Network


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

Hotspot


Dive into the research topics where Bing-Mae Chen is active.

Publication


Featured researches published by Bing-Mae Chen.


Journal of Immunology | 2010

Cutting Edge: Mechanical Forces Acting on T Cells Immobilized via the TCR Complex Can Trigger TCR Signaling

Ya-Chen Li; Bing-Mae Chen; Pei-Chun Wu; Tian-Lu Cheng; Lung-Sen Kao; Mi-Hua Tao; André Lieber; Steve R. Roffler

Engagement of the TCR by antigenic peptides presented by the MHC activates specific T cells to control infections. Recent theoretical considerations have suggested that mechanical forces acting on the TCR may be important for receptor triggering. In this study, we directly tested the hypothesis that physical forces acting on the TCR can initiate signaling in T cells by micromanipulation of individual T cells bound to artificial APCs expressing engineered TCR ligands. We find that mechanical forces acting on T cells bound to APCs via the TCR complex but not other surface receptors can initiate signaling in T cells in an Src kinase-dependent fashion. Our data indicate that T cells are mechanically sensitive when coupled to APCs by the TCR and indicates that the TCR may act as a mechanosensor. Our data provide new insight into TCR function.


Cancer Immunology, Immunotherapy | 1997

Poly(ethylene glycol) modification of β-glucuronidase-antibody conjugates for solid-tumor therapy by targeted activation of glucuronide prodrugs

Tian-Lu Cheng; Bing-Mae Chen; Lai-Yee Chan; Pin-Yi Wu; Ji-Wang Chern; Steve R. Roffler

Abstract Methoxypoly(ethylene glycol) (PEG) modification of Escherichia coliβ-glucuronidase (βG) was examined as a method to improve the stability and pharmacokinetics of antibody-βG conjugates for the targeted activation of glucuronide prodrugs at tumor cells. Introduction of 3 PEG molecules did not affect βG activity whereas higher degrees of PEG modification produced progressively greater loss of enzymatic activity. The enzyme was found to be stable in serum regardless of PEG modification. PEG-modified βG was coupled via a thioether bond to mAb RH1, an IgG2a antibody that binds to the surface of AS-30D hepatoma cells, to produce conjugates with 3 (RH1-βG-3PEG), 5.2 (RH1-βG-5PEG) or 9.8 (RH1-βG-10PEG) PEG molecules per βG with retention of 75%, 45% and 40% of the combined antigen-binding and enzymatic activity of the unmodified conjugate RH1-βG. In contrast to the rapid serum clearance of RH1-βG observed in mice, the PEG-modified conjugates displayed extended serum half-lives. RH1-βG-3PEG and RH1-βG-5PEG also exhibited reduced spleen uptake and greater tumor accumulation than RH1-βG. BHAMG, the glucuronide prodrug of p-hydroxyaniline mustard (pHAM), was relatively nontoxic in vivo. Injection of 6 mg/kg or 12 mg/kg pHAM i.v. depressed white blood cell numbers by 46% and 71% whereas 80 mg/kg BHAMG reduced these levels by 22%. Although the tumor/blood ratio of RH1-βG-5PEG was adversely affected by slow clearance from serum, combined therapy of small solid hepatoma tumors with this conjugate, followed 4 and 5 days later with i.v. injections of BHAMG, cured all of seven mice with severe combined immunodeficiency. Combined treatment with a control antibody-βG conjugate and BHAMG delayed tumor growth and cured two of six mice while treatment with pHAM or BHAMG alone was ineffective.


Cancer Gene Therapy | 2008

Tumor-targeting prodrug-activating bacteria for cancer therapy

Chiu Min Cheng; Y. L. Lu; Kuo-Hsiang Chuang; W. C. Hung; Jentaie Shiea; Y. C. Su; Chien-Han Kao; Bing-Mae Chen; Steve R. Roffler; Tian-Lu Cheng

Increasing the specificity of chemotherapy may improve the efficacy of cancer treatment. Toward this aim, we developed a strain of bacteria to express enzymes for selective prodrug activation and non-invasive imaging in tumors. β-glucuronidase and the luxCDABE gene cluster were expressed in the DH5α strain of Escherichia coli to generate DH5α-lux/βG. These bacteria emitted light for imaging and hydrolyzed the glucuronide prodrug 9ACG to the topoisomerase I inhibitor 9-aminocamptothecin (9AC). By optical imaging, colony-forming units (CFUs) and staining for βG activity, we found that DH5α-lux/βG preferentially localized and replicated within CL1-5 human lung tumors in mice. The intensity of luminescence, CFU and βG activity increased with time, indicating bacterial replication occurred in tumors. In comparison with DH5α-lux/βG, 9AC or 9ACG treatment, combined systemic administration of DH5α-lux/βG followed by 9ACG prodrug treatment significantly (P<0.005) delayed the growth of CL1-5 tumors. Our results demonstrate that prodrug-activating bacteria may be useful for selective cancer chemotherapy.


