Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Invest Ophthalmol Vis Sci ; 60(13): 4097-4108, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31574535

RESUMO

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.


Assuntos
Angiopoietina-1/antagonistas & inibidores , Angiopoietina-2/antagonistas & inibidores , Humor Aquoso/metabolismo , Glaucoma/fisiopatologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Angiopoietina-1/fisiologia , Angiopoietina-2/fisiologia , Animais , Anticorpos/farmacologia , Pressão Intraocular , Primatas , Fator A de Crescimento do Endotélio Vascular/fisiologia
2.
J Biol Chem ; 290(36): 21773-86, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26088137

RESUMO

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.


Assuntos
Indutores da Angiogênese/imunologia , Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Degeneração Macular/imunologia , Indutores da Angiogênese/química , Indutores da Angiogênese/metabolismo , Angiopoietina-2/antagonistas & inibidores , Angiopoietina-2/imunologia , Angiopoietina-2/metabolismo , Anticorpos Monoclonais/química , Anticorpos Monoclonais/uso terapêutico , Regiões Determinantes de Complementaridade/genética , Regiões Determinantes de Complementaridade/imunologia , Desenho de Fármacos , Estudos de Viabilidade , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Cinética , Degeneração Macular/tratamento farmacológico , Degeneração Macular/metabolismo , Modelos Moleculares , Terapia de Alvo Molecular/métodos , Mutação , Ligação Proteica/imunologia , Engenharia de Proteínas/métodos , Estrutura Terciária de Proteína , Reprodutibilidade dos Testes , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
PLoS One ; 8(7): e68755, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874750

RESUMO

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.


Assuntos
Linfonodos/patologia , Metástase Linfática/patologia , Fator C de Crescimento do Endotélio Vascular/metabolismo , Animais , Anticorpos/uso terapêutico , Feminino , Linfa/fisiologia , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Metástase Linfática/fisiopatologia , Metástase Linfática/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/antagonistas & inibidores
4.
Cancer Cell ; 20(4): 472-86, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22014573

RESUMO

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.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Antineoplásicos/uso terapêutico , Receptores ErbB/antagonistas & inibidores , Imunoglobulina G/uso terapêutico , Receptor ErbB-3/antagonistas & inibidores , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/toxicidade , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Especificidade de Anticorpos , Antineoplásicos/química , Antineoplásicos/toxicidade , Sítios de Ligação de Anticorpos , Ligação Competitiva , Cetuximab , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/química , Receptores ErbB/imunologia , Feminino , Humanos , Imunoglobulina G/efeitos adversos , Imunoglobulina G/química , Sistema de Sinalização das MAP Quinases , Macaca fascicularis , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/imunologia , Transdução de Sinais
5.
Nat Biotechnol ; 28(6): 585-93, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20495549

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Engenharia Genética , Mutação/genética , Neoplasias/genética , Neoplasias/terapia , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Cloridrato de Erlotinib , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Quinazolinas/uso terapêutico , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/imunologia , Gencitabina
6.
Science ; 323(5921): 1610-4, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19299620

RESUMO

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.


Assuntos
Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/imunologia , Receptor ErbB-2/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia , Animais , Anticorpos Biespecíficos/química , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Afinidade de Anticorpos , Especificidade de Anticorpos , Sítios de Ligação de Anticorpos/genética , Proliferação de Células/efeitos dos fármacos , Regiões Determinantes de Complementaridade/genética , Regiões Determinantes de Complementaridade/imunologia , Cristalografia por Raios X , Epitopos/imunologia , Epitopos/metabolismo , Engenharia Genética , Humanos , Camundongos , Modelos Moleculares , Mutagênese , Neoplasias Experimentais/tratamento farmacológico , Conformação Proteica , Estrutura Terciária de Proteína , Receptor ErbB-2/química , Receptor ErbB-2/metabolismo , Termodinâmica , Trastuzumab , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Invest Ophthalmol Vis Sci ; 49(2): 522-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18234994

RESUMO

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.


Assuntos
Inibidores da Angiogênese/metabolismo , Anticorpos Monoclonais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Bevacizumab , Western Blotting , Bovinos , Proliferação de Células/efeitos dos fármacos , Neovascularização de Coroide/patologia , Interações Medicamentosas , Eletroforese em Gel de Poliacrilamida , Endotélio Vascular/efeitos dos fármacos , Feminino , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Neoplasias Cutâneas/patologia , Ressonância de Plasmônio de Superfície , Fator A de Crescimento do Endotélio Vascular/farmacologia
8.
Proc Natl Acad Sci U S A ; 104(9): 3478-83, 2007 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-17360669

RESUMO

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.


Assuntos
Anticorpos Monoclonais/metabolismo , Afinidade de Anticorpos/genética , Carcinoma/metabolismo , Proliferação de Células/efeitos dos fármacos , Neovascularização Patológica/metabolismo , Proteínas Recombinantes/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutagênese , Especificidade da Espécie , Fator A de Crescimento do Endotélio Vascular/genética
9.
Blood ; 108(9): 3103-11, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16840730

RESUMO

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.


Assuntos
Fator Ativador de Células B/imunologia , Receptor do Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Anticorpos/uso terapêutico , Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/química , Sítios de Ligação , Cristalografia por Raios X , Ensaio de Imunoadsorção Enzimática , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Ativação Linfocitária , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese , Conformação Proteica , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
10.
J Biol Chem ; 281(10): 6625-31, 2006 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-16373345

RESUMO

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.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/fisiologia , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/fisiologia , Fator A de Crescimento do Endotélio Vascular/imunologia , Animais , Anticorpos Monoclonais Humanizados , Bevacizumab , Sítios de Ligação de Anticorpos , Reações Cruzadas , Epitopos , Humanos , Camundongos , Ligação Proteica/imunologia , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
11.
J Biol Chem ; 281(2): 951-61, 2006 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-16278208

RESUMO

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.


Assuntos
Fator A de Crescimento do Endotélio Vascular/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados , Bevacizumab , Linhagem Celular Tumoral , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina G/química , Cinética , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Transplante de Neoplasias , Neovascularização Patológica , Biblioteca de Peptídeos , Ligação Proteica , Especificidade da Espécie , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular/química , Fator A de Crescimento do Endotélio Vascular/imunologia
12.
J Immunol Methods ; 284(1-2): 119-32, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14736422

RESUMO

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.


Assuntos
Anticorpos Biespecíficos/química , Bacteriófago M13/química , Fragmentos Fab das Imunoglobulinas/química , Sequência de Aminoácidos , Anticorpos Biespecíficos/genética , Bacteriófago M13/genética , Sequência de Bases , Sítios de Ligação de Anticorpos , Western Blotting , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática , Fragmentos Fab das Imunoglobulinas/genética , Imunoglobulina G/química , Imunoglobulina G/genética , Zíper de Leucina , Mimetismo Molecular , Dados de Sequência Molecular , Biblioteca de Peptídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA