Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Nat Commun ; 13(1): 923, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35177668

RESUMO

N-methyl-D-aspartate receptors (NMDARs) are critically involved in basic brain functions and neurodegeneration as well as tumor invasiveness. Targeting specific subtypes of NMDARs with distinct activities has been considered an effective therapeutic strategy for neurological disorders and diseases. However, complete elimination of off-target effects of small chemical compounds has been challenging and thus, there is a need to explore alternative strategies for targeting NMDAR subtypes. Here we report identification of a functional antibody that specifically targets the GluN1-GluN2B NMDAR subtype and allosterically down-regulates ion channel activity as assessed by electrophysiology. Through biochemical analysis, x-ray crystallography, single-particle electron cryomicroscopy, and molecular dynamics simulations, we show that this inhibitory antibody recognizes the amino terminal domain of the GluN2B subunit and increases the population of the non-active conformational state. The current study demonstrates that antibodies may serve as specific reagents to regulate NMDAR functions for basic research and therapeutic objectives.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/ultraestrutura , Clonagem Molecular , Microscopia Crioeletrônica , Cristalografia por Raios X , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Fragmentos Fab das Imunoglobulinas/farmacologia , Fragmentos Fab das Imunoglobulinas/ultraestrutura , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/isolamento & purificação , Região Variável de Imunoglobulina/farmacologia , Região Variável de Imunoglobulina/ultraestrutura , Simulação de Dinâmica Molecular , Oócitos , Ratos , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/isolamento & purificação , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/ultraestrutura , Células Sf9 , Spodoptera , Xenopus laevis
2.
Nat Chem ; 13(6): 540-548, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33833446

RESUMO

The encoding of chemical compounds with amplifiable DNA tags facilitates the discovery of small-molecule ligands for proteins. To investigate the impact of stereo- and regiochemistry on ligand discovery, we synthesized a DNA-encoded library of 670,752 derivatives based on 2-azido-3-iodophenylpropionic acids. The library was selected against multiple proteins and yielded specific ligands. The selection fingerprints obtained for a set of protein targets of pharmaceutical relevance clearly showed the preferential enrichment of ortho-, meta- or para-regioisomers, which was experimentally verified by affinity measurements in the absence of DNA. The discovered ligands included novel selective enzyme inhibitors and binders to tumour-associated antigens, which enabled conditional chimeric antigen receptor T-cell activation and tumour targeting.


Assuntos
Sistemas de Liberação de Medicamentos , Região Variável de Imunoglobulina/farmacologia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Animais , Linhagem Celular Tumoral , DNA/química , Descoberta de Drogas , Fluorescência , Biblioteca Gênica , Humanos , Região Variável de Imunoglobulina/química , Camundongos , Microscopia de Fluorescência , Neoplasias , Neoplasias Experimentais
3.
Methods Enzymol ; 629: 383-399, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31727250

RESUMO

The programmed death 1 (PD-1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) are negative regulators of T-cell immune function. Removal of these "brakes" in T cells results in increased activation of the immune system and controlling and eradicating tumor. The development of immune checkpoint inhibitors (ICIs) is a revolutionary milestone in tumor immunotherapy. Obtaining the atomic structure of the human immune checkpoint receptor/ICI therapeutic antibody complex is essential for understanding its inhibition mechanism and the rational design of improved biotherapeutics. In this chapter, we describe the methods for efficient production of extracellular domain of human immune checkpoint receptors and Fv fragments of ICI therapeutic antibodies in milligram quantities sufficient for structural studies, taking examples of the PD-1/pembrolizumab Fv and CTLA-4-ipilimumab Fv complexes.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Região Variável de Imunoglobulina/farmacologia , Neoplasias/tratamento farmacológico , Difração de Raios X/métodos , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/uso terapêutico , Medicamentos Biossimilares/química , Medicamentos Biossimilares/farmacologia , Medicamentos Biossimilares/uso terapêutico , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Antígeno CTLA-4/isolamento & purificação , Humanos , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/uso terapêutico , Ipilimumab/química , Ipilimumab/farmacologia , Ipilimumab/uso terapêutico , Modelos Moleculares , Neoplasias/imunologia , Neoplasias/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/isolamento & purificação , Domínios Proteicos/genética , Domínios Proteicos/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
5.
AAPS J ; 20(6): 103, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30255287