International Journal of Cancer | 1997

CURE OF MALIGNANT ASCITES AND GENERATION OF PROTECTIVE IMMUNITY BY MONOCLONAL ANTIBODY-TARGETED ACTIVATION OF A GLUCURONIDE PRODRUG IN RATS

Bing-Mae Chen; Lai-Yee Chan; Shing-Ming Wang; Ming-Fang Wu; Ji-Wang Chern; Steve R. Roffler

We examined the in vivo efficacy of targeting β‐glucuronidase (βG) to activate a glucuronide prodrug (BHAMG) of p‐hydroxyaniline mustard (pHAM) at hepatoma ascites in Sprague‐Dawley rats. Injection i.p. of 500 μg RH1‐βG, a conjugate formed between recombinant βG and monoclonal antibody RH1 with specificity for an antigen expressed on AS‐30D rat hepatoma cells, into rats bearing AS‐30D ascites resulted in the accumulation of 54 μg conjugate per 109 tumor cells after 2 hr. Ascites fluid and serum contained 0.53 and 0 μg/ml, respectively, RH1‐βG 2 hr after injection of the conjugate. Conjugate binding to AS‐30D cells was heterogeneous and non‐saturated, as determined by flow cytometry. BHAMG was less toxic than pHAM to SD rats based on measures of animal mortality, weight loss and hematological toxicity. Treatment of rats bearing established hepatoma ascites with 500 μg RH1‐βG followed 2 hr later with a single i.p. injection of 30 mg/kg BHAMG or 3 i.p. injections of 10 mg/kg BHAMG 2, 3 and 4 hr later resulted in the cure of 6/8 and 8/8 animals, respectively. Treatment with BHAMG or pHAM alone did not produce cures, whereas treatment with a control antibody–βG conjugate and BHAMG produced significantly greater hematological toxicity compared to treatment with RH1‐βG and BHAMG. All cured rats were completely protected from rechallenge with 2 × 107 AS‐30D cells, indicating that successful treatment of animals induced protective immunity. Int. J. Cancer 73:392–402, 1997.


Gene Therapy | 2007

Gene expression imaging by enzymatic catalysis of a fluorescent probe via membrane-anchored β -glucuronidase

Y. C. Su; Kuo-Hsiang Chuang; Yeng-Tseng Wang; Chiu Min Cheng; Shinne-Ren Lin; Jaw-Yuan Wang; Jeng-Jong Hwang; Bing-Mae Chen; Kai-Chuan Chen; Steve R. Roffler; Ta-Chun Cheng

Development of nonimmunogenic and specific reporter genes to monitor gene expression in vivo is important for the optimization of gene therapy protocols. We developed a membrane-anchored form of mouse β-glucuronidase (mβG) as a reporter gene to hydrolyze a nonfluorescent glucuronide probe (fluorescein di-β-D-glucuronide, (FDGlcU) to a highly fluorescent reporter to assess the location and persistence of gene expression. A functional β-glucuronidase (βG) was stably expressed on the surface of murine CT26 colon adenocarcinoma cells where it selectively hydrolyzed the cell-impermeable FDGlcU probe. FDGlcU was also preferentially converted to fluorescent probe by (βG) on CT26 tumors. The fluorescent intensity in βG-expressing CT26 tumors was 240 times greater than the intensity in control tumors. Selective imaging of gene expression was also observed after intratumoral injection of adenoviral βG vector into carcinoma xenografts. Importantly, mβG did not induce an antibody response after hydrodynamic plasmid immunization of Balb/c mice, indicating that the reporter gene product displayed low immunogenicity. A membrane-anchored form of human βG also allowed in vivo imaging, demonstrating that human βG can be employed for imaging. This imaging system therefore, displays good selectivity with low immunogenicity and may help assess the location, magnitude and duration of gene expression in living animals and humans.


