Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 10(12): e0145078, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26674639

RESUMO

Defective clearance of apoptotic cells can result in sustained inflammation and subsequent autoimmunity. Macrophages, the "professional phagocyte" of the body, are responsible for efficient, non-phlogistic, apoptotic cell clearance. Controlling phagocytosis of apoptotic cells by macrophages is an attractive therapeutic opportunity to ameliorate inflammation. Using high content imaging, we have developed a system for evaluating the effects of antibody treatment on apoptotic cell uptake in primary human macrophages by comparing the Phagocytic Index (PI) for each antibody. Herein we demonstrate the feasibility of evaluating a panel of antibodies of unknown specificities obtained by immunization of mice with primary human macrophages and show that they can be distinguished based on individual PI measurements. In this study ~50% of antibodies obtained enhance phagocytosis of apoptotic cells while approximately 5% of the antibodies in the panel exhibit some inhibition. Though the specificities of the majority of antibodies are unknown, two of the antibodies that improved apoptotic cell uptake recognize recombinant MerTK; a receptor known to function in this capacity in vivo. The agonistic impact of these antibodies on efferocytosis could be demonstrated without addition of either of the MerTK ligands, Gas6 or ProS. These results validate applying the mechanism of this fundamental biological process as a means for identification of modulators that could potentially serve as therapeutics. This strategy for interrogating macrophages to discover molecules regulating apoptotic cell uptake is not limited by access to purified protein thereby increasing the possibility of finding novel apoptotic cell uptake pathways.


Assuntos
Anticorpos/imunologia , Apoptose , Macrófagos/imunologia , Fagocitose , Animais , Anticorpos/classificação , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/imunologia , c-Mer Tirosina Quinase
2.
Front Pharmacol ; 5: 225, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25339905

RESUMO

The neonatal Fc receptor (FcRn) in intestinal epithelium is the primary mechanism for transfer of maternal immunoglobulin G (IgG) from suckled milk to serum; but the factors contributing to the rapid uptake of IgG are poorly understood. These studies help to determine the contribution of cell surface FcRn in IgG uptake in 2-week-old rat pups by varying local pH and binding conditions. Variants of a human wild-type (WT) IgG monoclonal antibody (mAb WT) were assessed for binding affinity (KD) to rat (r)FcRn at pH 6.0 and subsequent off-rate at pH 7.4 (1/s) by surface plasmon resonance. Selected mAbs were administered intra-intestinally in isoflurane-anesthetized 2-week rat pups. Full length mAb in serum was quantified by immunoassay, (r)FcRn mRNA expression by reverse transcription polymerase chain reaction, and mAb epithelial localization was visualized by immunohistochemistry. After duodenal administration, serum levels of mAb variants correlated with their rFcRn off-rate at pH 7.4, but not their affinity at pH 6.0. The greatest serum levels of IgG were measured when mAb was administered in the duodenum where rFcRn mRNA expression is greatest, and was increased further by duodenal administration in pH 6.0 buffer. More intense human IgG immunostaining was detected in epithelium than the same variant administered at higher pH. These data suggest an increased contribution for cell surface receptor. We conclude that, in the neonate duodenum, receptor off-rates are as important as affinities for FcRn mediated uptake, and cell surface binding of IgG to rFcRn plays contributes to IgG uptake alongside pinocytosis; both of which responsible for increased IgG uptake.

3.
Pharm Res ; 31(4): 908-22, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24072267

RESUMO

PURPOSE: To evaluate transcytosis of immunoglobulin G (IgG) by the neonatal Fc receptor (FcRn) in adult primate intestine to determine whether this is a means for oral delivery of monoclonal antibodies (mAbs). METHODS: Relative regional expression of FcRn and localization in human intestinal mucosa by RT-PCR, ELISA & immunohistochemistry. Transcytosis of full-length mAbs (sandwich ELISA-based detection) across human intestinal segments mounted in Ussing-type chambers, human intestinal (caco-2) cell monolayers grown in transwells, and serum levels after regional intestinal delivery in isoflurane-anesthetized cynomolgus monkeys. RESULTS: In human intestine, there was an increasing proximal-distal gradient of mucosal FcRn mRNA and protein expression. In cynomolgus, serum mAb levels were greater after ileum-proximal colon infusion than after administration to stomach or proximal small intestine (1-5 mg/kg). Serum levels of wild-type mAb dosed into ileum/proximal colon (2 mg/kg) were 124 ± 104 ng/ml (n = 3) compared to 48 ± 48 ng/ml (n = 2) after a non-FcRn binding variant. In vitro, mAb transcytosis in polarized caco-2 cell monolayers and was not enhanced by increased apical cell surface IgG binding to FcRn. An unexpected finding in primate small intestine, was intense FcRn expression in enteroendocrine cells (chromagranin A, GLP-1 and GLP-2 containing). CONCLUSIONS: In adult primates, FcRn is expressed more highly in distal intestinal epithelial cells. However, mAb delivery to that region results in low serum levels, in part because apical surface FcRn binding does not influence mAb transcytosis. High FcRn expression in enteroendocrine cells could provide a novel means to target mAbs for metabolic diseases after systemic administration.


Assuntos
Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/biossíntese , Imunoglobulina G/metabolismo , Mucosa Intestinal/metabolismo , Receptores Fc/biossíntese , Transcitose/fisiologia , Adulto , Animais , Células CACO-2 , Feminino , Humanos , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , RNA Mensageiro/biossíntese , Ratos , Adulto Jovem
4.
Curr Drug Discov Technol ; 11(1): 65-73, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23978038

RESUMO

Since Kohler and Milstein developed the process of generating hybridomas by fusing antibody secreting B cells with an immortal myeloma cell line, the techniques used to develop monoclonal antibodies for use as human therapeutics have progressed significantly. Here, we will briefly review hybridoma technology and the evolution of therapeutic antibodies for the treatment of human disease. We will focus on the evolution of humanized mouse models for the generation of therapeutic human antibodies, comparing the early models, such as severe combined immunodeficient (SCID) mice which do not engraft human leukocytes well due to residual innate immunity, to the more recently developed models such as non-obese diabetic (NOD)/SCID IL-2Rγ-deficient mice in which numerous human hematopoietic lineages can be cultivated. Building on the identification of suitable host strains for the reconstitution of human immune cells, focus has now shifted onto humanizing the murine microenvironment in order to support human immune cell function. Although several recent studies have shown that the provision of human soluble factors can support maturation and function of human immune cells, particularly within the myeloid compartment, this does not appear to impact antibody production significantly. Moreover, models in which grafting of human tissues is performed to provide human microenvironments which support human leukocyte maturation do show improved humoral immune function, but require several surgical manipulations for generation of the model. Ultimately the most desirable scenario is to generate transgenic models that can be bred efficiently and express a sufficient number of human molecules to support functional human immune cells and several groups have made progress in making this idea a reality. These studies in the context of the generation of human antibodies will be discussed.


Assuntos
Anticorpos Monoclonais , Animais , Descoberta de Drogas , Transplante de Células-Tronco Hematopoéticas , Humanos , Hibridomas , Camundongos
5.
Ann N Y Acad Sci ; 1263: 1-12, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22823582

RESUMO

Since the original publication of the article "Development of the IL-12/23 antagonist ustekinumab in psoriasis: Past, present and future perspectives" in March 2011 (see Appendix),(1) there have been several new publications and developments of note. A number of new reports from the ustekinumab psoriasis clinical development program have been published. The analysis of efficacy and safety in the PHOENIX 1 long-term extension demonstrated that continuous stable maintenance dosing of ustekinumab was generally well tolerated and sustained durable efficacy through up to three years of treatment.(2) Pooled safety data from the phase 2 and phase 3 global trials showed that the safety profile of long-term continuous ustekinumab treatment through up to three years(3,4) and four years(5) of follow-up was favorable and comparable to what has been reported previously in the shorter-term ustekinumab psoriasis studies.(6-8) This represents the greatest exposure and longest follow-up of psoriasis patients treated with a biologic published to date. Additional phase 3 trials in Asian populations demonstrated similar high levels of efficacy and favorable safety profiles in Japanese,(9,10) Korean,(11,12) and Taiwanese(11,12) patients as those observed in trials conducted in mostly White populations in North America and Europe.(6-8) These data support the positive benefit:risk profile and consistency of response to ustekinumab over years of usage, and in multiple ethnic groups. Results from up to five years of treatment with ustekinumab in the long-term extensions of the phase 3 trials, and the efficacy, safety, and effect on quality of life in Chinese patients will be available in 2012. In addition to clinical trials of ustekinumab for the treatment of psoriasis, 24-week data from one phase 3 study of ustekinumab for the treatment of psoriatic arthritis has recently been presented(13) and another study is ongoing. A Phase 2b trial in Crohn's disease has also been presented,(14) and three phase 3 studies in Crohn's disease are currently in progress.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Descoberta de Drogas/tendências , Interleucina-12/antagonistas & inibidores , Interleucina-23/antagonistas & inibidores , Psoríase/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados , Descoberta de Drogas/métodos , Previsões , Humanos , Psoríase/imunologia , Transdução de Sinais/imunologia , Resultado do Tratamento , Ustekinumab
6.
MAbs ; 4(3): 319-25, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22531448

