Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 17(2): 521-531, 2018 02.
Article in English | MEDLINE | ID: mdl-29158469

ABSTRACT

Acquired resistance to cetuximab, an antibody that targets the EGFR, impacts clinical benefit in head and neck, and colorectal cancers. One of the mechanisms of resistance to cetuximab is the acquisition of mutations that map to the cetuximab epitope on EGFR and prevent drug binding. We find that necitumumab, another FDA-approved EGFR antibody, can bind to EGFR that harbors the most common cetuximab-resistant substitution, S468R (or S492R, depending on the amino acid numbering system). We determined an X-ray crystal structure to 2.8 Å resolution of the necitumumab Fab bound to an S468R variant of EGFR domain III. The arginine is accommodated in a large, preexisting cavity in the necitumumab paratope. We predict that this paratope shape will be permissive to other epitope substitutions, and show that necitumumab binds to most cetuximab- and panitumumab-resistant EGFR variants. We find that a simple computational approach can predict with high success which EGFR epitope substitutions abrogate antibody binding. This computational method will be valuable to determine whether necitumumab will bind to EGFR as new epitope resistance variants are identified. This method could also be useful for rapid evaluation of the effect on binding of alterations in other antibody/antigen interfaces. Together, these data suggest that necitumumab may be active in patients who are resistant to cetuximab or panitumumab through EGFR epitope mutation. Furthermore, our analysis leads us to speculate that antibodies with large paratope cavities may be less susceptible to resistance due to mutations mapping to the antigen epitope. Mol Cancer Ther; 17(2); 521-31. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cetuximab/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab/pharmacology , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Humans
2.
Neoplasia ; 13(1): 49-59, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21245940

ABSTRACT

The benefits of inhibiting vascular endothelial growth factor (VEGF) signaling in cancer patients are predominantly attributed to effects on tumor endothelial cells. Targeting non-endothelial stromal cells to further impact tumor cell growth and survival is being pursued through the inhibition of additional growth factor pathways important for the survival and/or proliferation of these cells. However, recent data suggest that VEGF receptor (VEGFR)-specific inhibitors may target lymphatic vessels and pericytes in addition to blood vessels. Here, in fact, we demonstrate that DC101 (40 mg/kg, thrice a week), an antibody specific to murine VEGFR2, significantly reduces all three of these stromal components in subcutaneous (SKRC-29) and orthotopic (786-O-LP) models of renal cell carcinoma (RCC) established in nu/nu athymic mice. Sunitinib (40 mg/kg, once daily), a receptor tyrosine kinase inhibitor of VEGFR2 and other growth factor receptors, also caused significant loss of tumor blood vessels in RCC models but had weaker effects than DC101 on pericytes and lymphatic vessels. In combination, sunitinib did not significantly add to the effects of DC101 on tumor blood vessels, lymphatic vessels, or pericytes. Nevertheless, sunitinib increased the effect of DC101 on tumor burden in the SKRC-29 model, perhaps related to its broader specificity. Our data have important implications for combination therapy design, supporting the conclusion that targeting VEGFR2 alone in RCC has the potential to have pleiotropic effects on tumor stroma.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Indoles/pharmacology , Kidney Neoplasms/drug therapy , Pyrroles/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/pathology , Disease Models, Animal , Drug Interactions , Female , Humans , Hypoxia-Inducible Factor 1/biosynthesis , Indoles/therapeutic use , Kidney Neoplasms/blood supply , Kidney Neoplasms/pathology , Lymphatic Vessels/drug effects , Lymphatic Vessels/pathology , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Neovascularization, Pathologic , Pericytes/drug effects , Pericytes/pathology , Pyrroles/therapeutic use , Stromal Cells/drug effects , Stromal Cells/pathology , Sunitinib , Tumor Burden , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-2/immunology , Von Hippel-Lindau Tumor Suppressor Protein/genetics
3.
Neoplasia ; 11(6): 594-604, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19484148

ABSTRACT

Platelet-derived growth factor receptor beta (PDGFRbeta) is upregulated in most of solid tumors. It is expressed by pericytes/smooth muscle cells, fibroblast, macrophage, and certain tumor cells. Several PDGF receptor-related antagonists are being developed as potential antitumor agents and have demonstrated promising antitumor activity in both preclinical and clinical settings. Here, we produced a fully human neutralizing antibody, IMC-2C5, directed against PDGFRbeta from an antibody phage display library. IMC-2C5 binds to both human and mouse PDGFRbeta and blocks PDGF-B from binding to the receptor. IMC-2C5 also blocks ligand-stimulated activation of PDGFRbeta and downstream signaling molecules in tumor cells. In animal studies, IMC-2C5 significantly delayed the growth of OVCAR-8 and NCI-H460 human tumor xenografts in nude mice but failed to show antitumor activities in OVCAR-5 and Caki-1 xenografts. Our results indicate that the antitumor efficacy of IMC-2C5 is primarily due to its effects on tumor stroma, rather than on tumor cells directly. Combination of IMC-2C5 and DC101, an anti-mouse vascular endothelial growth factor receptor 2 antibody, resulted in significantly enhanced antitumor activity in BxPC-3, NCI-H460, and HCT-116 xenografts, compared with DC101 alone, and the trend of additive effects to DC101 treatment in several other tumor models. ELISA analysis of NCI-H460 tumor homogenates showed that IMC-2C5 attenuated protein level of vascular endothelial growth factor and basic fibroblast growth factor elevated by DC101 treatment. Finally, IMC-2C5 showed a trend of additive effects when combined with DC101/chemotherapy in MIA-PaCa-2 and NCI-H460 models. Taken together, these results lend great support to the use of PDGFRbeta antagonists in combination with other antiangiogenic agents in the treatment of a broad range of human cancers.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Receptor, Platelet-Derived Growth Factor beta/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibody Affinity/immunology , Cell Line, Tumor , Drug Synergism , Drug Therapy, Combination , Female , Flow Cytometry , HCT116 Cells , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasms/pathology , Peptide Library , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Biochem Biophys Res Commun ; 357(4): 1142-7, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17462601

ABSTRACT

Platelet-derived growth factor (PDGF) and its receptors (PDGFR) play important roles in tumorigenesis through stimulating tumor growth and promoting angiogenesis via enhancing pericyte recruitment and vessel maturation. Here we produced a neutralizing antibody, 1B3, directed against mouse PDGFRbeta. 1B3 binds to PDGFRbeta with high affinity (9x10(-11)M) and blocks PDGF-BB from binding to the receptor with an IC(50) of approximately 1.2 nM. The antibody also blocks ligand-stimulated activation of PDGFRbeta and downstream signaling molecules, including Akt and MAPK p42/44, in tumor cells. In animal studies, 1B3 significantly enhanced the antitumor and the anti-angiogenic activities of DC101, an antibody directed against mouse vascular endothelial growth factor receptor 2, in a pancreatic (BxPC-3) and a non-small cell lung (NCI-H460) tumor xenograft models. Treatment with the combination of 1B3 and DC101 in BxPC-3 xenograft-bearing mice resulted in tumor regression in 58% of mice compared to that in 18% of mice treated with DC101 alone. Taken together, these results lend great support to use PDGFRbeta antagonists in combinations with other antitumor and/or anti-angiogenic agents in the treatment of a variety of cancers.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Receptor, Platelet-Derived Growth Factor beta/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Antibodies, Monoclonal/immunology , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
5.
J Immunol Methods ; 318(1-2): 65-74, 2007 Jan 10.
Article in English | MEDLINE | ID: mdl-17126853

ABSTRACT

Bispecific antibodies (BsAb) have been traditionally utilized to redirect cytotoxic effector cells and agents to kill tumor cells expressing the target antigens. Recently a new concept is emerging to develop BsAb that simultaneously block the functions of two tumor-associated targets, eg., growth factor receptors, for enhanced antitumor efficacies. Broad clinical applications of BsAb have been, and still are, significantly hampered by the difficulty in producing the materials in sufficient quantity and quality by traditional approaches. Here we describe a recombinant approach for the production of an Fc domain-containing, IgG-like tetravalent BsAb, using a single variable domain (sVD) antibody as a versatile building block. In this method, a sVD of a defined specificity is genetically fused to either the N-terminus of the light chain or the C-terminus of the heavy chain of a functional IgG antibody of a different specificity. A model BsAb was constructed using a sVD to mouse platelet derived growth factor receptor alpha and a conventional IgG antibody to mouse platelet derived growth factor receptor beta. The BsAb were expressed in mammalian cells and purified to homogeneity by a one-step Protein A affinity chromatography. Further, the BsAb retained the antigen binding specificity and the receptor neutralizing activity of both of its parent antibodies. Importantly, the BsAb inhibited the activation of both its target receptors in tumor cells stimulated by both platelet derived growth factor AA and BB, whereas the parent monospecific antibody only inhibited the activation of a single receptor stimulated by its cognate ligand. This format of BsAb should be readily applicable to the production of other BsAb recognizing any pairs of antigens.


