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










Publication year range
1.
Proc Natl Acad Sci U S A ; 117(18): 9952-9963, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32345717

ABSTRACT

Genetic polymorphisms in the region of the trimeric serine hydrolase high-temperature requirement 1 (HTRA1) are associated with increased risk of age-related macular degeneration (AMD) and disease progression, but the precise biological function of HtrA1 in the eye and its contribution to disease etiologies remain undefined. In this study, we have developed an HtrA1-blocking Fab fragment to test the therapeutic hypothesis that HtrA1 protease activity is involved in the progression of AMD. Next, we generated an activity-based small-molecule probe (ABP) to track target engagement in vivo. In addition, we used N-terminomic proteomic profiling in preclinical models to elucidate the in vivo repertoire of HtrA1-specific substrates, and identified substrates that can serve as robust pharmacodynamic biomarkers of HtrA1 activity. One of these HtrA1 substrates, Dickkopf-related protein 3 (DKK3), was successfully used as a biomarker to demonstrate the inhibition of HtrA1 activity in patients with AMD who were treated with the HtrA1-blocking Fab fragment. This pharmacodynamic biomarker provides important information on HtrA1 activity and pharmacological inhibition within the ocular compartment.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Antibodies, Anti-Idiotypic/pharmacology , Geographic Atrophy/drug therapy , High-Temperature Requirement A Serine Peptidase 1/genetics , Macular Degeneration/drug therapy , Adaptor Proteins, Signal Transducing/isolation & purification , Aged , Animals , Antibodies, Anti-Idiotypic/genetics , Antibodies, Anti-Idiotypic/immunology , Biomarkers/blood , Disease Progression , Female , Genetic Predisposition to Disease , Genotype , Geographic Atrophy/blood , Geographic Atrophy/genetics , Geographic Atrophy/immunology , High-Temperature Requirement A Serine Peptidase 1/antagonists & inhibitors , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/pharmacology , Macular Degeneration/blood , Macular Degeneration/genetics , Macular Degeneration/immunology , Male , Polymorphism, Single Nucleotide/genetics , Proteome/genetics , Proteome/immunology , Rats , Retina/drug effects , Retina/immunology , Retina/pathology , Small Molecule Libraries/pharmacology
2.
Invest Ophthalmol Vis Sci ; 60(13): 4097-4108, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31574535

ABSTRACT

Purpose: Investigate a significant, dose-related increase in IOP, leading to glaucomatous damage to the neuroretina and optic nerve following intravitreal (ITV) administration of a bispecific F(ab')2 [anti-VEGF/Angiopoietins [ANGPT]F(ab')2] molecule in adult monkeys. Methods: ITV ocular tolerability and investigation of anti-VEGF/ANGPT F(ab')2 (blocking both ANGPT1 and ANGPT2) was done in monkeys; mechanistic studies were done in neonatal mice. Results: Following the second ITV dose of anti-VEGF/ANGPT F(ab')2, all 1.5- and 4-mg/eye treated monkeys developed elevated IOP, which eventually was associated with optic disc cupping and thinning of the neuroretinal rim. Histopathologic examination showed nonreversible axonal degeneration in the optic nerves of animals administered 1.5 mg/eye and higher that was considered secondary to high IOP. Anti-ANGPT Fab also caused elevated IOP in monkeys, but anti-VEGF Fab did not contribute to the IOP increase. In addition, an anti-ANGPT2-selective antibody did not change IOP. In mice simultaneous blockade of ANGPT1 and ANGPT2 impaired the expansion and formation of Schlemm's canal (SC) vessels, similar to genetic ablation of Angpt1/Angpt2 and their receptor TIE2. As previously reported, blocking ANGPT2 alone did not affect SC formation in mice. Conclusions: Dual inhibition of ANGPT1/ANGPT2, but not ANGPT2 alone, leads to increased IOP and glaucomatous damage in monkeys. This confirms a role for TIE2/ANGPT signaling in the control of IOP in adults, a finding initially identified in transgenic mice. Dual pharmacologic inhibition of ANGPT1/ANGPT2 may affect aqueous drainage and homeostasis in adult monkeys and may be useful in developing novel models of glaucoma.


Subject(s)
Angiopoietin-1/antagonists & inhibitors , Angiopoietin-2/antagonists & inhibitors , Aqueous Humor/metabolism , Glaucoma/physiopathology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiopoietin-1/physiology , Angiopoietin-2/physiology , Animals , Antibodies/pharmacology , Intraocular Pressure , Primates , Vascular Endothelial Growth Factor A/physiology
3.
MAbs ; 9(6): 959-967, 2017.
Article in English | MEDLINE | ID: mdl-28585908

ABSTRACT

Monoclonal antibodies developed for therapeutic or diagnostic purposes need to demonstrate highly defined binding specificity profiles. Engineering of an antibody to enhance or reduce binding to related antigens is often needed to achieve the desired biologic activity without safety concern. Here, we describe a deep sequencing-aided engineering strategy to fine-tune the specificity of an angiopoietin-2 (Ang2)/vascular endothelial growth factor (VEGF) dual action Fab, 5A12.1 for the treatment of age-related macular degeneration. This antibody utilizes overlapping complementarity-determining region (CDR) sites for dual Ang2/VEGF interaction with KD in the sub-nanomolar range. However, it also exhibits significant (KD of 4 nM) binding to angiopoietin-1, which has high sequence identity with Ang2. We generated a large phage-displayed library of 5A12.1 Fab variants with all possible single mutations in the 6 CDRs. By tracking the change of prevalence of each mutation during various selection conditions, we identified 35 mutations predicted to decrease the affinity for Ang1 while maintaining the affinity for Ang2 and VEGF. We confirmed the specificity profiles for 25 of these single mutations as Fab protein. Structural analysis showed that some of the Fab mutations cluster near a potential Ang1/2 epitope residue that differs in the 2 proteins, while others are up to 15 Å away from the antigen-binding site and likely influence the binding interaction remotely. The approach presented here provides a robust and efficient method for specificity engineering that does not require prior knowledge of the antigen antibody interaction and can be broadly applied to antibody specificity engineering projects.

4.
Proc Natl Acad Sci U S A ; 114(4): E486-E495, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28057863

ABSTRACT

Somatic mutations within the antibody variable domains are critical to the immense capacity of the immune repertoire. Here, via a deep mutational scan, we dissect how mutations at all positions of the variable domains of a high-affinity anti-VEGF antibody G6.31 impact its antigen-binding function. The resulting mutational landscape demonstrates that large portions of antibody variable domain positions are open to mutation, and that beneficial mutations can be found throughout the variable domains. We determine the role of one antigen-distal light chain position 83, demonstrating that mutation at this site optimizes both antigen affinity and thermostability by modulating the interdomain conformational dynamics of the antigen-binding fragment. Furthermore, by analyzing a large number of human antibody sequences and structures, we demonstrate that somatic mutations occur frequently at position 83, with corresponding domain conformations observed for G6.31. Therefore, the modulation of interdomain dynamics represents an important mechanism during antibody maturation in vivo.


Subject(s)
Antibodies/genetics , Immunoglobulin Fab Fragments/genetics , Antibodies/chemistry , Antibodies/immunology , Antibody Affinity , Antigens/immunology , Binding Sites, Antibody , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Mutation , Protein Conformation
5.
Invest Ophthalmol Vis Sci ; 56(9): 5390-400, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26275136

ABSTRACT

PURPOSE: To design and select the next generation of ocular therapeutics, we performed a comprehensive ocular and systemic pharmacokinetic (PK) analysis of a variety of antibodies and antibody fragments, including a novel-designed bispecific antibody. METHODS: Molecules were administrated via intravitreal (IVT) or intravenous (IV) injections in rabbits, and antibody concentrations in each tissue were determined by ELISA. A novel mathematical model was developed to quantitate the structure-PK relationship. RESULTS: After IVT injection, differences in vitreal half-life observed across all molecules ranged between 3.2 and 5.2 days. Modification or elimination of the fragment crystallizable (Fc) region reduced serum half-life from 9 days for the IgG to 5 days for the neonatal Fc receptor (FcRn) null mAb, to 3.1 to 3.4 days for the other formats. The F(ab')2 was the optimal format for ocular therapeutics with comparable vitreal half-life to full-length antibodies, but with minimized systemic exposure. Concomitantly, the consistency among mathematical model predictions and observed data validated the model for future PK predictions. In addition, we showed a novel design to develop bispecific antibodies, here with activity targeting multiple angiogenesis pathways. CONCLUSIONS: We demonstrated that protein molecular weight and Fc region do not play a critical role in ocular PK, as they do systemically. Moreover, the mathematical model supports the selection of the "ideal therapeutic" by predicting ocular and systemic PK of any antibody format for any dose regimen. These findings have important implications for the design and selection of ocular therapeutics according to treatment needs, such as maximizing ocular half-life and minimizing systemic exposure.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies/immunology , Drug Design , Eye Diseases/drug therapy , Eye/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibody Affinity , Eye Diseases/immunology , Eye Diseases/metabolism , Intravitreal Injections , Male , Protein Binding , Rabbits
6.
J Biol Chem ; 290(36): 21773-86, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26088137

ABSTRACT

The development of dual targeting antibodies promises therapies with improved efficacy over mono-specific antibodies. Here, we engineered a Two-in-One VEGF/angiopoietin 2 antibody with dual action Fab (DAF) as a potential therapeutic for neovascular age-related macular degeneration. Crystal structures of the VEGF/angiopoietin 2 DAF in complex with its two antigens showed highly overlapping binding sites. To achieve sufficient affinity of the DAF to block both angiogenic factors, we turned to deep mutational scanning in the complementarity determining regions (CDRs). By mutating all three CDRs of each antibody chain simultaneously, we were able not only to identify affinity improving single mutations but also mutation pairs from different CDRs that synergistically improve both binding functions. Furthermore, insights into the cooperativity between mutations allowed us to identify fold-stabilizing mutations in the CDRs. The data obtained from deep mutational scanning reveal that the majority of the 52 CDR residues are utilized differently for the two antigen binding function and permit, for the first time, the engineering of several DAF variants with sub-nanomolar affinity against two structurally unrelated antigens. The improved variants show similar blocking activity of receptor binding as the high affinity mono-specific antibodies against these two proteins, demonstrating the feasibility of generating a dual specificity binding surface with comparable properties to individual high affinity mono-specific antibodies.


Subject(s)
Angiogenesis Inducing Agents/immunology , Antibodies, Monoclonal/immunology , Antibody Affinity/immunology , Antibody Specificity/immunology , Macular Degeneration/immunology , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/metabolism , Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/immunology , Angiopoietin-2/metabolism , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Drug Design , Feasibility Studies , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin Fab Fragments/immunology , Kinetics , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Models, Molecular , Molecular Targeted Therapy/methods , Mutation , Protein Binding/immunology , Protein Engineering/methods , Protein Structure, Tertiary , Reproducibility of Results , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
7.
MAbs ; 6(3): 622-7, 2014.
Article in English | MEDLINE | ID: mdl-24618680

ABSTRACT

A mono-specific antibody may recruit a second antigen binding specificity, thus converting to a dual-specific Two-in-One antibody through mutation at the light chain complementarity-determining regions (CDRs). It is, however, unknown whether mutation at the heavy chain CDRs may evolve such dual specificity. Herein, we examined the CDRs of a humanized interleukin 4 (IL4) antibody using alanine scanning and structural modeling, designed libraries of mutants in regions that tolerate mutation, and isolated dual specific antibodies harboring mutation at the heavy chain CDRs only. We then affinity improved an IL4/IL5 dual specific antibody to variants with dissociation constants in the low nanomolar range for both antigens. The results demonstrate the full capacity of antibodies to evolve dual binding specificity.


Subject(s)
Antibodies, Bispecific/genetics , Antibodies, Monoclonal, Humanized/genetics , Interleukin-4/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Monoclonal, Humanized/chemistry , Binding Sites, Antibody/genetics , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Mice , Models, Molecular , Mutagenesis , Peptide Library , Protein Engineering
8.
PLoS One ; 8(7): e68755, 2013.
Article in English | MEDLINE | ID: mdl-23874750

ABSTRACT

Tumor-associated lymphatics are postulated to provide a transit route for disseminating metastatic cells. This notion is supported by preclinical findings that inhibition of pro-lymphangiogenic signaling during tumor development reduces cell spread to sentinel lymph nodes (SLNs). However, it is unclear how lymphatics downstream of SLNs contribute to metastatic spread into distal organs, or if modulating distal lymph transport impacts disease progression. Utilizing murine models of metastasis, longitudinal in vivo imaging of lymph transport, and function blocking antibodies against two VEGF family members, we provide evidence that distal lymphatics undergo disease course-dependent up-regulation of lymph transport coincidental with structural remodeling. Inhibition of VEGF-C activity with antibodies against VEGF-C or NRP2 prevented these disease-associated changes. Furthermore, utilizing a novel model of adjuvant treatment, we demonstrate that antagonism of VEGF-C or NRP2 decreases post SLN metastasis. These data support a potential therapeutic strategy for inhibiting distant metastatic dissemination via targeting tumor-associated lymphatic remodeling.


Subject(s)
Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Vascular Endothelial Growth Factor C/metabolism , Animals , Antibodies/therapeutic use , Female , Lymph/physiology , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymphatic Metastasis/physiopathology , Lymphatic Metastasis/prevention & control , Male , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects , Vascular Endothelial Growth Factor C/antagonists & inhibitors
9.
PLoS One ; 7(4): e35844, 2012.
Article in English | MEDLINE | ID: mdl-22563408

ABSTRACT

Attempts to express eukaryotic multi-spanning membrane proteins at high-levels have been generally unsuccessful. In order to investigate the cause of this limitation and gain insight into the rate limiting processes involved, we have analyzed the effect of translation levels on the expression of several human membrane proteins in Escherichia coli (E. coli). These results demonstrate that excessive translation initiation rates of membrane proteins cause a block in protein synthesis and ultimately prevent the high-level accumulation of these proteins. Moderate translation rates allow coupling of peptide synthesis and membrane targeting, resulting in a significant increase in protein expression and accumulation over time. The current study evaluates four membrane proteins, CD20 (4-transmembrane (TM) helixes), the G-protein coupled receptors (GPCRs, 7-TMs) RA1c and EG-VEGFR1, and Patched 1 (12-TMs), and demonstrates the critical role of translation initiation rates in the targeting, insertion and folding of integral membrane proteins in the E. coli membrane.


Subject(s)
Membrane Proteins/biosynthesis , Antigens, CD20/genetics , Antigens, CD20/metabolism , Escherichia coli/metabolism , Humans , Membrane Proteins/genetics , Peptide Chain Initiation, Translational , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
10.
Cancer Cell ; 20(4): 472-86, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-22014573

ABSTRACT

Extensive crosstalk among ErbB/HER receptors suggests that blocking signaling from more than one family member may be essential to effectively treat cancer and limit drug resistance. We generated a conventional IgG molecule MEHD7945A with dual HER3/EGFR specificity by phage display engineering and used structural and mutational studies to understand how a single antigen recognition surface binds two epitopes with high affinity. As a human IgG1, MEHD7945A exhibited dual action by inhibiting EGFR- and HER3-mediated signaling in vitro and in vivo and the ability to engage immune effector functions. Compared with monospecific anti-HER antibodies, MEHD7945A was more broadly efficacious in multiple tumor models, showing that combined inhibition of EGFR and HER3 with a single antibody is beneficial.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antineoplastic Agents/therapeutic use , ErbB Receptors/antagonists & inhibitors , Immunoglobulin G/therapeutic use , Receptor, ErbB-3/antagonists & inhibitors , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/toxicity , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody Specificity , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Binding Sites, Antibody , Binding, Competitive , Cetuximab , Crystallography, X-Ray , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , ErbB Receptors/chemistry , ErbB Receptors/immunology , Female , Humans , Immunoglobulin G/adverse effects , Immunoglobulin G/chemistry , MAP Kinase Signaling System , Macaca fascicularis , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/immunology , Signal Transduction
11.
Nat Biotechnol ; 28(6): 585-93, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20495549

ABSTRACT

The low rate of approval of novel anti-cancer agents underscores the need for better preclinical models of therapeutic response as neither xenografts nor early-generation genetically engineered mouse models (GEMMs) reliably predict human clinical outcomes. Whereas recent, sporadic GEMMs emulate many aspects of their human disease counterpart more closely, their ability to predict clinical therapeutic responses has never been tested systematically. We evaluated the utility of two state-of-the-art, mutant Kras-driven GEMMs--one of non-small-cell lung carcinoma and another of pancreatic adenocarcinoma--by assessing responses to existing standard-of-care chemotherapeutics, and subsequently in combination with EGFR and VEGF inhibitors. Standard clinical endpoints were modeled to evaluate efficacy, including overall survival and progression-free survival using noninvasive imaging modalities. Comparisons with corresponding clinical trials indicate that these GEMMs model human responses well, and lay the foundation for the use of validated GEMMs in predicting outcome and interrogating mechanisms of therapeutic response and resistance.


Subject(s)
Disease Models, Animal , Genetic Engineering , Mutation/genetics , Neoplasms/genetics , Neoplasms/therapy , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Erlotinib Hydrochloride , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Quinazolines/therapeutic use , Survival Analysis , Vascular Endothelial Growth Factor A/immunology , Gemcitabine
12.
Science ; 323(5921): 1610-4, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19299620

ABSTRACT

The interface between antibody and antigen is often depicted as a lock and key, suggesting that an antibody surface can accommodate only one antigen. Here, we describe an antibody with an antigen binding site that binds two distinct proteins with high affinity. We isolated a variant of Herceptin, a therapeutic monoclonal antibody that binds the human epidermal growth factor receptor 2 (HER2), on the basis of its ability to simultaneously interact with vascular endothelial growth factor (VEGF). Crystallographic and mutagenesis studies revealed that distinct amino acids of this antibody, called bH1, engage HER2 and VEGF energetically, but there is extensive overlap between the antibody surface areas contacting the two antigens. An affinity-improved version of bH1 inhibits both HER2- and VEGF-mediated cell proliferation in vitro and tumor progression in mouse models. Such "two-in-one" antibodies challenge the monoclonal antibody paradigm of one binding site, one antigen. They could also provide new opportunities for antibody-based therapy.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Receptor, ErbB-2/immunology , Vascular Endothelial Growth Factor A/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody Affinity , Antibody Specificity , Binding Sites, Antibody/genetics , Cell Proliferation/drug effects , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Crystallography, X-Ray , Epitopes/immunology , Epitopes/metabolism , Genetic Engineering , Humans , Mice , Models, Molecular , Mutagenesis , Neoplasms, Experimental/drug therapy , Protein Conformation , Protein Structure, Tertiary , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Thermodynamics , Trastuzumab , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
13.
Methods Mol Biol ; 525: 353-76, xiii, 2009.
Article in English | MEDLINE | ID: mdl-19252851

ABSTRACT

Affinity maturation is an important part of the therapeutic antibody development process as in vivo activity often requires high binding affinity. Here, we describe a targeted approach for affinity improvement of therapeutic antibodies. Sets of CDR residues that are solvent accessible and relatively diverse in natural antibodies are targeted for diversification. Degenerate oligonucleotides are used to generate combinatorial phage-displayed antibody libraries with varying degree of diversity at randomized positions from which high-affinity antibodies can be selected. An advantage of using antibodies for therapy is their exquisite target specificity, which enables selective antigen binding and reduces off-target effects. However, it can be useful, and often it is necessary, to generate cross-reactive antibodies binding to not only the human antigen but also the corresponding non-human primate or rodent orthologs. Such cross-reactive antibodies can be used to validate the therapeutic targeting and examine the safety profile in preclinical animal models before committing to a costly development track. We show how affinity improvement and cross-species binding can be achieved in a one-step process.


Subject(s)
Antibodies/therapeutic use , Antibody Affinity/immunology , Antibody Specificity/immunology , Molecular Biology/methods , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/genetics , Base Sequence , Clone Cells , Codon/genetics , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin G/biosynthesis , Inhibitory Concentration 50 , Mice , Molecular Sequence Data , Oligonucleotides/genetics , Peptide Library
14.
Invest Ophthalmol Vis Sci ; 49(2): 522-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18234994

ABSTRACT

PURPOSE: Bevacizumab is a humanized anti-human VEGF-A monoclonal antibody (mAb) approved by the United States Food and Drug Administration for cancer therapy and used off label to treat neovascular age-related macular degeneration. Earlier studies characterized bevacizumab as species specific and lacking the ability to neutralize murine (m) VEGF-A. However, a recent study reported that bevacizumab is a potent inhibitor of hemangiogenesis and lymphangiogenesis in murine models. The authors sought to reassess the interaction between bevacizumab and mVEGF-A. METHODS: The authors performed Western blot analysis, plasmon resonance by BIAcore, and endothelial cell proliferation assays to characterize the interaction between bevacizumab and mVEGF-A. They also tested whether bevacizumab had any effects in two in vivo murine models, laser-induced choroidal neovascularization (CNV) and melanoma growth. RESULTS: Western blot detected a very weak interaction, but BIAcore detected no measurable interaction between mVEGF and bevacizumab. Bevacizumab failed to inhibit mVEGF-stimulated endothelial cell proliferation. In addition, bevacizumab was indistinguishable from the control antibody in the CNV and tumor models, whereas a cross-reactive anti-VEGF-A mAb had dramatic inhibitory effects. CONCLUSIONS: Bevacizumab has an extremely weak interaction with mVEGF-A, which fails to result in immunoneutralization as assessed by several bioassays.


Subject(s)
Angiogenesis Inhibitors/metabolism , Antibodies, Monoclonal/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Blotting, Western , Cattle , Cell Proliferation/drug effects , Choroidal Neovascularization/pathology , Drug Interactions , Electrophoresis, Polyacrylamide Gel , Endothelium, Vascular/drug effects , Female , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Skin Neoplasms/pathology , Surface Plasmon Resonance , Vascular Endothelial Growth Factor A/pharmacology
15.
Proc Natl Acad Sci U S A ; 104(9): 3478-83, 2007 Feb 27.
Article in English | MEDLINE | ID: mdl-17360669

ABSTRACT

VEGF-A is important in tumor angiogenesis, and a humanized anti-VEGF-A monoclonal antibody (bevacizumab) has been approved by the FDA as a treatment for metastatic colorectal and nonsquamous, non-small-cell lung cancer in combination with chemotherapy. However, contributions of both tumor- and stromal-cell derived VEGF-A to vascularization of human tumors grown in immunodeficient mice hindered direct comparison between the pharmacological effects of anti-VEGF antibodies with different abilities to block host VEGF. Therefore, by gene replacement technology, we engineered mice to express a humanized form of VEGF-A (hum-X VEGF) that is recognized by many anti-VEGF antibodies and has biochemical and biological properties comparable with WT mouse and human VEGF-A. The hum-X VEGF mouse model was then used to compare the activity and safety of a panel of VEGF Mabs with different affinities for VEGF-A. Although in vitro studies clearly showed a correlation between binding affinity and potency at blocking endothelial cell proliferation stimulated by VEGF, in vivo experiments failed to document any consistent correlation between antibody affinity and the ability to inhibit tumor growth and angiogenesis in most animal models. However, higher-affinity antibodies were more likely to result in glomerulosclerosis during long-term treatment.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibody Affinity/genetics , Carcinoma/metabolism , Cell Proliferation/drug effects , Neovascularization, Pathologic/metabolism , Recombinant Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Humans , Kidney/drug effects , Kidney/pathology , Mice , Mice, Transgenic , Molecular Sequence Data , Mutagenesis , Species Specificity , Vascular Endothelial Growth Factor A/genetics
16.
Blood ; 108(9): 3103-11, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16840730

ABSTRACT

BR3, which is expressed on all mature B cells, is a specific receptor for the B-cell survival and maturation factor BAFF (B-cell-activating factor belonging to the tumor necrosis factor [TNF] family). In order to investigate the consequences of targeting BR3 in murine models and to assess the potential of BR3 antibodies as human therapeutics, synthetic antibody phage libraries were employed to identify BAFF-blocking antibodies cross-reactive to murine and human BR3, which share 52% identity in their extracellular domains. We found an antibody, CB1, which exhibits muM affinity for murine BR3 and very weak affinity for the human receptor. CB3s, an affinity-matured variant of CB1, has sub-nM affinity for BR3 from both species. Alanine scanning and crystallographic structural analysis of the CB3s/BR3 complex reveal that CB3s mimics BAFF by interacting with a similar region of the BR3 surface. Despite this similarity in binding epitopes, CB1 variants antagonize BAFF-dependent human B-cell proliferation in vitro and are effective at reducing murine B-cell populations in vivo, showing significant promise as therapeutics for human B-cell-mediated diseases.


Subject(s)
B-Cell Activating Factor/immunology , B-Cell Activation Factor Receptor/genetics , B-Cell Activation Factor Receptor/immunology , B-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antibodies/immunology , Antibodies/therapeutic use , B-Cell Activating Factor/genetics , B-Cell Activation Factor Receptor/chemistry , Binding Sites , Crystallography, X-Ray , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/immunology , Lymphocyte Activation , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid
17.
J Biol Chem ; 281(10): 6625-31, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16373345

ABSTRACT

In the quest to discover new research tools and to develop better agents in the fight against cancer, two antibodies, G6 and B20-4, were isolated from synthetic antibody phage libraries. Unlike the AVASTINtrade mark antibody, a recently approved agent for the treatment of patients with colorectal cancer, B20-4 and G6 bind and block both human and murine vascular endothelial growth factor (VEGF). Here we have analyzed and compared the binding epitopes on VEGF for these three antibodies using alanine-scanning mutagenesis and structural analyses. The epitopes recognized by both synthetic antibodies are conserved between human and mouse VEGF, and they match closely to the receptor epitopes both structurally and functionally. In contrast, the Avastin epitope overlaps minimally with the receptor binding surface and centers around a residue that is not conserved in mouse. Our structural and functional analyses elucidate the cross-species reactivity of all three antibodies and emphasize the potential advantages of antibody generation using phage display as the resulting antibodies do not depend on sequence differences across species and preferentially target natural protein-protein interaction surfaces.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/physiology , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/physiology , Vascular Endothelial Growth Factor A/immunology , Animals , Antibodies, Monoclonal, Humanized , Bevacizumab , Binding Sites, Antibody , Cross Reactions , Epitopes , Humans , Mice , Protein Binding/immunology , Protein Structure, Tertiary , Structure-Activity Relationship
18.
J Biol Chem ; 281(2): 951-61, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16278208

ABSTRACT

To fully assess the role of VEGF-A in tumor angiogenesis, antibodies that can block all sources of vascular endothelial growth factor (VEGF) are desired. Selectively targeting tumor-derived VEGF overlooks the contribution of host stromal VEGF. Other strategies, such as targeting VEGF receptors directly or using receptor decoys, result in inhibiting not only VEGF-A but also VEGF homologues (e.g. placental growth factor, VEGF-B, and VEGF-C), which may play a role in angiogenesis. Here we report the identification of novel anti-VEGF antibodies, B20 and G6, from synthetic antibody phage libraries, which block both human and murine VEGF action in vitro. Their affinity-improved variants completely inhibit three human tumor xenografts in mice of skeletal muscle, colorectal, and pancreatic origins (A673, HM-7, and HPAC). Avastin, which only inhibits the tumor-derived human VEGF, is approximately 90% effective at inhibiting HM-7 and A673 growth but is <50% effective at inhibiting HPAC growth. Indeed, HPAC tumors contain more host stroma invasion and stroma-derived VEGF than other tumors. Thus, the functional contribution of stromal VEGF varies greatly among tumors, and systemic blockade of both tumor and stroma-derived VEGF is sufficient for inhibiting the growth of tumor xenografts.


Subject(s)
Vascular Endothelial Growth Factor A/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Bevacizumab , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/chemistry , Kinetics , Mice , Mice, Nude , Molecular Sequence Data , Muscle, Skeletal/metabolism , Neoplasm Transplantation , Neovascularization, Pathologic , Peptide Library , Protein Binding , Species Specificity , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/immunology
19.
J Mol Biol ; 340(5): 1073-93, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15236968

ABSTRACT

Phage-displayed synthetic antibody libraries were built on a single human framework by introducing synthetic diversity at solvent-exposed positions within the heavy chain complementarity-determining regions (CDRs). The design strategy of mimicking natural diversity using tailored codons had been validated previously with scFv libraries, which produced antibodies that bound to antigen, murine vascular endothelial growth factor (mVEGF), with affinities in the 100nM range. To improve library performance, we constructed monovalent and bivalent antigen-binding fragment (Fab) libraries, and explored different CDR-H3 diversities by varying the amino acid composition and CDR length. A Fab with sub-nanomolar affinity for mVEGF was obtained from a library with CDR-H3 diversity designed to contain all 20 naturally occurring amino acids. We then expanded the library by increasing the variability of CDR-H3 length and using tailored codons that mimicked the amino acid composition of natural CDR-H3 sequences. The library was tested against a panel of 13 protein antigens and high-affinity Fabs were obtained for most antigens. Furthermore, the heavy chain of an anti-mVEGF clone was recombined with a library of light chain CDRs, and the affinity was improved from low nanomolar to low picomolar. The results demonstrated that high-affinity human antibodies can be generated from libraries with completely synthetic CDRs displayed on a single scaffold.


Subject(s)
Antibodies/chemistry , Antibodies/immunology , Antibody Affinity , Bacteriophages/genetics , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Peptide Library , Antibodies/genetics , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Kinetics , Models, Molecular , Protein Engineering , Protein Structure, Tertiary
20.
J Immunol Methods ; 284(1-2): 119-32, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14736422

ABSTRACT

We report the development of a system for displaying bivalent antibody fragments on M13 bacteriophage in a manner that effectively mimics the binding behavior of natural antibodies. In the "bivalent display" format, two copies of antigen binding sites are displayed on the coat of a single phage particle. Bivalent display was first achieved by the insertion of a dimerization domain, consisting of an IgG1 hinge region and a homodimerizing GCN4 leucine zipper, between a Fab and the C-terminal domain of the M13 gene-3 minor coat protein. In a phagemid-based display system, the resulting "Fab'-zip-phage" particles display bivalent Fabs that resemble natural IgGs. An important functional consequence of bivalent display is an avidity effect, which results in a greatly reduced off-rate for phage bound to immobilized antigen. The avidity effect improved the capture and retention of bivalent Fab'-zip-phage relative to monovalent Fab-phage both with antigen immobilized on plates and with cell surface antigen. To examine the requirements for bivalent display on phage, we systematically trimmed down the dimerization domain and found that a single cysteine was sufficient to confer the same avidity effect conferred by the complete dimerization domain. Bivalent antibody phage display should be useful for many applications. In particular, the technology should aid in the production of antibodies against difficult antigens, and also, in selections that require dimerization for activity.


Subject(s)
Antibodies, Bispecific/chemistry , Bacteriophage M13/chemistry , Immunoglobulin Fab Fragments/chemistry , Amino Acid Sequence , Antibodies, Bispecific/genetics , Bacteriophage M13/genetics , Base Sequence , Binding Sites, Antibody , Blotting, Western , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Immunoglobulin Fab Fragments/genetics , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Leucine Zippers , Molecular Mimicry , Molecular Sequence Data , Peptide Library
SELECTION OF CITATIONS
SEARCH DETAIL
...