RESUMO

The successful development of antibody therapeutics depends on the molecules having properties that are suitable for manufacturing, as well as use by patients. Because high solubility is a desirable property for antibodies, screening for solubility has become an essential step during the early candidate selection process. In considering the screening process, we formed a hypothesis that hybridoma antibodies are filtered by nature to possess high solubility and tested this hypothesis using a large number of murine hybridoma-derived antibodies. Using the cross-interaction chromatography (CIC) method, we screened the solubility of 92 murine hybridoma-derived monoclonal antibodies and found that all of these molecules exhibited CIC profiles that are indicative of high solubility (> 100mg/mL). Further investigations revealed that variable region N-linked glycosylation or isoelectric parameters are unlikely to contribute to the high solubility of these antibodies. These results support the general hypothesis that hybridoma monoclonal antibodies are highly soluble.


Assuntos
Anticorpos Monoclonais/química , Hibridomas/metabolismo , Animais , Anticorpos Monoclonais/isolamento & purificação , Técnicas de Imunoadsorção , Focalização Isoelétrica , Camundongos , Solubilidade
7.
MAbs ; 3(6): 535-45, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22123062

RESUMO

Monoclonal antibody (mAb) therapy was first established upon the approval of a mouse antibody for treatment of human acute organ rejection. However, the high incidence of immune response against the mouse mAb restricted therapeutic utility. Development of chimeric, "humanized" and human mAbs broadened therapeutic application to immune-mediated diseases requiring long-term treatment. Indeed, mAb therapeutics targeting soluble cytokines are highly effective in numerous immune-mediated disorders. A recent example is ustekinumab, a first-in-class therapeutic human immunoglobulin G1 kappa mAb that binds to the interleukins (IL)-12 and IL-23, cytokines that modulate lymphocyte function, including T-helper (Th) 1 and Th17 cell subsets. Ustekinumab was generated via recombinant human IL-12 immunization of human immunoglobulin (hu-Ig) transgenic mice. Ustekinumab binds to the p40 subunit common to IL-12 and IL-23 and prevents their interaction with the IL-12 receptor ß1 subunit of the IL-12 and IL-23 receptor complexes. Ustekinumab is approved for treatment of moderate-to-severe plaque psoriasis and has demonstrated efficacy in Crohn disease and psoriatic arthritis. The clinical characterization of ustekinumab continues to clarify our understanding of human immune pathologies and may offer a novel therapeutic option for certain immune-mediated diseases.


Assuntos
Anticorpos Monoclonais/imunologia , Interleucina-12/imunologia , Interleucina-23/imunologia , Psoríase/terapia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Humanos , Camundongos , Psoríase/imunologia , Resultado do Tratamento , Ustekinumab
8.
Fibrogenesis Tissue Repair ; 4(1): 23, 2011 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22011363

RESUMO

BACKGROUND: Fibrocytes are a population of circulating bone-marrow-derived cells that express surface markers for leukocytes and mesenchymal cells, and are capable of differentiating into myofibroblasts. They have been observed at sites of active fibrosis and increased circulating numbers correlate with mortality in idiopathic pulmonary fibrosis (IPF). Inhibition of chemokine (C-C motif) receptor 2 (CCR2) during experimental models of lung fibrosis reduces lung collagen deposition, as well as reducing lung fibrocyte accumulation. The aim of the present study was to determine whether human and mouse fibrocytes express functional CCR2. RESULTS: Following optimized and identical human and murine fibrocyte isolation, both cell sources were shown to be positive for CCR2 by flow cytometry and this expression colocalized with collagen I and CD45. Human blood fibrocytes stimulated with the CCR2 ligand chemokine (C-C motif) ligand 2 (CCL2), demonstrated increased proliferation (P < 0.005) and differentiation into myofibroblasts (P < 0.001), as well as a chemotactic response (P < 0.05). Murine fibrocytes also responded to CCR2 stimulation, with CCL12 being more potent than CCL2. CONCLUSIONS: This study directly compares the functional responses of human and murine fibrocytes to CCR2 ligands, and following comparable isolation techniques. We have shown comparable biological effects, strengthening the translatability of the murine models to human disease with respect to targeting the CCR2 axis to ameliorate disease in IPF patients.

9.
Hybridoma (Larchmt) ; 30(2): 153-62, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21529288

RESUMO

ST2L is a transmembrane receptor that belongs to the IL-1 receptor family. The receptor is expressed on various cell types including Th2 cells, mast cells, basophils, growth-activated fibroblasts, and vascular endothelial cells. ST2L activation by its ligand IL-33 has been implicated in Th2-mediated immunity, inflammation, and allergic responses in vivo. Inhibition of ST2L activity can attenuate Th2-dominated immune responses such as lung eosinophilia, airway hyper-responsiveness, and arthritis in animal models. Here we report the generation and in vitro characterization of a panel of rat anti-mouse ST2L monoclonal antibodies. We demonstrate that the antibodies specifically bind to recombinant receptor protein and that a subset of the binders inhibits mouse ST2L activity in multiple in vitro assays. Four of the identified anti-mouse ST2L antibodies were shown to prevent IL-33 from binding to ST2L, down-regulate IL-33-induced NF-κB signaling, and neutralize the ability of IL-33 to stimulate mouse Th2 cell proliferation. The characterized monoclonal antibodies are important tools that will be used to study mouse ST2L receptor functionality in vivo.


Assuntos
Anticorpos Monoclonais , Interleucinas/imunologia , Receptores de Interleucina-1/imunologia , Proteínas Recombinantes/imunologia , Transdução de Sinais/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais/farmacologia , Especificidade de Anticorpos , Biotina/química , Biotina/metabolismo , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo , Células HEK293 , Humanos , Hibridomas/imunologia , Hibridomas/metabolismo , Hipersensibilidade/tratamento farmacológico , Hipersensibilidade/imunologia , Imunoconjugados/química , Imunoconjugados/metabolismo , Inflamação/tratamento farmacológico , Inflamação/imunologia , Interleucina-1/imunologia , Interleucina-33 , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Plasmídeos/genética , Plasmídeos/metabolismo , Ratos , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/imunologia , Células Th2/imunologia , Transfecção
10.
Ann N Y Acad Sci ; 1222: 30-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21434940

RESUMO

The development of ustekinumab as a first-in-class anti-interleukin (IL) 12/23p40 therapeutic agent for psoriasis represents an important example of modern and rational drug design and development. Psoriasis is a chronic, systemic, immune-mediated skin disorder with considerable clinical, psychosocial, and economic burden. Ustekinumab is a human monoclonal antibody (mAb) that binds the p40 subunit common to IL-12 and IL-23, key cytokines in psoriasis pathogenesis. The therapeutic mAb was developed using human gamma-1 immunoglobulin (IgG)-expressing transgenic mice, which created a molecule with endogenous IgG(1) biologic properties and low immunogenicity. Ustekinumab was well tolerated in clinical studies and yielded rapid, significant, and sustained efficacy plus improved quality of life/work performance and reduced depression/anxiety. Its pharmacologic properties afford the most convenient dosing regimen among approved biologics, representing a significant advancement in the treatment of moderate to severe psoriasis. Ustekinumab also holds promise for other immune-mediated disorders with significant unmet need.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Desenho de Fármacos , Interleucina-12/antagonistas & inibidores , Interleucina-23/antagonistas & inibidores , Psoríase/tratamento farmacológico , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais Humanizados , Descoberta de Drogas/tendências , Previsões , Humanos , Fatores Imunológicos/uso terapêutico , Interleucina-12/imunologia , Interleucina-23/imunologia , Camundongos , Modelos Biológicos , Ustekinumab
11.
Assay Drug Dev Technol ; 9(4): 420-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21294636

