28 research outputs found

    Positron emission tomography imaging of endometrial cancer using engineered anti-EMP2 antibody fragments.

    Get PDF
    PurposeAs imaging of the cell surface tetraspan protein epithelial membrane protein-2 (EMP2) expression in malignant tumors may provide important prognostic and predictive diagnostic information, the goal of this study is to determine if antibody fragments to EMP2 may be useful for imaging EMP2 positive tumors.ProceduresThe normal tissue distribution of EMP2 protein expression was evaluated by immunohistochemistry and found to be discretely expressed in both mouse and human tissues. To detect EMP2 in tumors, a recombinant human anti-EMP2 minibody (scFv-hinge-C(H)3 dimer; 80 kDa) was designed to recognize a common epitope in mice and humans and characterized. In human tumor cell lines, the antibody binding induced EMP2 internalization and degradation, prompting the need for a residualizing imaging strategy. Following conjugation to DOTA (1,4,7,10-tetraazacyclododecane-N,N',N',N'″-tetraacetic acid), the minibody was radiolabeled with (64)Cu (t (1/2) = 12.7 h) and evaluated in mice as a positron emission tomography (PET) imaging agent for human EMP2-expressing endometrial tumor xenografts.ResultsThe residualizing agent, (64)Cu-DOTA anti-EMP2 minibody, achieved high uptake in endometrial cancer xenografts overexpressing EMP2 (10.2 ± 2.6, percent injected dose per gram (%ID/g) ± SD) with moderate uptake in wild-type HEC1A tumors (6.0 ± 0.1). In both cases, precise tumor delineation was observed from the PET images. In contrast, low uptake was observed with anti-EMP2 minibodies in EMP2-negative tumors (1.9 ± 0.5).ConclusionsThis new immune-PET agent may be useful for preclinical assessment of anti-EMP2 targeting in vivo. It may also have value for imaging of tumor localization and therapeutic response in patients with EMP2-positive malignancies

    Rationale and preclinical efficacy of a novel anti-EMP2 antibody for the treatment of invasive breast cancer

    Get PDF
    Despite significant advances in biology and medicine, the incidence and mortality due to breast cancer worldwide is still unacceptably high. Thus, there is an urgent need to discover new molecular targets. In this article, we show evidence for a novel target in human breast cancer, the tetraspan protein epithelial membrane protein-2 (EMP2). Using tissue tumor arrays, protein expression of EMP2 was measured and found to be minimal in normal mammary tissue, but it was upregulated in 63% of invasive breast cancer tumors and in 73% of triple-negative tumors tested. To test the hypothesis that EMP2 may be a suitable target for therapy, we constructed a fully human immunoglobulin G1 (IgG1) antibody specific for a conserved domain of human and murine EMP2. Treatment of breast cancer cells with the anti-EMP2 IgG1 significantly inhibited EMP2-mediated signaling, blocked FAK/Src signaling, inhibited invasion, and promoted apoptosis in vitro. In both human xenograft and syngeneic metastatic tumor monotherapy models, anti-EMP2 IgG1 retarded tumor growth without detectable systemic toxicity. This antitumor effect was, in part, attributable to a potent antibody-dependent cell-mediated cytotoxicity response as well as direct cytotoxicity induced by the monoclonal antibody. Together, these results identify EMP2 as a novel therapeutic target for invasive breast cancer.Fil: Fu, Maoyong. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Maresh , Erin L.. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Helguera, Gustavo Fernando. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental (i); Argentina. Universidad de Buenos Aires; ArgentinaFil: Kiyohara, Meagan. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Qin, yu. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Ashki, Negin. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Daniels Wells, Tracy R.. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Aziz, Najib. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Gordon, Lynn K.. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Braun, Jonathan. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Elshimali, Yahya. Charles Drew University. Department of Pathology; Estados UnidosFil: Soslow, Robert A.. Memorial Sloan-Kettering Cancer Center. Department of Pathology; Estados UnidosFil: Penichet, Manuel L.. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Goodglick, Lee. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados UnidosFil: Wadehra, Madhuri. University of California Los Angeles. David Geffen School of Medicine at UCLA; Estados Unido

    Epithelial Membrane Protein-2 Promotes Endometrial Tumor Formation through Activation of FAK and Src

    Get PDF
    Endometrial cancer is the most common gynecologic malignancy diagnosed among women in developed countries. One recent biomarker strongly associated with disease progression and survival is epithelial membrane protein-2 (EMP2), a tetraspan protein known to associate with and modify surface expression of certain integrin isoforms. In this study, we show using a xenograft model system that EMP2 expression is necessary for efficient endometrial tumor formation, and we have started to characterize the mechanism by which EMP2 contributes to this malignant phenotype. In endometrial cancer cells, the focal adhesion kinase (FAK)/Src pathway appears to regulate migration as measured through wound healing assays. Manipulation of EMP2 levels in endometrial cancer cells regulates the phosphorylation of FAK and Src, and promotes their distribution into lipid raft domains. Notably, cells with low levels of EMP2 fail to migrate and poorly form tumors in vivo. These findings reveal the pivotal role of EMP2 in endometrial cancer carcinogenesis, and suggest that the association of elevated EMP2 levels with endometrial cancer prognosis may be causally linked to its effect on integrin-mediated signaling
    corecore