Subject(s)
Antibodies, Bispecific/immunology , Immunoglobulin Variable Region/genetics , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacology , Antibody Affinity/immunology , Antibody Specificity/immunology , Becaplermin , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Electrophoresis, Polyacrylamide Gel , Genetic Vectors/genetics , Humans , Immunoglobulin G/immunology , Immunoglobulin Variable Region/immunology , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/immunology , Kinetics , Mice , Models, Molecular , Phosphorylation/drug effects , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/immunology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/immunology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Transfection
6.
J Biol Chem ; 281(16): 10706-14, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16481314

ABSTRACT

Both laboratory and early clinical studies to date have demonstrated that bispecific antibodies (BsAb) may have potentially significant application in cancer therapy. The clinical development of BsAb as therapeutics has been hampered, however, by the difficulty in preparing the materials in sufficient quantity and quality by traditional methods. In recent years, a variety of recombinant methods has been developed for efficient production of BsAb, both as antibody fragments and as full-length IgG-like molecules. Here we describe a novel recombinant approach for the production of an Fc domain-containing, IgG-like tetravalent BsAb, with two antigen-binding sites to each of its target antigens, by genetically fusing a single variable domain antibody to the N terminus of the light chain of a functional IgG antibody of different specificity. A model BsAb was constructed using a single variable domain antibody to mouse platelet-derived growth factor receptor alpha and a conventional IgG antibody to mouse vascular endothelial growth factor receptor 2. The BsAb was expressed in mammalian cells and purified to homogeneity by one-step protein A affinity chromatography. Furthermore, the BsAb retains the antigen binding specificity and the receptor neutralizing activity of both of its parent antibodies. This design and expression of Fc domain-containing, IgG-like BsAb should be applicable to the construction of similar BsAb from antibodies recognizing any pair of antigens.


Subject(s)
Antibodies, Bispecific/chemistry , Immunoglobulin G/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Antibody Specificity , Binding Sites , Chromatography , Chromatography, Affinity , Cross-Linking Reagents/pharmacology , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Escherichia coli/metabolism , Immunoglobulin Fragments , Immunologic Techniques , Inhibitory Concentration 50 , Kinetics , Mice , Molecular Sequence Data , Neoplasms/immunology , Neoplasms/metabolism , Peptide Library , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Protein Binding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Mol Cancer Ther ; 4(11): 1801-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16276002

ABSTRACT

Etk, the 70-kDa member of the Tec family of nonreceptor protein tyrosine kinases, is expressed in a variety of hematopoietic, epithelial, and endothelial cells and was shown to be involved in several cellular processes, including proliferation, differentiation, and motility. In this study, we describe a novel approach using a human single-domain antibody phage display library for the generation of intrabodies directed against Etk. These single-domain antibodies bind specifically to recombinant Etk and efficiently block its kinase activity. When expressed in transformed cells, these antibodies associated tightly with Etk, leading to significant blockade of Etk enzymatic activity and inhibition of clonogenic cell growth in soft agar. Our results indicate that Etk may play a role in Src-induced cellular transformation and thus may represent a good target for cancer intervention. Furthermore, our single-domain antibody-based intrabody system proves to be an excellent tool for future intracellular targeting of other signaling molecules.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/metabolism , Protein-Tyrosine Kinases/physiology , Agar/chemistry , Animals , Blotting, Western , Cell Differentiation , Cell Proliferation , Cloning, Molecular , DNA/chemistry , Dose-Response Relationship, Drug , Enzyme Activation , Glutathione Transferase/metabolism , Humans , Immunoprecipitation , Mice , NIH 3T3 Cells , Peptide Library , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , RNA Interference , Recombinant Proteins/chemistry , Signal Transduction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...