International Journal of Cancer | 2001

Potentiation of antitumor immunity by antibody‐directed enzyme prodrug therapy

Bing-Mae Chen; Tian-Lu Cheng; Shey-Cherng Tzou; Steve R. Roffler

Antibody‐directed enzyme prodrug therapy (ADEPT) has displayed antitumor activity in animal models and clinical trials. We examined whether antitumor immunity is generated during ADEPT by employing an immunoenzyme composed of the monoclonal antibody (MAb) RH1 conjugated to β‐glucuronidase to target rat AS‐30D hepatocellular carcinoma tumors. A glucuronide prodrug of p‐hydroxyaniline mustard was used to treat malignant ascites after immunoenzyme localization at the cancer cells. ADEPT cured more than 96% of Sprague‐Dawley rats bearing advanced malignant ascites, and all cured rats were protected from a lethal challenge of AS‐30D cells. Immunization with radiation‐killed AS‐30D cells or AS‐30D cells coated with immunoenzyme did not provide tumor protection. Likewise, ex vivo treatment of tumor cells by ADEPT before injection into rats did not protect against a tumor challenge. AS‐30D and N1‐S1 hepatocellular carcinoma cells but not unrelated syngeneic tumor cells were lysed by peritoneal exudate cells isolated from ADEPT‐cured rats. Depletion of CD8+ but not CD4+ T cells or natural killer (NK) cells reduced the cytolytic activity of peritoneal lymphocytes. ADEPT did not cure tumor‐bearing rats depleted of CD4+ and CD8+ T cells even though it was curative when given 7 days after tumor transplantation in rats with an intact immune system, indicating that ADEPT can synergize with host immunity to increase therapeutic efficacy. These results have important implications for the clinical application of ADEPT.


Bioconjugate Chemistry | 2011

A Humanized Immunoenzyme with Enhanced Activity for Glucuronide Prodrug Activation in the Tumor Microenvironment

Kai-Chuan Chen; Shih-Yen Wu; Yu-Lin Leu; Zeljko M. Prijovich; Bing-Mae Chen; Hsin-Ell Wang; Tian-Lu Cheng; Steve R. Roffler

Antibody-directed enzyme prodrug therapy (ADEPT) utilizing β-glucuronidase is a promising method to enhance the therapeutic index of cancer chemotherapy. In this approach, an immunoenzyme (antibody-β-glucuronidase fusion protein) is employed to selectively activate anticancer glucuronide prodrugs in the tumor microenvironment. A major roadblock to the clinical translation of this therapeutic strategy, however, is the low enzymatic activity and strong immunogenicity of the current generation of immunoenzymes. To overcome this problem, we fused a humanized single-chain antibody (scFv) of mAb CC49 to S2, a human β-glucuronidase (hβG) variant that displays enhanced catalytic activity for prodrug hydrolysis. Here, we show that hcc49-S2 displayed 100-fold greater binding avidity than hcc49 scFv, possessed greater enzymatic activity than wild-type hβG, and more effectively killed antigen-positive cancer cells exposed to an anticancer glucuronide prodrug as compared to an analogous hβG immunoenzyme. Treatment of tumor-bearing mice with hcc49-S2 followed by prodrug significantly delayed tumor growth as compared to hcc49-hβG. Our study shows that hcc49-S2 is a promising targeted enzyme for cancer treatment and demonstrates that enhancement of human enzyme catalytic activity is a powerful approach to improve immunoenzyme efficacy.


Cancer Gene Therapy | 2003

Stable expression of chimeric anti-CD3 receptors on mammalian cells for stimulation of antitumor immunity

Kuang-Wen Liao; Bing-Mae Chen; Tang-Bi Liu; Shey-Cherng Tzou; Ya-Min Lin; Kai-Feng Lin; Chien-I Su; Steve R. Roffler