RESUMO

Among the numerous antibody-drug conjugate (ADC) clinical candidates, one of the most prevalent types utilizes the interchain cysteines in antibodies to conjugate auristatin via a maleimide-containing linker. In this class of ADCs, there are a paucity of systematic studies characterizing how IgG subclass influences the biophysical properties and in vivo pharmacokinetics of the ADC molecules. In the current investigation, we studied cysteine-conjugated ADCs using a model system consisting of human IgG1, IgG2, and IgG4 antibodies with the same variable region. Our findings identified some unforeseen differences among the three ADCs. Drug conjugation profiling by LC-MS revealed that 50% of inter heavy-light chain disulfide bonds are disrupted to conjugate drugs in IgG1 antibody while only 10% in IgG2 antibody and 20% in IgG4 antibody. The solution behavior of the ADCs was interrogated in concentrating experiments and diffusion interaction parameter measurements. We found that drug conjugation affected the solution property of the three antibodies differently, with the IgG2-based ADC having the most increased propensity to aggregate. Rat PK studies using a sensitive LC-MS-based bioanalytical method showed that the IgG1-based ADC has poor peripheral linker-payload stability while the IgG2- and IgG4-based ADCs are stable. The conjugate stability of the IgG2-based ADC was further confirmed in a cynomolgus monkey PK study. Overall, the IgG2-based ADC exhibited the best PK/conjugate stability but also the most deterioration in stability among the three ADCs. Our findings provide important information and present multifactorial considerations for the selection of IgG subclass during ADC drug discovery when employing stochastic cysteine conjugation.


Assuntos
Anticorpos Monoclonais/farmacocinética , Cisteína/química , Imunoconjugados/farmacocinética , Imunoglobulina G/farmacologia , Região Variável de Imunoglobulina/farmacologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Células CHO , Cricetulus , Estabilidade de Medicamentos , Imunoconjugados/administração & dosagem , Imunoconjugados/química , Imunoglobulina G/química , Região Variável de Imunoglobulina/química , Macaca fascicularis , Masculino , Ratos , Solubilidade
6.
Oncotarget ; 8(12): 20187-20197, 2017 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-28423625

RESUMO

The over-expression of basic fibroblast growth factor (bFGF) plays a crucial role in the development, invasion and metastasis of lung cancer. Therefore, neutralizing antibodies against bFGF may inhibit the growth of lung cancer. In this study, a Disulfide-stabilized diabody (ds-Diabody) against bFGF was constructed by site-directed mutation and overlap extension PCR (SOE-PCR) at the position of VH44 and VL100 in the scFv. The ds-Diabody was constructed and expressed in Pichia pastoris. We found that the ds-Diabody against bFGF could efficiently suppress the proliferation, migration and invasion of human lung cancer A549 cells in vitro. Moreover, in A549 cells, the ds-Diabody against bFGF could inhibit bFGF-induced activation of downstream signaling regulators, such as phospho-Akt and phospho-MAPK. In the nude mouse xenograft model of lung cancer, the ds-Diabody against bFGF could significantly inhibit tumor growth and decrease the densities of micro-vessels and lymphatic vessels in tumor tissue. Our data indicate that the ds-Diabody against bFGF could effectively suppress the lung cancer growth through blockade of bFGF signaling pathway and inhibition of tumor angiogenesis, which may make it a potential therapeutic candidate antibody drug for human lung cancer therapy.


Assuntos
Dissulfetos/química , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Região Variável de Imunoglobulina/farmacologia , Neoplasias Pulmonares/terapia , Neovascularização Patológica/prevenção & controle , Anticorpos de Cadeia Única/farmacologia , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Fator 2 de Crescimento de Fibroblastos/imunologia , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Toxins (Basel) ; 8(8)2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27463727

RESUMO

Recombinant immunotoxins (RITs) are fusions of an Fv-based targeting moiety and a toxin. Pseudomonas exotoxin A (PE) has been used to make several immunotoxins that have been evaluated in clinical trials. Immunogenicity of the bacterial toxin and off-target toxicity have limited the efficacy of these immunotoxins. To address these issues, we have previously made RITs in which the Fv is connected to domain III (PE24) by a furin cleavage site (FCS), thereby removing unneeded sequences of domain II. However, the PE24 containing RITs do not contain the naturally occurring disulfide bond around the furin cleavage sequence, because it was removed when domain II was deleted. This could potentially allow PE24 containing immunotoxins to be cleaved and inactivated before internalization by cell surface furin or other proteases in the blood stream or tumor microenvironment. Here, we describe five new RITs in which a disulfide bond is engineered to protect the FCS. The most active of these, SS1-Fab-DS3-PE24, shows a longer serum half-life than an RIT without the disulfide bond and has the same anti-tumor activity, despite being less cytotoxic in vitro. These results have significance for the production of de-immunized, low toxicity, PE24-based immunotoxins with a longer serum half-life.


Assuntos
ADP Ribose Transferases/farmacologia , Toxinas Bacterianas/farmacologia , Proliferação de Células/efeitos dos fármacos , Dissulfetos/farmacologia , Desenho de Fármacos , Exotoxinas/farmacologia , Furina/metabolismo , Região Variável de Imunoglobulina/farmacologia , Imunotoxinas/farmacologia , Neoplasias/tratamento farmacológico , Fatores de Virulência/farmacologia , ADP Ribose Transferases/sangue , ADP Ribose Transferases/química , Animais , Toxinas Bacterianas/sangue , Toxinas Bacterianas/química , Linhagem Celular Tumoral , Dissulfetos/sangue , Dissulfetos/química , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Exotoxinas/sangue , Exotoxinas/química , Meia-Vida , Humanos , Região Variável de Imunoglobulina/sangue , Região Variável de Imunoglobulina/química , Imunotoxinas/sangue , Imunotoxinas/química , Concentração Inibidora 50 , Mesotelina , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Oxirredução , Domínios Proteicos , Proteínas Recombinantes/sangue , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Relação Estrutura-Atividade , Microambiente Tumoral , Fatores de Virulência/sangue , Fatores de Virulência/química , Ensaios Antitumorais Modelo de Xenoenxerto , Exotoxina A de Pseudomonas aeruginosa
8.
Clin Cancer Res ; 22(19): 4913-4922, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27114443

RESUMO

PURPOSE: Recombinant immunotoxins (rITs) targeting CD22 are highly active in hairy cell leukemia, but less so in acute lymphoblastic leukemia (ALL). This study aims to understand the variable activity of an rIT against ALL toward improving responses in clinical application. EXPERIMENTAL DESIGN: We determined in vitro activity of rITs by WST-8 assays and the time needed to kill ALL cell lines and patient-derived ALL blasts by flow cytometry. The findings were translated into two systemic ALL xenograft models. Differences in time needed to kill KOPN-8 cells for distinct rITs were addressed biochemically. RESULTS: In vitro activity (IC50) of anti-CD22 rIT varied 210-fold from 0.02 to 4.6 ng/mL. Activity also varied greatly depending on the time ALL cells were exposed to immunotoxin from < 30 minutes to > 4 days. For KOPN-8, the difference in exposure time was related to intracellular rIT processing. We showed in newly developed ALL xenograft models, where immunotoxins have a short half-life, that the needed exposure time in vitro predicted the responses in vivo By replacing bolus dose with small doses at frequent intervals or with continuous infusion, responses were substantially improved. We confirmed exposure time variability on patient-derived ALL samples and showed a correlation between exposure time needed to reach maximal cytotoxicity in vitro and their clinical response. CONCLUSIONS: The exposure time needed for rITs targeting CD22 to kill ALL cells varies widely. Our results suggest that ALL patients would have a better response rate to anti-CD22 immunotoxins if treated by continuous infusion rather than by bolus injections. Clin Cancer Res; 22(19); 4913-22. ©2016 AACR.


Assuntos
Toxinas Bacterianas/farmacologia , Região Variável de Imunoglobulina/farmacologia , Imunotoxinas/farmacologia , Terapia de Alvo Molecular/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Projetos de Pesquisa , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/antagonistas & inibidores , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Reprod Sci ; 22(9): 1143-52, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25850899

RESUMO

Endometriosis is caused by the displacement of endometrium outside the uterus contributing heavily to infertility and debilitating pelvic pain. Ectopic adhesion and growth are believed to occur under the influence of a favorable hormonal environment and immunological factors. The objective of this study is to analyze the effect of a targeted therapy with an antibody-based pharmacodelivery of interleukin 4 (F8-IL4) in a mouse model of experimentally induced endometriosis. Endometriosis-like lesions were induced in Balb/c mice. The animals were treated intravenously with F8-IL4 or with untargeted IL4 (KSF-IL4). Twelve days after disease induction, the lesions were isolated. A significant reduction in the number of total lesions/mouse and in the total volume of lesions/mouse was observed in mice treated with F8-IL4 compared to controls (P = .029 and P = .006, respectively), while no difference was found between KSF-IL4-treated mice and their controls. Gene expression was evaluated by quantitative real-time polymerase chain reaction. Expression of genes involved in cell adhesion, extracellular matrix invasion, and neovascularization was significantly downregulated in F8-IL4-treated mice compared to their controls (integrin ß1: P = .02; metalloproteinase [MMP] 3: P = .02; MMP9: P = .04; vascular endothelial growth factor: P = .04). Gene expression of inflammatory cytokines (tumor necrosis factor α, IL1ß, IL1α, and IL6) did not vary in the ectopic lesions isolated from F8-IL4-treated mice compared to their controls. Immunohistochemistry demonstrated a significantly reduced expression of E-cadherin and ß-catenin in the lesions of mice treated with F8-IL4. Our results show that the antibody-mediated targeted delivery of IL4 inhibits the development of endometriosis in a syngeneic mouse model by likely impairing adhesion, invasion, and vascularization of the ectopic endometrium.


Assuntos
Anticorpos Monoclonais/farmacologia , Sistemas de Liberação de Medicamentos , Endometriose/prevenção & controle , Endométrio/efeitos dos fármacos , Imunoconjugados/farmacologia , Região Variável de Imunoglobulina/farmacologia , Interleucina-4/farmacologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Caderinas/genética , Caderinas/metabolismo , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Química Farmacêutica , Citocinas/genética , Citocinas/metabolismo , Citoproteção , Modelos Animais de Doenças , Endometriose/genética , Endometriose/metabolismo , Endometriose/patologia , Endométrio/irrigação sanguínea , Endométrio/metabolismo , Endométrio/patologia , Feminino , Regulação da Expressão Gênica , Imunoconjugados/administração & dosagem , Imunoconjugados/metabolismo , Região Variável de Imunoglobulina/administração & dosagem , Região Variável de Imunoglobulina/metabolismo , Mediadores da Inflamação/metabolismo , Injeções Intravenosas , Interleucina-4/administração & dosagem , Interleucina-4/metabolismo , Camundongos Endogâmicos BALB C , Neovascularização Patológica , Proteínas Recombinantes de Fusão/farmacologia , Fatores de Tempo , beta Catenina/genética , beta Catenina/metabolismo
10.
J Am Chem Soc ; 137(12): 4042-5, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25775396

RESUMO

The bovine antibody BLV1H12, which has an ultralong CDR3H, provides a novel scaffold for engineering new functions into the antibody's variable region. By modifying the ß-strand "stalk" of BLV1H12 with sequences derived from natural or synthetic protease inhibitors, we have generated antibodies that inhibit bovine trypsin and human neutrophil elastase (HNE) with low nanomolar affinities. We were also able to generate a humanized variant using a human immunoglobulin scaffold that shares a high degree of homology with BLV1H12. Further optimization yielded a highly selective humanized anti-HNE antibody with sub-nanomolar affinity. This work demonstrates a novel strategy for generating antibodies with potent and selective inhibitory activities against extracellular proteases involved in human disease.


Assuntos
Anticorpos/química , Anticorpos/farmacologia , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/farmacologia , Afinidade de Anticorpos , Bovinos , Humanos , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/farmacologia , Elastase de Leucócito/antagonistas & inibidores , Elastase de Leucócito/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Engenharia de Proteínas , Tripsina/imunologia , Inibidores da Tripsina/química , Inibidores da Tripsina/farmacologia
11.
MAbs ; 7(3): 605-19, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25764208

RESUMO

Interleukin-1 (IL-1) cytokines such as IL-1α, IL-1ß, and IL-1Ra contribute to immune regulation and inflammatory processes by exerting a wide range of cellular responses, including expression of cytokines and chemokines, matrix metalloproteinases, and nitric oxide synthetase. IL-1α and IL-1ß bind to IL-1R1 complexed to the IL-1 receptor accessory protein and induce similar physiological effects. Preclinical and clinical studies provide significant evidence for the role of IL-1 in the pathogenesis of osteoarthritis (OA), including cartilage degradation, bone sclerosis, and synovial proliferation. Here, we describe the generation and characterization of ABT-981, a dual variable domain immunoglobulin (DVD-Ig) of the IgG1/k subtype that specifically and potently neutralizes IL-1α and IL-1ß. In ABT-981, the IL-1ß variable domain resides in the outer domain of the DVD-Ig, whereas the IL-1α variable domain is located in the inner position. ABT-981 specifically binds to IL-1α and IL-1ß, and is physically capable of binding 2 human IL-1α and 2 human IL-1ß molecules simultaneously. Single-dose intravenous and subcutaneous pharmacokinetics studies indicate that ABT-981 has a half-life of 8.0 to 10.4 d in cynomolgus monkey and 10.0 to 20.3 d in rodents. ABT-981 exhibits suitable drug-like-properties including affinity, potency, specificity, half-life, and stability for evaluation in human clinical trials. ABT-981 offers an exciting new approach for the treatment of OA, potentially addressing both disease modification and symptom relief as a disease-modifying OA drug.


Assuntos
Anticorpos Neutralizantes/química , Imunoglobulina G/química , Região Variável de Imunoglobulina/química , Interleucina-1alfa/antagonistas & inibidores , Interleucina-1beta/antagonistas & inibidores , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Afinidade de Anticorpos , Especificidade de Anticorpos , Feminino , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/farmacologia , Interleucina-1alfa/química , Interleucina-1alfa/imunologia , Interleucina-1beta/química , Interleucina-1beta/imunologia , Camundongos
12.
MAbs ; 7(2): 440-50, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25679409

RESUMO

Panitumumab, as a commercially available antibody, is an effective anticancer therapeutic against epidermal growth factor receptor (EGFR), although it exerts weak antibody-dependent cell-mediated cytotoxicity (ADCC) activity owing to its IgG2 nature. Here, we firstly engineered panitumumab by grafting its variable region into an IgG1 backbone. The engineered panitumumab (denoted as Pan) retained binding activity identical to the parental antibody while exhibiting stronger ADCC activity in vitro and more potent antitumor effect in vivo. To further enhance the target selectivity of Pan, we generated Pan-P by tethering an epitope-blocking peptide to Pan via a tumor-specific protease selective linker. Pan-P showed almost 40-fold weaker affinity compared with Pan, but functional activity was restored to a similar extent as Pan when Pan-P was selectively activated by urokinase-type plasminogen activator (uPA). More importantly, targeted localization of Pan-P was observed in tumor samples from colorectal cancer (CRC) patients and tumor-bearing nude mice, strongly indicating that specific activation also existed ex vivo and in vivo. Furthermore, Pan-P also exhibited effective in vivo antitumor potency similar to Pan. Taken together, our data evidence the enhanced antitumor potency and excellent target selectivity of Pan-P, suggesting its potential use for minimizing on-target toxicity in anti-EGFR therapy.


Assuntos
Anticorpos Monoclonais , Anticorpos Antineoplásicos , Neoplasias Colorretais/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Região Variável de Imunoglobulina , Engenharia de Proteínas , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Antineoplásicos/genética , Anticorpos Antineoplásicos/imunologia , Anticorpos Antineoplásicos/farmacologia , Células CHO , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Cricetinae , Cricetulus , Receptores ErbB/genética , Receptores ErbB/imunologia , Feminino , Humanos , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Panitumumabe , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Mol Pharm ; 11(1): 81-9, 2014 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-24171467

RESUMO

A single chain Fraction variable (scFv) employs antibody-like target recognition specificity. Osteoclasts, responsible for bone resorption, express Receptor Activator of Nuclear factor Kappa B (RANK) receptors. This study aimed to express, characterize, and evaluate scFv against RANK receptors that may serve as a platform to target osteoclasts. Using phage display technology, scFv against RANK receptor was expressed and characterized by DNA sequencing, sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), matrix-assisted laser desorption-ionization time-of-flight (MALDI TOF), enzyme-linked immunosorbent assay (ELISA), Western blot, and immunocytochemistry. The potential for cytotoxicity was evaluated using an MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide) assay, and its cross reactivity was evaluated using ELISA. Osteoclast-like cells were generated from RAW 264.7 cells, and the osteoclast targeting ability of scFv was evaluated using immunocytochemistry. ScFv's antiresorptive efficacy was studied using a tartrate-resistant acid phosphatase (TRAP) assay and resorption assay. Anti-RANK scFv was successfully expressed and characterized. No cross reactivity with other tumor necrosis factor receptor (TNFR) members and no cytotoxic effect on a non-RANK bearing cell line were observed. It showed specificity toward a RANK receptor and an inhibitory effect on osteoclast activity. With the increase in development trends for biologics as therapeutics and growing knowledge on the importance of osteoclast targeted therapy, this study may provide a drug delivery strategy to target osteoclasts, thereby leading to a promising therapy for resorptive bone diseases.


Assuntos
Reabsorção Óssea , Sistemas de Liberação de Medicamentos , Região Variável de Imunoglobulina/farmacologia , Osteoclastos/efeitos dos fármacos , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Anticorpos de Cadeia Única/farmacologia , Sequência de Aminoácidos , Animais , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Técnicas Imunoenzimáticas , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/isolamento & purificação , Camundongos , Dados de Sequência Molecular , Osteoclastos/citologia , Osteoclastos/imunologia , Biblioteca de Peptídeos , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/isolamento & purificação , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
14.
J Hepatol ; 57(1): 17-23, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22414763

RESUMO

BACKGROUND & AIMS: Hepatitis C virus (HCV)-induced end-stage liver disease is currently the major indication for liver transplantation in the Western world. After transplantation, the donor liver almost inevitably becomes infected by the circulating virus and disease progression is accelerated in immune suppressed transplant patients. The current standard therapy, based on pegylated interferon and ribavirin, induces severe side effects and is often ineffective in this population. Therefore, new strategies to prevent graft re-infection are urgently needed. We have previously shown that monoclonal antibodies (mAbs) against the HCV co-receptor scavenger receptor class B type I (SR-BI/Cla1) inhibit infection by different HCV genotypes in cell culture. METHODS: Using phage display libraries, we have generated a large set of novel human mAbs against SR-BI and evaluated their effectiveness in preventing HCV infection and direct cell-to-cell spread in vitro and in vivo using uPA-SCID mice with a humanized liver. RESULTS: Eleven human monoclonal antibodies were generated that specifically recognize SR-BI. Two antibodies, mAb8 and mAb151, displayed the highest binding and inhibitory properties and also interfered with direct cell-to-cell spread in vitro. Studies in humanized mice showed that both antibodies were capable of preventing HCV infection and could block intrahepatic spread and virus amplification when administered 3 days after infection. Interestingly, anti-SR-BI therapy was effective against an HCV variant that escaped the control of the adaptive immune response in a liver transplant patient. CONCLUSIONS: The anti-SR-BI mAbs generated in this study may represent novel therapeutic tools to prevent HCV re-infection of liver allografts.


Assuntos
Anticorpos Monoclonais/farmacologia , Hepacivirus/imunologia , Hepatite C Crônica/prevenção & controle , Hepatócitos , Receptores Depuradores Classe B/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Células CHO , Cricetinae , Genótipo , Células Hep G2 , Hepacivirus/genética , Hepacivirus/crescimento & desenvolvimento , Hepatite C Crônica/imunologia , Hepatócitos/imunologia , Hepatócitos/transplante , Hepatócitos/virologia , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/farmacologia , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Biblioteca de Peptídeos , Quimeras de Transplante , Transplante Heterólogo
15.
J Biol Chem ; 287(18): 14912-22, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22334655

RESUMO

Complementarity-determining regions (CDRs) from monoclonal antibodies tested as synthetic peptides display anti-infective and antitumor activities, independent of the specificity of the native antibody. Previously, we have shown that the synthetic peptide C7H2, based on the heavy chain CDR 2 from monoclonal antibody C7, a mAb directed to a mannoprotein of Candida albicans, significantly reduced B16F10 melanoma growth and lung colony formation by triggering tumor apoptosis. The mechanism, however, by which C7H2 induced apoptosis in tumor cells remained unknown. Here, we demonstrate that C7H2 interacts with components of the tumor cells cytoskeleton, being rapidly internalized after binding to the tumor cell surface. Mass spectrometry analysis and in vitro validation revealed that ß-actin is the receptor of C7H2 in the tumor cells. C7H2 induces ß-actin polymerization and F-actin stabilization, linked with abundant generation of superoxide anions and apoptosis. Major phenotypes following peptide binding were chromatin condensation, DNA fragmentation, annexin V binding, lamin disruption, caspase 8 and 3 activation, and organelle alterations. Finally, we evaluated the cytotoxic efficacy of C7H2 in a panel of human tumor cell lines. All tumor cell lines studied were equally susceptible to C7H2 in vitro. The C7H2 amide without further derivatization significantly reduced lung metastasis of mice endovenously challenged with B16F10-Nex2 melanoma cells. No significant cytotoxicity was observed toward nontumorigenic cell lines on short incubation in vitro or in naïve mice injected with a high dose of the peptide. We believe that C7H2 is a promising peptide to be developed as an anticancer drug.


Assuntos
Actinas/imunologia , Anticorpos Monoclonais Murinos/farmacologia , Anticorpos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cadeias Pesadas de Imunoglobulinas/farmacologia , Região Variável de Imunoglobulina/farmacologia , Melanoma/prevenção & controle , Proteínas de Neoplasias/imunologia , Animais , Anticorpos Monoclonais Murinos/imunologia , Antineoplásicos/imunologia , Candida albicans/imunologia , Caspase 3/imunologia , Caspase 8/imunologia , Linhagem Celular Tumoral , Fragmentação do DNA/efeitos dos fármacos , DNA de Neoplasias/imunologia , Proteínas Fúngicas/imunologia , Humanos , Cadeias Pesadas de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/imunologia , Masculino , Melanoma/imunologia , Melanoma/patologia , Glicoproteínas de Membrana/imunologia , Camundongos , Metástase Neoplásica
16.
Toxicon ; 59(2): 272-82, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22126923

RESUMO

Tetrodotoxin (TTX) is a heterocyclic guanidinium alkaloid (C11H17N3O8) with molecular mass of ∼320 Da. The TTX and toxic analogs block sodium ion activity of mammalian nerve cells resulting in failure to conduct nerve impulse which manifested clinically in host as variable degrees of organ paralysis. Human intoxication occurs after consuming food containing the toxins. Current treatment of the poisoning is supportive and symptomatic. There has been no specific drug or antidote for the TTX mediated malady. In this study, phage clones displaying human single chain antibody fragments (HuScFv) were selected from a human ScFv phage display library. HuScFv derived from phagemid transformed Escherichia coli clones (clones s16 and s35) bound to the TTX as tested by indirect ELISA and band shift assay. Homology modeling and molecular docking revealed that VL domain of the s16-HuScFv interacted with the hydroxyl groups of C6, C9, C10 and C11 of the TTX by means of Tyr 223, Ser226 and Tyr228, while the Asp53 and Asp55 of the VH domain of s35-HuScFv interacted with the positions 1 and 2 of the guanidinium group and the hydroxyl groups at C9 and C10 of the TTX. The s16- and s35-HuScFv neutralized the TTX bioactivity in nerve cell based- and mouse bio-assays. Moreover, the HuScFv could rescue the intoxicated mice from the TTX mediated lethality. Thus, the HuScFv derived from the transformed E. coli clones have high potential as a safe, effective and specific therapeutic remedy for TTX intoxication in humans and warrant further trials.


Assuntos
Anticorpos de Cadeia Única/farmacologia , Canais de Sódio/efeitos dos fármacos , Tetrodotoxina/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Bioensaio , Cromatografia Líquida de Alta Pressão/métodos , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática/métodos , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Região Variável de Imunoglobulina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Polimorfismo de Fragmento de Restrição , Análise de Sequência de DNA , Canais de Sódio/metabolismo , Espectrometria de Massas em Tandem , Tetraodontiformes , Tetrodotoxina/toxicidade
17.
Neuroreport ; 22(9): 433-6, 2011 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-21544003

RESUMO

Cocaine and amphetamine-regulated transcript (CART) peptides are neurotransmitters with important roles in drug abuse. The increase of CART expression in the brain induced by psychostimulants is associated with changes of behavior in addicted animals. We expressed and purified the single-chain variable fragments antibody (scFv) against CART55-102, and observed the effect of CART scFv on the expression of cocaine-induced behavior sensitization in mice. Results showed that the titer of CART scFv was 1.6 µg/ml. Single administration of CART scFv (intraperitoneal 0.04, 0.2, and 1 mg/kg) reduced the increasing locomotor activity induced by chronic cocaine intake in mice (P<0.05-0.01), but failed to affect the locomotor activity of naive mice. These results suggested that CART scFv may be a potential therapeutic tool to treat drug abuse.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/metabolismo , Região Variável de Imunoglobulina/farmacologia , Atividade Motora/efeitos dos fármacos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Fragmentos de Peptídeos/antagonistas & inibidores , Anticorpos de Cadeia Única/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Tecido Nervoso/imunologia , Fragmentos de Peptídeos/imunologia
18.
Genes Immun ; 12(5): 360-9, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21326316

RESUMO

Administration of an artificial peptide (pConsensus) based on anti-DNA IgG sequences that contain major histocompatibility complex class I and class II T-cell determinants, induces immune tolerance in NZB/NZW F1 female (BWF1) mice. To understand the molecular basis of CD8(+) Ti-mediated suppression, we previously performed microarray analysis to identify genes that were differentially expressed following tolerance induction with pCons. CD8(+) T cells from mice tolerized with pCons showed more than two-fold increase in Ifi202b mRNA, an interferon inducible gene, versus cells from untolerized mice. Ifi202b expression increased through weeks 1-4 after tolerization and then decreased, reapproaching baseline levels at 6 weeks. In vitro polyclonal activation of tolerized CD8(+) T cells significantly increased Ifi202b mRNA expression. Importantly, silencing of Ifi202b abrogated the suppressive capacity of CD8(+) Ti cells. This was associated with decreased expression of Foxp3, and decreased gene and protein expression of transforming growth factor (TGF)ß and interleukin-2 (IL-2), but not of interferon (IFN)-γ, IL-10, or IL-17. Silencing of another IFN-induced gene upregulated in tolerized CD8(+) T cells, IFNAR1, had no effect on the ability of CD8(+) T cells to suppress autoantibody production. Our findings indicate a potential role for Ifi202b in the suppressive capacity of peptide-induced regulatory CD8(+) Ti cells through effects on the expression of Foxp3 and the synthesis of TGFß.


Assuntos
Anticorpos Antinucleares/biossíntese , Linfócitos T CD8-Positivos/imunologia , DNA/imunologia , Tolerância Imunológica , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Animais , Anticorpos Antinucleares/química , Linfócitos B/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Inativação Gênica , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/genética , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/farmacologia , Fatores Imunológicos/imunologia , Fatores Imunológicos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Camundongos , Camundongos Endogâmicos NZB , Peptídeos , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
19.
J Immunol Methods ; 366(1-2): 60-8, 2011 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-21277307

RESUMO

Aspergillus fumigatus causes the highly lethal form of invasive aspergillosis (IA). In the present study to develop a novel anti-fungal drug for protection against invasive disease, we identified a single chain fragment variable (scFv) antibody (scFv AF1) by panning against A. fumigatus membrane fraction (AMF) or HM-1 killer toxin (HM-1) neutralizing monoclonal antibody (nmAb-KT) as antigen. The key step was elution of bound phages with phosphate buffered saline (PBS) at pH 7.0 containing AMF. The specificity of soluble scFv AF1 antibody to antigens was verified by ELISA, which specifically binds to both AMF and nmAb-KT. After nucleotide sequencing, clone expression and purification by HisTrap HP affinity column, scFv AF1 showed in vitro anti-fungal activity against A. fumigatus. By SPR analysis it showed high binding affinity to nmAb-KT (K(d)=5.22×10(-11) M). The method used to isolate scFv AF1 was a new method and we believe that it will be applicable to isolate the specific scFv against any kind of membrane protein of yeast or fungus.


Assuntos
Anticorpos Anti-Idiotípicos/isolamento & purificação , Anticorpos Antifúngicos/isolamento & purificação , Aspergillus fumigatus/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Anti-Idiotípicos/genética , Anticorpos Anti-Idiotípicos/farmacologia , Anticorpos Antifúngicos/genética , Anticorpos Antifúngicos/farmacologia , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Neutralizantes/farmacologia , Afinidade de Anticorpos , Especificidade de Anticorpos , Antifúngicos/isolamento & purificação , Antifúngicos/farmacologia , Antígenos de Fungos , Aspergillus fumigatus/patogenicidade , Sequência de Bases , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Proteínas Fúngicas/imunologia , Humanos , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/isolamento & purificação , Fragmentos de Imunoglobulinas/farmacologia , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/isolamento & purificação , Região Variável de Imunoglobulina/farmacologia , Técnicas In Vitro , Fatores Matadores de Levedura/imunologia , Proteínas de Membrana/imunologia , Camundongos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície
20.
J Biol Chem ; 286(5): 3707-16, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-20980262

RESUMO

Interfering intracellular antibodies are valuable for biological studies as drug surrogates and as potential macromolecular drugs per se. Their application is still limited because of the difficulty of acquisition of functional intracellular antibodies. We describe the use of the new intracellular antibody capture procedure (IAC(3)) to facilitate direct isolation of functional single domain antibody fragments using four independent target molecules (LMO2, TP53, CRAF1, and Hoxa9) from a set of diverse libraries. Initially, these have variability in only one of the three antigen-binding CDR regions of VH or VL and first round single domains are affinity matured by iterative randomization of the two other CDRs and reselection. We highlight the approach using a single domain binding to LMO2 protein. Our results show that interfering with LMO2 protein function demonstrates a role specifically in erythroid differentiation, confirm a necessary and sufficient function for LMO2 as a cancer therapy target in T-cell neoplasia and allowed for the first time production of soluble recombinant LMO2 protein by co-expression with intracellular domain antibodies. Co-crystallization of LMO2 and the anti-LMO2 VH protein was successful. These results demonstrate that this third generation IAC(3) offers a robust toolbox for various biomedical applications and consolidates functional features of the LMO2 protein complex, which includes the importance of Lmo2-Ldb1 protein interaction.


Assuntos
Proteínas de Ligação a DNA/imunologia , Região Variável de Imunoglobulina/farmacologia , Metaloproteínas/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Anticorpos , Antineoplásicos/imunologia , Diferenciação Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Células Eritroides , Cadeias Pesadas de Imunoglobulinas , Região Variável de Imunoglobulina/uso terapêutico , Proteínas com Domínio LIM , Metaloproteínas/metabolismo , Metaloproteínas/fisiologia , Camundongos , Biblioteca de Peptídeos , Ligação Proteica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...