Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 16(2): 497-508, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068083

ABSTRACT

PURPOSE: Targeted therapeutics have significantly changed the outcome for patients diagnosed with cancer. Still, effective therapeutic intervention does not exist for many cancers and much remains to be done. The objective of this study was to identify novel genes that potentially regulate tumor growth, to target these gene products with monoclonal antibodies, and to examine the therapeutic potential of these antibodies. EXPERIMENTAL DESIGN: Using cDNA microarray analysis, we identified genes overexpressed in several solid malignancies. We generated a mouse monoclonal antibody, 19.2.1, and its humanized counterpart, PDL192, to one such target, TweakR (TWEAK receptor, Fn14, TNFRSF12A, CD266), and characterized the antitumor activities in vitro and in mouse xenograft models. RESULTS: Both 19.2.1 (mouse IgG2a) and PDL192 (human IgG1), like TWEAK, the natural ligand of TweakR, inhibited the growth of several TweakR-expressing cancer cell lines in anchorage-dependent and anchorage-independent assays in vitro. Both antibodies showed significant antitumor activity in multiple mouse xenograft models. PDL192 and 19.2.1 also induced antibody-dependent cellular cytotoxicity (ADCC) of cancer cell lines in vitro. A chimeric version of 19.2.1 containing the mouse IgG1 Fc region (19.2.1 x G1) exhibited significantly less ADCC than 19.2.1. However, 19.2. 1x G1 showed differential activity in vivo, with activity equivalent to 19.2.1 in one model, but significantly less efficacy than 19.2.1 in a second model. These results indicate that PDL192 and 19.2.1 mediate their antitumor effects by signaling through TweakR, resulting in reduced tumor cell proliferation, and by ADCC.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Cell Proliferation/drug effects , Neoplasms/pathology , Tumor Necrosis Factors/immunology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Cytokine TWEAK , Dose-Response Relationship, Drug , Humans , Immunotherapy/methods , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Neoplasm Metastasis , Neoplasms/therapy , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
J Exp Ther Oncol ; 5(4): 273-86, 2006.
Article in English | MEDLINE | ID: mdl-17024968

ABSTRACT

Integrin alpha5beta1, the principal fibronectin receptor, is an important survival factor, playing a key role in angiogenesis. Angiogenesis is critical for tumor growth, and anti-angiogenic therapies have met clinical success. To validate the therapeutic potential of an anti-alpha5beta1 strategy, we generated volociximab (M200) a chimeric human IgG4 version of the alpha5beta1 function-blocking murine antibody IIA1; and F200, the Fab derivative. Volociximab, F200 and IIA1 showed similar activity by ELISA (EC50= 0.2nM), Biacore (Kd= 0.1-0.4nM) and inhibition of fibronectin binding (IC50= 2-3nM). The inhibitory potential of alpha5beta1 antibodies was compared to HuMV833, an anti-VEGF antibody. Both volociximab and HuMV833 inhibited HUVEC proliferation (IC50 of volociximab = 0.2-0.5nM; IC50 of HuMV833 = 45nM). However, IIA1, volociximab and F200 were also potent inhibitors of an in vitro model of angiogenesis (HUVEC tube formation assay), unlike HuMV833. Additionally, volociximab inhibited in vitro tube formation induced by VEGF and/or bFGF, suggesting a mechanism of action independent of growth factor stimulus. In fact, inhibition of alpha5beta1 function by volociximab induced apoptosis of actively proliferating, but not resting, endothelial cells. Volociximab does not cross-react with rodent alpha5beta1, therefore in vivo validation of an anti-alpha5beta1 approach was conducted in a cynomolgus model of choroidal revascularization. Volociximab and F200 were potent inhibitors of neovessel formation in this model. These data demonstrate that volociximab has therapeutic potential in diseases in which new vessel formation is a component of the pathology.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies/therapeutic use , Drug Evaluation, Preclinical , Integrin alpha5beta1/immunology , Animals , Antibodies, Monoclonal, Murine-Derived , COS Cells , Chlorocebus aethiops , Extracellular Matrix/metabolism , Humans , Inhibitory Concentration 50 , Integrin alpha5beta1/chemistry , Kinetics , Macaca fascicularis , Macular Degeneration/drug therapy , Neovascularization, Pathologic , Rituximab
3.
Mol Cancer Ther ; 3(8): 921-32, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15299075

ABSTRACT

Current treatments for advanced stage, hormone-resistant prostate cancer are largely ineffective, leading to high patient mortality and morbidity. To fulfill this unmet medical need, we used global gene expression profiling to identify new potential antibody-drug conjugate (ADC) targets that showed maximal prostate cancer-specific expression. TMEFF2, a gene encoding a plasma membrane protein with two follistatin-like domains and one epidermal growth factor-like domain, had limited normal tissue distribution and was highly overexpressed in prostate cancer. Immunohistochemistry analysis using a specific monoclonal antibody (mAb) to human TMEFF2 showed significant protein expression in 74% of primary prostate cancers and 42% of metastatic lesions from lymph nodes and bone that represented both hormone-naïve and hormone-resistant disease. To evaluate anti-TMEFF2 mAbs as potential ADCs, one mAb was conjugated to the cytotoxic agent auristatin E via a cathepsin B-sensitive valine-citrulline linker. This ADC, Pr1-vcMMAE, was used to treat male severe combined immunodeficient mice bearing xenografted LNCaP and CWR22 prostate cancers expressing TMEFF2. Doses of 3 to 10 mg/kg of this specific ADC resulted in significant and sustained tumor growth inhibition, whereas an isotype control ADC had no significant effect. Similar efficacy and specificity was shown with huPr1-vcMMAE, a humanized anti-TMEFF2 ADC. No overt in vivo toxicity was observed with either murine or human ADC, despite significant cross-reactivity of anti-TMEFF2 mAb with the murine TMEFF2 protein, implying minimal toxicity to other body tissues. These data support the further evaluation and clinical testing of huPr1-vcMMAE as a novel therapeutic for the treatment of metastatic and hormone-resistant prostate cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Membrane Proteins/chemistry , Membrane Proteins/immunology , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Oligopeptides/therapeutic use , Prostatic Neoplasms/drug therapy , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Brain/metabolism , Cell Membrane/metabolism , Cell Proliferation , Cloning, Molecular , DNA, Complementary/metabolism , Flow Cytometry , Follistatin/chemistry , Humans , Hybridomas/chemistry , Immunohistochemistry , Kinetics , Lymphatic Metastasis , Male , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Oligopeptides/chemistry , Prostate/metabolism , Prostatic Neoplasms/metabolism , Protein Structure, Tertiary , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Surface Plasmon Resonance , Time Factors , Transfection
4.
Cancer Res ; 63(19): 6387-94, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14559828

ABSTRACT

We have used the Eos Hu03 GeneChip array, which represents over 92% of the transcribed human genome, to measure gene expression in a panel of normal and diseased human tissues. This analysis revealed that E-selectin mRNA is selectively overexpressed in prostate cancer epithelium, a finding that correlated strongly with E-selectin protein expression as assessed by immunohistochemistry. Antibodies against E-selectin that blocked function failed to impede cancer cell growth, suggesting that overexpression of E-selectin was not essential for cell growth. However, a novel auristatin E-based antibody drug conjugate (ADC), E-selectin antibody valine-citrulline monomethyl-auristatin E, was a potent and selective agent against E-selectin-expressing cancer cell lines in vitro, with the degree of cytotoxicity varying with surface antigen density. Interestingly, sensitivity to the ADC differed among cell lines from different tissues expressing similar amounts of E-selectin and was found to correlate with sensitivity to free auristatin E. Furthermore, E-selectin-expressing tumors grown as xenografts in severe combined immunodeficient mice were responsive to treatment with E-selectin antibody valine-citrulline monomethyl-auristatin E in vivo, with more than 85% inhibition of tumor growth observed in treated mice. These findings demonstrate that an E-selectin-targeting ADC has potential as a prostate cancer therapy and validates a genomics-based paradigm for the identification of cancer-specific antigens suitable for targeted therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , E-Selectin/biosynthesis , Immunotoxins/metabolism , Oligopeptides/administration & dosage , Prostatic Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody Specificity , E-Selectin/genetics , E-Selectin/immunology , Gene Expression Regulation, Neoplastic , Humans , Immunotoxins/immunology , Immunotoxins/pharmacology , Male , Mice , Mice, SCID , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...