation. In this study, the development of multicellular and apoptotic resistance in human mesothelioma cell lines is examined using a 3D spheroid model cultured in vitro, which more closely depicts in vivo tumor resistance. We hypothesized that poor drug penetration of spheroid tumors is associated with the number of cellular contacts related to its 3D shape. In addition, we asked whether its resistance to cell death was directly related to the Bcl-2-regulated mitochondrial pathway. To test this, we cultured tumors cells both as monolayers and spheroids and measured the concentration of a fluorescently-labeled immunotoxin, SS1P, as a percentage of live cells. Antigen expression was determined via flow cytometry, and tumors were visualized using transmission and scanning electron microscopy, where a higher number of tight junctions were found in the core area of the spheroid. Additionally, we analyzed the expression of several proteins related to apoptosis in cell lysates for both monolayers and spheroids by western blotting. We found that some human mesothelioma cells form spheroids within 24 hours in the absence of Matrigel. The expression levels of mesothelin and CA125 - two established tumor antigens - are comparable between mesothelioma cells obtained from monolayer and spheroid; however, the cytotoxicity assay (IC50) of the anti-mesothelin immunotoxin SS1P shows that it is at least 100 times less active for spheroids than for monolayers. SS1P also shows incomplete penetration in spheroids, with limited penetration after 4 to 8 hours. Regarding cellular contacts, E-Cadherin is expressed in both monolayers and spheroids, but is significantly increased in spheroids. However, no bands are seen for prosurvival protein Bcl-2, and no difference in expression is found for prosurvival protein MCL-1. As for pro-apoptotic molecules, Bax and Bak, Bax did not show any changes, whereas Bak appeared only in monolayer and was absent for spheroid. Our results indicate that the upregulation of E-Cadherin expression in spheroids may be related to the increased number of tight junctions, thereby inhibiting the penetration of immunotoxins. Likewise, downregulation of the pro-apoptotic molecule Bak may cause resistance to immunotoxin-induced mitochondria-dependent apoptosis in spheroids. We conclude that using a biologically relevant 3D spheroid model more closely resembles similar types of resistance to that of in vivo tumors. We believe that it is a useful in vitro tumor model that will provide greater opportunity for investigating and screening antibody therapeutics. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 648.
Journal of Cancer | 2011
Yen Phung; Dario Barbone; V. Courtney Broaddus; Mitchell Ho