Expression of CD80 or CD86 costimulatory molecules on tumor cells can produce rejection of immunogenic but not poorly immunogenic tumors. We have previously shown that anti-CD3 single-chain antibodies expressed on the surface of cells can directly activate T cells. We therefore investigated whether anti-CD3 “receptors” could enhance CD86-mediated rejection of poorly immunogenic tumors. Expression of anti-CD3 receptors on cells was increased by introduction of membrane-proximal “spacer” domains containing glycosylation sites between the single-chain antibody and the transmembrane domain of the chimeric receptors. Removal of glycosylation sites in the spacer reduced surface expression due to increased shedding of chimeric receptors from the cell surface. Induction of T-cell proliferation by anti-CD3 receptors did not correlate with the expression level of chimeric protein, but rather depended on the physical properties of the spacer. Anti-CD3 receptors effectively induced T-cell cytotoxicity, whereas coexpression with CD80 or CD86 was required for generating T-cell proliferation and IL-2 secretion. Although expression of CD86 did not significantly delay the growth of poorly immunogenic B16-F1 tumors, expression of anti-CD3 receptors with CD86 produced complete tumor rejections in 50% of mice and induced significant protection against wild-type B16-F1 tumor cells. Our results show that spacer domains can dramatically influence the surface expression and the biological activity of chimeric antibody receptors. The strong antitumor activity produced by anti-CD3 receptors and CD86 on tumor cells indicates that this strategy may be beneficial for the gene-mediated therapy of poorly immunogenic tumors.


Gene Therapy | 2006

A membrane antibody receptor for noninvasive imaging of gene expression

Steve R. Roffler; Hsin-Ell Wang; H.-M. Yu; W.-D. Chang; Chiu-Min Cheng; Yun-Chi Lu; Bing-Mae Chen; Ta-Chun Cheng

Monitoring gene expression is important to optimize gene therapy protocols and ensure that the proper tissue distribution is achieved in clinical practice. We developed a noninvasive imaging system based on the expression of artificial antibody receptors to trap hapten-labeled imaging probes. Functional membrane-bound anti-dansyl antibodies (DNS receptor) were stably expressed on melanoma cells in vitro and in vivo. A bivalent (DNS)2-diethylenetriaminepentaacetic 111Indium probe specifically bound to cells that expressed DNS receptors but not control scFv receptors. Importantly, the 111In probe preferentially localized to DNS receptors but not control receptors on tumors in mice as assessed by gamma camera imaging. By 48 h after intravenous injection, the uptake of the probe in tumors expressing DNS receptors was 72 times greater than the amount of probe in the blood. This targeting strategy may allow noninvasive assessment of the location, extent and persistence of gene expression in living animals and in the clinic.


Cancer Gene Therapy | 2004

Hapten-directed targeting to single-chain antibody receptors.

Tian-Lu Cheng; Kuang-Wen Liao; Shey-Cherng Tzou; Chiu-Min Cheng; Bing-Mae Chen; Steve R. Roffler

Artificial recombinant receptors may be useful for selectively targeting imaging and therapeutic agents to sites of gene expression. To evaluate this approach, we developed transgenes to express highly on cells a single-chain antibody (scFv) against the hapten 4-ethoxymethylene-2-phenyl-2-oxazoline-5-one (phOx). A phOx enzyme conjugate was created by covalently attaching phOx molecules to polyethylene glycol (PEG)-modified β-glucuronidase. Cells expressing phOx scFv but not control scFv receptors were selectively killed after exposure to ß-glucuronidase derivatized with phOx and PEG (phOx-βG-PEG) and a glucuronide prodrug (p-hydroxy aniline mustard β-D-glucuronide, HAMG) of p-hydroxyaniline mustard. Targeted activation of HAMG produced bystander killing of receptor-negative cells in mixed populations containing as few as 10% phOx-receptor-positive cells. Functional phOx scFv receptors were stably expressed on B16-F1 melanoma tumors in vivo. Treatment of mice bearing established phOx-receptor-positive tumors with phOx-βG-PEG and HAMG significantly (P⩽.0005) suppressed tumor growth as compared with treatment with βG-PEG and HAMG or prodrug alone. phOx was unstable in the serum, suggesting alternative haptens may be more suitable for in vivo applications. Our results show that therapeutic agents can be targeted to artificial hapten receptors in vitro and in vivo. The expression of artificial receptors on target cells may allow preferential delivery of therapeutic or imaging molecules to sites of transgene expression.

Collaboration


Dive into the Bing-Mae Chen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chien-Han Kao

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Kuo-Hsiang Chuang

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Shey-Cherng Tzou

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chiu-Min Cheng

National Kaohsiung Marine University

View shared research outputs
Top Co-Authors

Avatar

Hsin-Ell Wang

National Yang-Ming University

View shared research outputs
Top Co-Authors

Avatar

Ji-Wang Chern

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Ta-Chun Cheng

Kaohsiung Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge