Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 23(11): 1722-1733, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26234505

RESUMO

FS102 is a HER2-specific Fcab (Fc fragment with antigen binding), which binds HER2 with high affinity and recognizes an epitope that does not overlap with those of trastuzumab or pertuzumab. In tumor cells that express high levels of HER2, FS102 caused profound HER2 internalization and degradation leading to tumor cell apoptosis. The antitumor effect of FS102 in patient-derived xenografts (PDXs) correlated strongly with the HER2 amplification status of the tumors. Superior activity of FS102 over trastuzumab or the combination of trastuzumab and pertuzumab was observed in vitro and in vivo when the gene copy number of HER2 was equal to or exceeded 10 per cell based on quantitative polymerase chain reaction (qPCR). Thus, FS102 induced complete and sustained tumor regression in a significant proportion of HER2-high PDX tumor models. We hypothesize that the unique structure and/or epitope of FS102 enables the Fcab to internalize and degrade cell surface HER2 more efficiently than standard of care antibodies. In turn, increased depletion of HER2 commits the cells to apoptosis as a result of oncogene shock. FS102 has the potential of a biomarker-driven therapeutic that derives superior antitumor effects from a unique mechanism-of-action in tumor cells which are oncogenically addicted to the HER2 pathway due to overexpression.


Assuntos
Apoptose/efeitos dos fármacos , Fragmentos Fc das Imunoglobulinas/farmacologia , Neoplasias/tratamento farmacológico , Receptor ErbB-2/antagonistas & inibidores , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proliferação de Células , Humanos , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptor ErbB-2/imunologia , Transdução de Sinais , Trastuzumab/farmacologia , Trastuzumab/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Immunol ; 187(8): 4229-35, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21930965

RESUMO

The immune system is known to generate a diverse panel of high-affinity Abs by adaptively improving the recognition of pathogens during ongoing immune responses. In this study, we report the biological limits for Ag-driven affinity maturation and repertoire diversification by analyzing Ab repertoires in two adult volunteers after each of three consecutive booster vaccinations with tetanus toxoid. Maturation of on-rates and off-rates occurred independently, indicating a kinetically controlled affinity maturation process. The third vaccination induced no significant changes in the distribution of somatic mutations and binding rate constants implying that the limits for affinity maturation and repertoire diversification had been reached. These fully matured Ab repertoires remained similar in size, genetically diverse, and dynamic. Somatic mutations and kinetic rate constants showed normal and log-normal distribution profiles, respectively. Mean values can therefore be considered as biological constants defining the observed boundaries. At physiological temperature, affinity maturation peaked at k(on) = 1.6 × 10(4) M(-1) s(-1) and k(off) = 1.7 × 10(-4) s(-1) leading to a maximum mean affinity of K(D) = 1.0 × 10(-9) M. At ambient temperature, the average affinity increased to K(D) = 3.4 × 10(-10) M mainly due to slower off-rates. This experimentally determined set of constants can be used as a benchmark for analysis of the maturation level of human Abs and Ab responses.


Assuntos
Afinidade de Anticorpos/imunologia , Imunização Secundária , Hipermutação Somática de Imunoglobulina/imunologia , Toxoide Tetânico/imunologia , Vacinação/métodos , Sequência de Aminoácidos , Afinidade de Anticorpos/genética , Humanos , Região Variável de Imunoglobulina/genética , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Hipermutação Somática de Imunoglobulina/genética , Ressonância de Plasmônio de Superfície
3.
J Virol ; 85(4): 1820-33, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21147924

RESUMO

The eradication of smallpox (variola) and the subsequent cessation of routine vaccination have left modern society vulnerable to bioterrorism employing this devastating contagious disease. The existing, licensed vaccines based on live vaccinia virus (VACV) are contraindicated for a substantial number of people, and prophylactic vaccination of large populations is not reasonable when there is little risk of exposure. Consequently, there is an emerging need to develop efficient and safe therapeutics to be used shortly before or after exposure, either alone or in combination with vaccination. We have characterized the human antibody response to smallpox vaccine (VACV Lister) in immunized volunteers and isolated a large number of VACV-specific antibodies that recognize a variety of different VACV antigens. Using this broad antibody panel, we have generated a fully human, recombinant analogue to plasma-derived vaccinia immunoglobulin (VIG), which mirrors the diversity and specificity of the human antibody immune response and offers the advantage of unlimited supply and reproducible specificity and activity. The recombinant VIG was found to display a high specific binding activity toward VACV antigens, potent in vitro VACV neutralizing activity, and a highly protective efficacy against VACV challenge in the mouse tail lesion model when given either prophylactically or therapeutically. Altogether, the results suggest that this compound has the potential to be used as an effective postexposure prophylaxis or treatment of disease caused by orthopoxviruses.


Assuntos
Anticorpos Antivirais/biossíntese , Antígenos Virais/imunologia , Vacina Antivariólica/imunologia , Varíola/prevenção & controle , Vacinas Sintéticas/imunologia , Vaccinia virus/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/química , Anticorpos Antivirais/genética , Formação de Anticorpos , Especificidade de Anticorpos , Antígenos Virais/química , Antígenos Virais/genética , Doadores de Sangue , Feminino , Variação Genética , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/química , Imunoglobulina G/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Análise de Sequência de DNA , Varíola/tratamento farmacológico , Varíola/imunologia , Vacina Antivariólica/administração & dosagem , Vacina Antivariólica/genética , Vacinação , Vacinas Sintéticas/genética , Vacinas Sintéticas/uso terapêutico , Vaccinia virus/classificação , Vaccinia virus/genética
4.
Mol Immunol ; 44(4): 412-22, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16581131

RESUMO

Anti-Rhesus D immunoglobulin purified from human sera is used as a prophylactic reagent in Rhesus D negative women at risk of alloimmunization during pregnancy. We are currently developing a Rhesus D antigen-specific recombinant polyclonal antibody drug lead for replacing the existing blood derived-products. By analyzing the RhD-specific antibody VH repertoires from eight alloimmunized women we found, in agreement with previous studies, a strong preference for the VH 3-33 "superspecies" gene segments which encompasses the IGHV3-30-3*01, IGHV3-30*18, and IGHV3-33*01 VH alleles. Even more extensive genetic restriction was observed among five donors, which produced antibodies of identical V-D-J usage and CDR3 loop length and joining regions of similar amino acid composition. In addition, we find a high degree of sequence relatedness to previously isolated anti-Rhesus D antibodies. Such close homology in VH domains indicates that significant structural restrictions are operating in the selection of antibodies recognizing RhD as seen for T cell receptors. Moreover, some VH domains were isolated in their germline configuration indicating that anti-RhD antibodies of relatively high affinity are present in the naïve antibody repertoire of Rhesus negative individuals which offers an explanation for the strong and clinically significant immunogenicity of the Rhesus D.


Assuntos
Anticorpos/imunologia , Cadeias Pesadas de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/imunologia , Sistema do Grupo Sanguíneo Rh-Hr/imunologia , Sequência de Aminoácidos , Anticorpos/sangue , Anticorpos/genética , Afinidade de Anticorpos , Especificidade de Anticorpos , Sequência de Bases , Feminino , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Dados de Sequência Molecular , Gravidez , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Alinhamento de Sequência
5.
J Mol Biol ; 358(3): 764-72, 2006 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-16563430

RESUMO

The humoral immune system in higher vertebrates is unique in its ability to generate highly diverse antibody responses against most pathogens as well as against certain malignancies. Several technologies have been developed to exploit this vast source of potentially therapeutic antibodies, including hybridoma technology, phage display and yeast display. Here, we present a novel, high-throughput technology (the Symplex Technology) for rapid direct cloning and identification of human antigen-specific high-affinity antibodies from single antibody-producing cells of immune individuals. The utility of the technology was demonstrated by isolation of diverse sets of unique high-affinity antibodies against tetanus toxoid and influenza virus from immunized volunteers. Hence, the Symplex Technology is a new method for the rapid isolation of high-affinity antibodies directly from humans.


Assuntos
Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/isolamento & purificação , Cadeias Leves de Imunoglobulina/imunologia , Cadeias Leves de Imunoglobulina/isolamento & purificação , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/metabolismo , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/metabolismo , Cinética , Dados de Sequência Molecular , Biblioteca de Peptídeos , Filogenia , Toxina Tetânica/imunologia
6.
Drug Discov Today ; 11(13-14): 655-60, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16793535

RESUMO

Antibodies have been used as therapeutics in various forms for over a century. Traditional immunoglobulin therapy has the advantage of reflecting the diversity of the natural immune response but has very limited clinical applications. However, over the past ten years more than 30 monoclonal antibodies have been successfully introduced on to the drug market. The monoclonal approach provides the advantage of specificity, but lacks efficacy in the treatment of diseases caused by complex antigens. Recombinant polyclonal antibodies, the third generation of antibody therapeutics, have the ability to tackle complex and highly mutagenic targets, and will undoubtedly offer a promising commercial future.


Assuntos
Anticorpos/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Imunoterapia/tendências , Linfoma/tratamento farmacológico , Animais , Anticorpos/genética , Anticorpos/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/imunologia , Neoplasias da Mama/imunologia , Epitopos/imunologia , Humanos , Linfoma/imunologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Tecnologia Farmacêutica/tendências
7.
Curr Pharm Des ; 12(16): 2007-15, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16787244

RESUMO

The mammalian immune system eliminates pathogens by generating a specific antibody response. Polyclonality is a key feature of this immune response: the immune system produces antibodies which bind to different structures on a given pathogen thereby increasing the likelihood of its elimination. The vast majority of current recombinant antibody drugs rely on monospecific monoclonal antibodies. Inherently, such antibodies do not represent the benefits of polyclonality utilized by a natural immune system and this has impeded the identification of efficacious antibody drugs against infectious agents, including viruses. The development of novel technologies has allowed the identification and manufacturing of antigen-specific recombinant polyclonal human antibodies, so-called symphobodies. This review describes the rationale for designing drugs based on symphobodies against pathogenic viruses, including HIV, vaccinia and smallpox virus, and respiratory syncytial virus.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunização Passiva/métodos , Proteínas Recombinantes/uso terapêutico , Viroses/terapia , Anticorpos Monoclonais/genética , Vírus do Sarcoma Aviário/imunologia , Guerra Biológica/prevenção & controle , HIV/imunologia , Humanos , Orthopoxvirus/imunologia
8.
MAbs ; 8(1): 1-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26716992

RESUMO

An important step in drug development is the assignment of an International Nonproprietary Name (INN) by the World Health Organization (WHO) that provides healthcare professionals with a unique and universally available designated name to identify each pharmaceutical substance. Monoclonal antibody INNs comprise a -mab suffix preceded by a substem indicating the antibody type, e.g., chimeric (-xi-), humanized (-zu-), or human (-u-). The WHO publishes INN definitions that specify how new monoclonal antibody therapeutics are categorized and adapts the definitions to new technologies. However, rapid progress in antibody technologies has blurred the boundaries between existing antibody categories and created a burgeoning array of new antibody formats. Thus, revising the INN system for antibodies is akin to aiming for a rapidly moving target. The WHO recently revised INN definitions for antibodies now to be based on amino acid sequence identity. These new definitions, however, are critically flawed as they are ambiguous and go against decades of scientific literature. A key concern is the imposition of an arbitrary threshold for identity against human germline antibody variable region sequences. This leads to inconsistent classification of somatically mutated human antibodies, humanized antibodies as well as antibodies derived from semi-synthetic/synthetic libraries and transgenic animals. Such sequence-based classification implies clear functional distinction between categories (e.g., immunogenicity). However, there is no scientific evidence to support this. Dialog between the WHO INN Expert Group and key stakeholders is needed to develop a new INN system for antibodies and to avoid confusion and miscommunication between researchers and clinicians prescribing antibodies.


Assuntos
Anticorpos , Animais , Humanos , Terminologia como Assunto
9.
IDrugs ; 8(5): 404-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15883923

RESUMO

Current antibody therapeutics can be grouped into two generations, each distinguished by a unique feature of the immune system: diversity and specificity. Antibodies from human blood (immunoglobulin) represent the first generation, and are characterized by the natural diversity of human antibody responses. The second generation consists of recombinant monoclonal antibodies (mAbs), which are characterized by high specificity toward a single, often well-described antigen. The natural immune response comprises a plurality of specificities, many of which do not compete for binding, whereas molecules in a mAb all compete for binding to the same epitope. Thus, the epitope is more likely to become a limiting factor for mAb binding to complex targets compared with a polyclonal antibody. Also, epitope-escape by mutation or natural variation is less likely to be a problem for polyclonal antibodies. Technologies attempting to develop truly human recombinant antigen-specific polyclonal antibodies, such as the Sympress technology, are closing a natural circle between the first generations of antibody technologies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Tecnologia Farmacêutica/métodos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Indústria Farmacêutica/economia , Indústria Farmacêutica/tendências , Epitopos/genética , Epitopos/imunologia , Humanos , Tecnologia Farmacêutica/tendências
10.
Expert Opin Biol Ther ; 4(3): 387-96, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15006732

RESUMO

The potential use of biological agents such as viruses, bacteria or bacterial toxins as weapons of mass destruction has fuelled significant national and international research and development in novel prophylactic or therapeutic countermeasures. Such measures need to be fast-acting and broadly specific, a hallmark of target-specific polyclonal antibodies (pAbs). As reviewed here, pathogen-specific antibodies in the form of human or animal serum have long been recognised as effective therapies in a number of infectious diseases. This review focuses in particular on the potential biowarfare agents prioritised by the National Institute of Allergy and Infectious Diseases and the Centers for Disease Control and Prevention (CDC), referred to as the category A organisms. Furthermore, it is propose that the last decade of development in recombinant antibody technologies offers the possibility for developing highly specific human monoclonal or polyclonal pathogen-specific antibodies. In particular, pathogen-specific polyclonal human antibodies offer certain advantages over existing hyperimmune serum products, monoclonal antibodies, small molecule drugs and vaccines. Here, the rationale for designing pAb-based therapeutics against the CDC category A microbial agents causing anthrax, botulism, plague, smallpox, tularaemia and viral haemorrhagic fevers, as well as the overall design of such therapeutics, are discussed.


Assuntos
Anticorpos/uso terapêutico , Infecções Bacterianas/terapia , Animais , Anticorpos Monoclonais/uso terapêutico , Infecções Bacterianas/microbiologia , Humanos , Imunização Passiva , Proteínas Recombinantes/uso terapêutico
11.
MAbs ; 3(6): 584-95, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22123060

RESUMO

The epidermal growth factor receptor (EGFR) is frequently dysregulated in human malignancies and a validated target for cancer therapy. Two monoclonal anti-EGFR antibodies (cetuximab and panitumumab) are approved for clinical use. However, the percentage of patients responding to treatment is low and many patients experiencing an initial response eventually relapse. Thus, the need for more efficacious treatments remains. Previous studies have reported that mixtures of antibodies targeting multiple distinct epitopes are more effective than single mAbs at inhibiting growth of human cancer cells in vitro and in vivo. The current work describes the rational approach that led to discovery and selection of a novel anti-EGFR antibody mixture Sym004, which is currently in Phase 2 clinical testing. Twenty-four selected anti-EGFR antibodies were systematically tested in dual and triple mixtures for their ability to inhibit cancer cells in vitro and tumor growth in vivo. The results show that targeting EGFR dependent cancer cells with mixtures of antibodies is superior at inhibiting their growth both in vitro and in vivo. In particular, antibody mixtures targeting non-overlapping epitopes on domain III are efficient and indeed Sym004 is composed of two monoclonal antibodies targeting this domain. The superior growth inhibitory activity of mixtures correlated with their ability to induce efficient EGFR degradation.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/uso terapêutico , Receptores ErbB/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antineoplásicos/química , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Mapeamento de Epitopos , Epitopos/química , Epitopos/imunologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Humanos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Ressonância de Plasmônio de Superfície , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Res ; 70(2): 588-97, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20068188

RESUMO

Epidermal growth factor receptor (EGFR) is a validated therapeutic target in cancer and EGFR antagonists with greater effectiveness than existing clinical agents remain of interest. Here, we report a novel approach based on Sym004, a mixture of two anti-EGFR monoclonal antibodies directed against distinct nonoverlapping epitopes in EGFR extracellular domain III. Like anti-EGFR monoclonal antibodies in current clinical use, Sym004 inhibits cancer cell growth and survival by blocking ligand-binding receptor activation and phosphorylation and downstream receptor signaling. However, unlike the other antibodies, Sym004 induces rapid and efficient removal of the receptor from the cancer cell surface by triggering EGFR internalization and degradation. Compared with reference anti-EGFR monoclonal antibodies, Sym004 exhibited more pronounced growth inhibition in vitro and superior efficacy in vivo. Together, these findings illustrate a strategy to target EGFR more effectively than existing clinical antibodies.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptores ErbB/antagonistas & inibidores , Neoplasias/terapia , Animais , Anticorpos Monoclonais/imunologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo , Sinergismo Farmacológico , Epitopos/imunologia , Receptores ErbB/imunologia , Receptores ErbB/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/enzimologia , Fosforilação , Estrutura Terciária de Proteína , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Immunol ; 177(6): 3782-90, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16951339

RESUMO

The immunogenicity of therapeutic Abs is a concern as anti-drug Abs may impact negatively on the pharmacodynamics and safety profile of Ab drugs. The factors governing induction of anti-drug Abs are not fully understood. In this study, we describe a model based on mouse-human chimeric Abs for the study of Ab immunogenicity in vivo. Six chimeric Abs containing human V regions and mouse C regions were generated from six human anti-Rhesus D Abs and the Ag-binding characteristics of the parental human Abs were retained. Analysis of the immune response toward the individual chimeric Abs revealed the induction of anti-variable domain Abs including anti-idiotypic Abs against some of these, thereby demonstrating the applicability of the model for studying anti-drug Ab responses in vivo. Immunization of BALB/c, C57, and outbred NMRI mice with a polyclonal composition consisting of all six chimeric Abs demonstrated that the immunogenicity of the individual Abs was haplotype dependent. Chimeric Abs, which were nonimmunogenic when administered individually, did not become immunogenic as part of the polyclonal composition, implying the absence of epitope spreading. Ex vivo Ab-binding studies established a clear correlation between the level of immunogenicity of the Abs comprised in the composition and the impact on the pharmacology of the Abs. These analyses demonstrate that under these conditions this polyclonal Ab composition was generally less susceptible to blocking Abs than the respective mAbs.


Assuntos
Anticorpos Anti-Idiotípicos/metabolismo , Região Variável de Imunoglobulina/imunologia , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Anti-Idiotípicos/administração & dosagem , Anticorpos Anti-Idiotípicos/biossíntese , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/metabolismo , Sítios de Ligação de Anticorpos/genética , Feminino , Humanos , Soros Imunes/metabolismo , Imunoglobulina G/metabolismo , Região Variável de Imunoglobulina/administração & dosagem , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Sistema do Grupo Sanguíneo Rh-Hr/imunologia
15.
Int Arch Allergy Immunol ; 140(3): 261-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16699287

RESUMO

BACKGROUND: Recombinant allergen-specific immunoglobulin G (IgG) antibody therapy can reduce allergic asthma symptoms by inhibiting the immunoglobulin E (IgE)-mediated allergic response. This study investigated the effect of intranasally administered allergen-specific monoclonal (mAb) and polyclonal (pAb) antibody on airway inflammation and hyperresponsiveness (AHR) in a mouse model of human asthma. METHODS: Ovalbumin (OVA)-specific IgG2b antibodies were generated by phage display using spleens from OVA-immunized mice, and screening against OVA and finally expressed in CHO cells. Sensitized mice were treated intranasally with either a recombinant anti-OVA mAb (gc32) or a polyclonal preparation comprising seven selected antibodies (including gc32). Control mice received diluent only, OVA only, a control polymeric IgG or dexamethasone. Following challenge with nebulized OVA, investigators assessed airway inflammation by histology and cellular composition of the bronchoalveolar fluid, and methacholine-induced airway hyperresponsiveness (AHR). Serum levels of total and OVA-specific IgE were measured by ELISA. RESULTS: Sensitized mice developed airway inflammation and AHR in response to OVA challenge. Intranasally administered OVA-specific murine polyclonal or monoclonal IgG2b antibodies both reduced OVA-induced lung inflammation. Polyclonal, but not anti-OVA mAb, also reduced AHR and eosinophil influx into the airway lumen. Both anti-OVA antibody preparations reduced levels of specific IgE with no effect on total IgE levels. CONCLUSIONS: Intranasal treatment with allergen-specific pAb reduces pulmonary inflammation and AHR in a mouse model of allergic asthma, but allergen-specific mAb reduces inflammation only. Allergen-specific recombinant pAb offers a potentially valuable therapeutic approach to the management of allergic asthma.


Assuntos
Anticorpos/uso terapêutico , Asma/imunologia , Asma/terapia , Imunoterapia/métodos , Administração Intranasal , Animais , Anticorpos/administração & dosagem , Anticorpos/imunologia , Especificidade de Anticorpos , Líquido da Lavagem Broncoalveolar/citologia , Contagem de Células , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Histocitoquímica , Hipersensibilidade Imediata/imunologia , Hipersensibilidade Imediata/prevenção & controle , Imunoglobulina E/sangue , Imunoterapia/normas , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico
16.
Biotechnol Bioeng ; 94(2): 396-405, 2006 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-16596663

RESUMO

We describe the expression and consistent production of a first target-specific recombinant human polyclonal antibody. An anti-Rhesus D recombinant polyclonal antibody, Sym001, comprised of 25 unique human IgG1 antibodies, was produced by the novel Sympress expression technology. This strategy is based on site-specific integration of antibody genes in CHO cells, using the FRT/Flp-In recombinase system. This allows integration of the expression construct at the same genomic site in the host cells, thereby reducing genomic position effects. Different bioreactor batches of Sym001 displayed highly consistent manufacturing yield, antibody composition, binding potency, and functional activity. The results demonstrate that diverse recombinant human polyclonal antibody compositions can be reproducibly generated under conditions directly applicable to industrial manufacturing settings and present a first recombinant polyclonal antibody which could be used for treatment of hemolytic disease of the newborn and/or idiopathic thrombocytopenic purpura.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Marcação de Genes , Engenharia Genética , Animais , Anticorpos Monoclonais/genética , Reatores Biológicos , Células CHO , Cricetinae , DNA Nucleotidiltransferases , Eritrócitos/imunologia , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA