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.
MAbs ; 3(3): 273-88, 2011.
Article in English | MEDLINE | ID: mdl-21393993

ABSTRACT

The epidermal growth factor receptor (EGFR) and the type I insulin-like growth factor receptor (IGF-1R) are two cell surface receptor tyrosine kinases known to cooperate to promote tumor progression and drug resistance. Combined blockade of EGFR and IGF-1R has shown improved anti-tumor activity in preclinical models. Here, we report the characterization of a stable IgG-like bispecific antibody (BsAb) dual-targeting EGFR and IGF-1R that was developed for cancer therapy. The BsAb molecule (EI-04), constructed with a stability-engineered single chain variable fragment (scFv) against IGF-1R attached to the carboxyl-terminus of an IgG against EGFR, displays favorable biophysical properties for biopharmaceutical development. Biochemically, EI-04 bound to human EGFR and IGF-1R with sub nanomolar affinity, co-engaged the two receptors simultaneously, and blocked the binding of their respective ligands with similar potency compared to the parental monoclonal antibodies (mAbs). In tumor cells, EI-04 effectively inhibited EGFR and IGF-1R phosphorylation, and concurrently blocked downstream AKT and ERK activation, resulting in greater inhibition of tumor cell growth and cell cycle progression than the single mAbs. EI-04, likely due to its tetravalent bispecific format, exhibited high avidity binding to BxPC3 tumor cells co-expressing EGFR and IGF-1R, and consequently improved potency at inhibiting IGF-driven cell growth over the mAb combination. Importantly, EI-04 demonstrated enhanced in vivo anti-tumor efficacy over the parental mAbs in two xenograft models, and even over the mAb combination in the BxPC3 model. Our data support the clinical investigation of EI-04 as a superior cancer therapeutic in treating EGFR and IGF-1R pathway responsive tumors.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , ErbB Receptors/immunology , Neoplasms/immunology , Receptor, IGF Type 1/immunology , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , Antibody Affinity/immunology , Antibody Specificity/immunology , Blotting, Western , CHO Cells , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Humans , Immunoglobulin G/immunology , Mice , Mice, Nude , Mice, SCID , Neoplasms/drug therapy , Neoplasms/pathology , Phosphorylation/drug effects , Receptor, IGF Type 1/metabolism , Signal Transduction/drug effects , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 9(9): 2593-604, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20716637

ABSTRACT

The insulin-like growth factor-I receptor (IGF-IR) is a cell surface receptor tyrosine kinase that mediates cell survival signaling and supports tumor progression in multiple tumor types. We identified a spectrum of inhibitory IGF-IR antibodies with diverse binding epitopes and ligand-blocking properties. By binding distinct inhibitory epitopes, two of these antibodies, BIIB4 and BIIB5, block both IGF-I and IGF-II binding to IGF-IR using competitive and allosteric mechanisms, respectively. Here, we explored the inhibitory effects of combining BIIB4 and BIIB5. In biochemical assays, the combination of BIIB4 and BIIB5 improved both the potency and extent of IGF-I and IGF-II blockade compared with either antibody alone. In tumor cells, the combination of BIIB4 and BIIB5 accelerated IGF-IR downregulation and more efficiently inhibited IGF-IR activation as well as downstream signaling, particularly AKT phosphorylation. In several carcinoma cell lines, the antibody combination more effectively inhibited ligand-driven cell growth than either BIIB4 or BIIB5 alone. Notably, the enhanced tumor growth-inhibitory activity of the BIIB4 and BIIB5 combination was much more pronounced at high ligand concentrations, where the individual antibodies exhibited substantially reduced activity. Compared with single antibodies, the BIIB4 and BIIB5 combination also significantly further enhanced the antitumor activity of the epidermal growth factor receptor inhibitor erlotinib and the mTOR inhibitor rapamycin. Moreover, in osteosarcoma and hepatocellular carcinoma xenograft models, the BIIB4 and BIIB5 combination significantly reduced tumor growth to a greater degree than each single antibody. Taken together, our results suggest that targeting multiple distinct inhibitory epitopes on IGF-IR may be a more effective strategy of affecting the IGF-IR pathway in cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Bone Neoplasms/drug therapy , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Disease Progression , Down-Regulation/drug effects , Epitopes/immunology , Female , Hep G2 Cells , Humans , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/antagonists & inhibitors , Insulin-Like Growth Factor II/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Osteosarcoma/drug therapy , Osteosarcoma/immunology , Osteosarcoma/pathology , Receptor, IGF Type 1/immunology , Receptor, IGF Type 1/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
3.
J Biol Chem ; 284(15): 10254-67, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19211557

ABSTRACT

Therapeutic antibodies directed against the type 1 insulin-like growth factor receptor (IGF-1R) have recently gained significant momentum in the clinic because of preliminary data generated in human patients with cancer. These antibodies inhibit ligand-mediated activation of IGF-1R and the resulting down-stream signaling cascade. Here we generated a panel of antibodies against IGF-1R and screened them for their ability to block the binding of both IGF-1 and IGF-2 at escalating ligand concentrations (>1 microm) to investigate allosteric versus competitive blocking mechanisms. Four distinct inhibitory classes were found as follows: 1) allosteric IGF-1 blockers, 2) allosteric IGF-2 blockers, 3) allosteric IGF-1 and IGF-2 blockers, and 4) competitive IGF-1 and IGF-2 blockers. The epitopes of representative antibodies from each of these classes were mapped using a purified IGF-1R library containing 64 mutations. Most of these antibodies bound overlapping surfaces on the cysteine-rich repeat and L2 domains. One class of allosteric IGF-1 and IGF-2 blocker was identified that bound a separate epitope on the outer surface of the FnIII-1 domain. Using various biophysical techniques, we show that the dual IGF blockers inhibit ligand binding using a spectrum of mechanisms ranging from highly allosteric to purely competitive. Binding of IGF-1 or the inhibitory antibodies was associated with conformational changes in IGF-1R, linked to the ordering of dynamic or unstructured regions of the receptor. These results suggest IGF-1R uses disorder/order within its polypeptide sequence to regulate its activity. Interestingly, the activity of representative allosteric and competitive inhibitors on H322M tumor cell growth in vitro was reflective of their individual ligand-blocking properties. Many of the antibodies in the clinic likely adopt one of the inhibitory mechanisms described here, and the outcome of future clinical studies may reveal whether a particular inhibitory mechanism leads to optimal clinical efficacy.


Subject(s)
Epitopes/chemistry , Receptors, Somatomedin/chemistry , Allosteric Site , Animals , CHO Cells , Calorimetry, Differential Scanning , Cricetinae , Cricetulus , Epitope Mapping , Humans , Insulin-Like Growth Factor II/chemistry , Kinetics , Ligands , Molecular Conformation , Receptor, IGF Type 1/metabolism
4.
Proteins ; 76(1): 99-114, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19089973

ABSTRACT

Engineered antibodies are a large and growing class of protein therapeutics comprising both marketed products and many molecules in clinical trials in various disease indications. We investigated naturally conserved networks of amino acids that support antibody V(H) and V(L) function, with the goal of generating information to assist in the engineering of robust antibody or antibody-like therapeutics. We generated a large and diverse sequence alignment of V-class Ig-folds, of which V(H) and V(L) domains are family members. To identify conserved amino acid networks, covariations between residues at all possible position pairs were quantified as correlation coefficients (phi-values). We provide rosters of the key conserved amino acid pairs in antibody V(H) and V(L) domains, for reference and use by the antibody research community. The majority of the most strongly conserved amino acid pairs in V(H) and V(L) are at or adjacent to the V(H)-V(L) interface suggesting that the ability to heterodimerize is a constraining feature of antibody evolution. For the V(H) domain, but not the V(L) domain, residue pairs at the variable-constant domain interface (V(H)-C(H)1 interface) are also strongly conserved. The same network of conserved V(H) positions involved in interactions with both the V(L) and C(H)1 domains is found in camelid V(HH) domains, which have evolved to lack interactions with V(L) and C(H)1 domains in their mature structures; however, the amino acids at these positions are different, reflecting their different function. Overall, the data describe naturally occurring amino acid networks in antibody Fv regions that can be referenced when designing antibodies or antibody-like fragments with the goal of improving their biophysical properties.


Subject(s)
Antibodies/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/immunology , Protein Engineering/methods , Amino Acid Sequence , Conserved Sequence , Databases, Protein , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Immunoglobulin Variable Region/genetics , Molecular Sequence Data , Protein Folding , Sequence Alignment
5.
J Biol Chem ; 280(50): 41494-503, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16221669

ABSTRACT

HuCC49 deltaCH2 is a heavy chain constant domain 2 domain-deleted antibody under development as a radioimmunotherapeutic for treating carcinomas overexpressing the TAG-72 tumor antigen. Mammalian cell culture biosynthesis of HuCC49 deltaCH2 produces two isoforms (form A and form B) in an approximate 1:1 ratio, and consequently separation and purification of the desired form A isoform adversely impact process and yield. A protein engineering strategy was used to develop a panel of hinge-engineered HuCC49 deltaCH2 antibodies to identify hinge sequences to optimize production of the form A isoform. We found that adding a single proline residue at Kabat position 243, immediately adjacent to the carboxyl end of the core middle hinge CPPC domain, resulted in an increase from 39 to 51% form A isoform relative to the parent HuCC49 deltaCH2 antibody. Insertion of the amino acids proline-alanine-proline (PAP) at positions 243-245 enhanced production of the form A isoform to 72%. Insertion of a cysteine-rich 15-amino acid IgG3 hinge motif (CPEPKSCDTPPPCPR) in both of these mutant antibodies resulted in secretion of predominantly form A isoform with little or no detectable form B. Yields exceeding 98% of the form A isoform have been realized. Preliminary peptide mapping and mass spectrometry analysis suggest that at least two, and as many as five, inter-heavy chain disulfide linkages may be present.


Subject(s)
Protein Engineering/methods , Radioimmunotherapy/methods , Amino Acid Motifs , Animals , Antibodies/chemistry , Antibodies, Monoclonal/chemistry , Antibodies, Neoplasm/chemistry , Binding, Competitive , Blotting, Western , CHO Cells , Cattle , Cricetinae , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , Genetic Vectors , Hypoxia , Immunohistochemistry , Mass Spectrometry , Models, Biological , Models, Molecular , Mucins/chemistry , Mutation , Oligonucleotides/chemistry , Peptide Mapping , Peptides/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation , Protein Isoforms , Protein Structure, Tertiary , Spectrometry, Fluorescence , Time Factors
6.
Int Immunopharmacol ; 2(4): 453-61, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11962725

ABSTRACT

A chimeric macaque/human (PRIMATIZED) anti-CD23 antibody, p6G5G1, demonstrated a strong inhibitory effect on IL-4 and anti-CD40 antibody-stimulated IgE production by human peripheral blood mononuclear cells (PBMCs). RNA analysis by both reverse transcription-polymerase chain reaction (RT-PCR) and Northern blot showed that p6G5G1 inhibited germline Cepsilon RNA synthesis, but had no effect on CD23 mRNA levels. These data suggest that p6G5G1 may inhibit immunoglobulin class switching to IgE through the inhibition of germline Cepsilon RNA synthesis. Early addition of p6G5G1 after stimulation by IL-4 and anti-CD40 was critical for IgE inhibition. In contrast, later addition of p6G5G1 still showed inhibition of increased levels of surface CD23, which is normally upregulated by stimulation with IL-4 and anti-CD40.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , B-Lymphocytes/drug effects , Immunoglobulin Constant Regions/genetics , Immunoglobulin E/genetics , Receptors, IgE/immunology , Transcription, Genetic/drug effects , Animals , B-Lymphocytes/metabolism , Flow Cytometry , Humans , Immunoglobulin Class Switching/drug effects , Immunoglobulin E/biosynthesis , Macaca , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, IgE/analysis , Receptors, IgE/biosynthesis , Receptors, IgE/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
7.
Cancer Control ; 9(2): 152-66, 2002.
Article in English | MEDLINE | ID: mdl-11965235

ABSTRACT

BACKGROUND: The approval of monoclonal antibodies (MAbs) as antibody-targeted therapy in the management of patients with hematologic malignancies has led to new treatment options for this group of patients. The ability to target antibodies to novel functional receptors can increase their therapeutic efficacy. METHODS: The authors reviewed improvements in MAb design to enhance their effectiveness over the existing therapeutic MAb currently approved for treating hematologic malignancies. RESULTS: Three classes of therapeutic MAbs showing promise in human clinical trials for treatment of hematologic malignancies include unconjugated MAb, drug conjugates in which the antibody preferentially delivers a potent cytotoxic drug to the tumor, and radioactive immunotherapy in which the antibody delivers a sterilizing dose of radiation to the tumor. CONCLUSIONS: A better appreciation of how MAbs are metabolized in the body and localized to tumors is resulting in the development of new antibody constructs with improved biodistribution profiles.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Hematologic Neoplasms/therapy , Animals , Clinical Trials, Phase III as Topic , Combined Modality Therapy , Drug Therapy , Forecasting , Hematologic Neoplasms/immunology , Host vs Graft Reaction/immunology , Humans , Immune System/drug effects , Immune System/immunology , Multicenter Studies as Topic , Radioimmunotherapy/standards , Treatment Outcome , United States/epidemiology , United States Food and Drug Administration
SELECTION OF CITATIONS
SEARCH DETAIL
...