RESUMO

Interleukin-6 (IL-6) family of cytokines are multifunctional proteins that play an important role in host defenses, acute phase reactions, immune responses, hematopoiesis, and tumorigenesis. The cytokines are produced by various lymphoid and nonlymphoid cells and mediate their biological activity through initial low-affinity binding to cell surface receptors, which are specific for their respective ligands. Ligand-specific receptor binding results in the receptor heterodimerization with ubiquitously expressed signal-transducing transmembrane component gp130 followed by activation of the gp130-associated Janus kinase, which, in turn, phosphorylates signal transducer and activator of transcription 3 (STAT3). Phosphorylated STAT3 (pSTAT3) dimerizes and translocates to the nucleus, where it activates gene transcription. Activation of STAT3 is essential to IL-6 family-associated physiological effects. Therefore, the ability to assess STAT3 phosphorylation is important for drug discovery efforts targeting IL-6 family cytokines. Various reagents and technologies are available to detect the effect of IL-6 type cytokines in treated cells. The present study describes the development of two pSTAT3 detection assays: the high-throughput screening assay based on Meso-Scale Discovery technology, which utilizes electrochemoluminescent signal measurements for the detection of pSTAT3 in treated cell extracts, and the secondary characterization assay based on fluorescent imaging analysis, which monitors pSTAT3 nuclear translocation in cells after activation. We have successfully utilized these assays to screen a small library of secreted proteins and identified inducers of STAT3 phosphorylation. The results obtained in this study demonstrate that both assays are robust, reliable, and amenable to high-throughput screening applications.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Fator de Transcrição STAT3/análise , Bibliotecas de Moléculas Pequenas/análise , Linhagem Celular Tumoral , Receptor gp130 de Citocina/fisiologia , Descoberta de Drogas , Fluorescência , Humanos , Interleucina-6/fisiologia , Janus Quinases/fisiologia , Fosforilação , Proteínas/análise , Proteínas/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
12.
MAbs ; 2(4): 428-39, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20519961

RESUMO

We prepared and characterized golimumab (CNTO148), a human IgG1 tumor necrosis factor alpha (TNFα) antagonist monoclonal antibody chosen for clinical development based on its molecular properties. Golimumab was compared with infliximab, adalimumab and etanercept for affinity and in vitro TNFα neutralization. The affinity of golimumab for soluble human TNFα, as determined by surface plasmon resonance, was similar to that of etanercept (18 pM versus 11 pM), greater than that of infliximab (44 pM) and significantly greater than that of adalimumab (127 pM, p=0.018).  The concentration of golimumab necessary to neutralize TNFα-induced E-selectin expression on human endothelial cells by 50% was significantly less than those for infliximab (3.2 fold; p=0.017) and adalimumab (3.3-fold; p=0.008) and comparable to that for etanercept. The conformational stability of golimumab was greater than that of infliximab (primary melting temperature [Tm] 74.8 °C vs. 69.5 °C) as assessed by differential scanning calorimetry.  In addition, golimumab showed minimal aggregation over the intended shelf life when formulated as a high concentration liquid product (100 mg/mL) for subcutaneous administration.  In vivo, golimumab at doses of 1 and 10 mg/kg significantly delayed disease progression in a mouse model of human TNFα-induced arthritis when compared with untreated mice, while infliximab was effective only at 10 mg/kg. Golimumab also significantly reduced histological scores for arthritis severity and cartilage damage, as well as serum levels of pro-inflammatory cytokines and chemokines associated with arthritis. Thus, we have demonstrated that golimumab is a highly stable human monoclonal antibody with high affinity and capacity to neutralize human TNFα in vitro and in vivo.


Assuntos
Anticorpos Monoclonais/farmacologia , Artrite/imunologia , Cartilagem/efeitos dos fármacos , Imunoglobulina G/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Adalimumab , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Monoclonais Humanizados/farmacologia , Afinidade de Anticorpos , Artrite/induzido quimicamente , Cartilagem/patologia , Modelos Animais de Doenças , Progressão da Doença , Selectina E/genética , Selectina E/metabolismo , Etanercepte , Regulação da Expressão Gênica/efeitos dos fármacos , Hibridomas , Imunoglobulina G/isolamento & purificação , Mediadores da Inflamação/metabolismo , Infliximab , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Conformação Proteica , Receptores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/imunologia
13.
J Mol Biol ; 398(2): 214-31, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20226193

RESUMO

Humanization of a potent neutralizing mouse anti-human IL-13 antibody (m836) using a method called human framework adaptation (HFA) is reported. HFA consists of two steps: human framework selection (HFS) and specificity-determining residue optimization (SDRO). The HFS step involved generation of a library of m836 antigen binding sites combined with diverse human germline framework regions (FRs), which were selected based on structural and sequence similarities between mouse variable domains and a repertoire of human antibody germline genes. SDRO consisted of diversifying specificity-determining residues and selecting variants with improved affinity using phage display. HFS of m836 resulted in a 5-fold loss of affinity, whereas SDRO increased the affinity up to 100-fold compared to the HFS antibody. Crystal structures of Fabs in complex with IL-13 were obtained for m836, the HFS variant chosen for SDRO, and one of the highest-affinity SDRO variants. Analysis of the structures revealed that major conformational changes in FR-H1 and FR-H3 occurred after FR replacement, but none of them had an evident direct impact on residues in contact with IL-13. Instead, subtle changes affected the V(L)/V(H) (variable-light domain/variable-heavy domain) interface and were likely responsible for the 5-fold decreased affinity. After SDRO, increased affinity resulted mainly from rearrangements in hydrogen-bonding pattern at the antibody/antigen interface. Comparison with m836 putative germline genes suggested interesting analogies between natural affinity maturation and the engineering process that led to the potent HFA anti-human IL-13 antibody.


Assuntos
Anticorpos Neutralizantes/imunologia , Região Variável de Imunoglobulina/imunologia , Interleucina-13/antagonistas & inibidores , Interleucina-13/imunologia , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/genética , Reações Antígeno-Anticorpo , Sítios de Ligação , Cristalografia por Raios X , Humanos , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/genética , Camundongos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Alinhamento de Sequência
14.
Hybridoma (Larchmt) ; 27(1): 25-30, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18294073

RESUMO

ABSTRACT Herein we describe the use of an agonistic anti-murine CD40 MAb as a B cell proliferative agent to enhance the generation of monoclonal antibodies (MAbs) in Balb/c mice. While hybridoma technology has been validated repeatedly over the decades, little work has been described to improve upon the overall numbers of in vivo B cells and specific antibodies obtained from a fusion. To begin to address this situation, strategies to boost B lymphocyte yields for hybridoma production were employed. Anti-CD40 agonist antibodies have been reported to activate and amplify human resting B lymphocytes in vitro, resulting in increased cell numbers available for the generation of human hybridomas or B cell clones. An agonistic anti-murine CD40 MAb was administered to immunized mice 3 days prior to splenic harvest, and B lymphocyte yields were found to be approximately 2-fold higher in treated animals when compared to untreated animals. Moreover, the resulting hybridoma fusions using lymphocytes from treated animals yielded 5- to 10-fold more antigen reactive hybrids when compared to untreated animals. This novel addition to conventional approaches utilizes the proliferative effects of agonistic anti-CD40 MAbs to markedly enhance monoclonal antibody generation.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos CD40/agonistas , Hibridomas/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Antígenos CD40/imunologia , Fusão Celular , Feminino , Humanos , Hibridomas/citologia , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia , Baço/imunologia
15.
Cell Immunol ; 248(2): 103-14, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18048020

RESUMO

Toll-like receptors are a family of pattern-recognition receptors that contribute to the innate immune response. Toll-like receptor 3 (TLR3) signals in response to foreign, endogenous and synthetic ligands including viral dsRNA, bacterial RNA, mitochondrial RNA, endogenous necrotic cell mRNA and the synthetic dsRNA analog, poly(I:C). We have generated a monoclonal antibody (mAb CNTO2424) that recognizes the extracellular domain (ECD) of human TLR3 in a conformation-dependent manner. CNTO2424 down-regulates poly(I:C)-induced production of IL-6, IL-8, MCP-1, RANTES, and IP-10 in human lung epithelial cells. In addition, mAb CNTO2424 was able to interfere with the known TLR3-dependent signaling pathways, namely NF-kappaB, IRF-3/ISRE, and p38 MAPK. The generation of this neutralizing anti-TLR3 mAb provides a unique tool to better understand TLR3 signaling and potential cross-talk between TLR3 and other molecules.


Assuntos
Anticorpos Monoclonais , Receptor 3 Toll-Like/antagonistas & inibidores , Receptor 3 Toll-Like/imunologia , Animais , Anticorpos Bloqueadores/metabolismo , Anticorpos Monoclonais/metabolismo , Sítios de Ligação de Anticorpos , Linhagem Celular , Linhagem Celular Transformada , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Projetos Piloto , Receptor 3 Toll-Like/metabolismo
16.
Hum Antibodies ; 15(3): 61-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17065737

RESUMO

The generation of anti-variable region monoclonal antibodies (mAbs) against therapeutic antibodies is essential in the pharmacokinetic/pharmacodynamic (PK/PD) assessments of the drugs in clinical study samples. Sandwich EIA and other methods are typically employed to achieve sensitivity and selectivity for the PK/PD analyses. These assays usually require generation of mAb reagents that bind specifically to the drug in non-competing pair combinations. Thus, large panels of anti-variable region mAbs must be generated in an expeditious manner to increase the probability of success. Herein we describe a novel immunization method that utilizes type 1 interferons (IFNs) as immunomodulators coupled with an agonistic anti-CD40 mAb as a B cell proliferative agent to drive immune responses. This novel protocol allows for rapid and robust mAb reagent generation without the use of conventional protein-denaturing adjuvants. The use of IFNs allowed for the generation of comparable and in some cases, increased numbers of anti-variable region mAbs in a dramatically shorter timeframe. This IFN based, immunostimulatory approach utilizing a non-denaturing adjuvant, likely presents conformational epitopes and may optimize the humoral response for the rapid generation of anti-variable region reagents to therapeutic mAb candidates.


Assuntos
Adjuvantes Imunológicos/farmacologia , Anticorpos Monoclonais/biossíntese , Interferons/imunologia , Animais , Anticorpos Monoclonais/imunologia , Linfócitos B/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Sensibilidade e Especificidade , Baço/imunologia
17.
Hybridoma (Larchmt) ; 25(2): 68-74, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16704306

RESUMO

The selection of efficacious anti-tumor monoclonal antibodies (MAbs) for biological applications is a lengthy and labor-intensive process. In vitro characterization of one hybridoma fusion may reveal large numbers of tumor antigen-specific hybridomas. Very often, many of these tumor-specific antibodies need to be assessed in vivo using several different murine xenograft tumor prevention models to determine biological efficacy. The production and purification of sufficient quantities of many antigen-specific hybridomas is time-consuming, and several months can pass between initial determination of MAb specificity and bioactivity. Moreover, many tumor-specific MAbs selected using in vitro binding studies have no in vivo anti-tumor efficacy. These studies describe an in vivo screening method either to eliminate non-efficacious MAbs or to rank-order several tumor-specific MAbs in an expeditious manner. Proof-of-concept studies were conducted using two hybridomas secreting fully characterized neutralizing human anti-tumor MAbs (CNTO MAbs). Nu-/nu- mice were injected with CNTO MAb-secreting hybridoma cells in Matrigel cell matrix, followed by injection of target human tumor cells 4 days later (when circulating CNTO MAbs were detected in serum). Both the tumor take-rate and the mean tumor volumes were reduced significantly in mice treated with CNTO MAbsecreting hybridomas compared with mice treated with non-antibody-secreting cells. A panel of human antitumor antigen-specific MAbs with unknown biological efficacy was then evaluated by this method. The hybridomas exhibited a varied pattern of anti-tumor protection, indicating that some hybrids were secreting neutralizing anti-tumor MAbs, while others appeared to be less efficacious. These studies demonstrate a rapid, biologically relevant "yes/no" in vivo screening method for the evaluation of anti-tumor antigen MAbs.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Animais , Anticorpos Monoclonais/sangue , Antígenos de Neoplasias/imunologia , Antineoplásicos/isolamento & purificação , Linhagem Celular , Colágeno , Combinação de Medicamentos , Feminino , Humanos , Hibridomas/imunologia , Hibridomas/metabolismo , Laminina , Camundongos , Camundongos Nus , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Proteoglicanas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Virchows Arch ; 448(3): 295-300, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16323007

RESUMO

The regenerating protein (Reg)-like protein (RELP, also known as Reg IV) is a recently characterized fourth member of the human Reg protein family. The Reg proteins are small, about 20-kD-sized, secretory proteins of C-lectin type. The previously known Reg proteins have been functionally implicated in regeneration, proliferation, and differentiation of the pancreas, liver, and gastrointestinal mucosa. To study the tissue expression of RELP, we raised a monoclonal antibody to RELP. By immunohistochemistry and in situ hybridization, we found a robust de novo expression of RELP in the neoplastic goblet cells of appendiceal mucinous cystadenomas and in the epithelial implants of pseudomyxoma peritonei (PMP). Our findings indicate that RELP serves as a marker for appendiceal mucinous cystadenomas and PMP, and that RELP may contribute to the pathogenesis of these disorders.


Assuntos
Neoplasias do Apêndice/metabolismo , Cistadenoma Mucinoso/metabolismo , Células Caliciformes/metabolismo , Lectinas Tipo C/metabolismo , Pseudomixoma Peritoneal/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias do Apêndice/genética , Neoplasias do Apêndice/patologia , Biomarcadores Tumorais/metabolismo , Cistadenoma Mucinoso/genética , Cistadenoma Mucinoso/patologia , Feminino , Técnica Direta de Fluorescência para Anticorpo , Expressão Gênica , Células Caliciformes/patologia , Humanos , Técnicas Imunoenzimáticas , Hibridização In Situ , Lectinas Tipo C/genética , Masculino , Pessoa de Meia-Idade , Proteínas Associadas a Pancreatite , Pseudomixoma Peritoneal/genética , Pseudomixoma Peritoneal/patologia , RNA Mensageiro/metabolismo
19.
Hum Antibodies ; 15(4): 155-62, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17522437

RESUMO

The generation of anti-variable region monoclonal antibodies (mAbs) against therapeutic antibodies is essential in the pharmacokinetic/pharmacodynamic (PK/PD) assessments of the drugs in clinical study samples. Sandwich EIA and other methods are typically employed to achieve sensitivity and selectivity for the PK/PD analyses. These assays usually require generation of mAb reagents that bind specifically to the therapeutic mAb candidate in non-competing pair combinations. Thus, large panels of anti-variable region mAbs must be generated in an expeditious manner to increase the probability of success. Previously, we described a novel immunization method using type 1 interferons (IFNs) coupled with an agonistic anti-CD40 mAb to drive immune responses (Staquet et al., Human Antibodies 15 (2006), 61-69). This protocol allows for rapid and robust generation of large panels of anti-variable region mAbs. In order to quickly characterize and efficiently identify optimal anti-variable region antibody pairs early in the hybridoma process using crude supernatants, an inexpensive, high-throughput ELISA method was developed. The ability to rapidly identify appropriate mAb pairs will save resources by eliminating the time-consuming and laborious process of subcloning irrelevant hybridomas.


Assuntos
Anticorpos Monoclonais/biossíntese , Antígenos CD40/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Região Variável de Imunoglobulina/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Linfócitos B , Biotecnologia/métodos , Feminino , Humanos , Imunização , Imunoglobulina G/administração & dosagem , Imunoglobulina G/imunologia , Interferon Tipo I , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Tempo
20.
J Pharmacol Exp Ther ; 313(1): 8-15, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15644434

RESUMO

In vivo models have demonstrated that interleukin-13 (IL-13) plays an important role in asthma; however, few studies have evaluated the effect of inhibition of IL-13 on established and persistent disease. In the present study, we have investigated the effect of a therapeutic dosing regimen with an anti-IL-13 monoclonal antibody (mAb) in a chronic mouse model of persistent asthma. BALB/c mice were sensitized to allergen [ovalbumin (OVA); on days 1 and 8] and challenged with OVA weekly from day 22. Anti-IL-13 mAb or vehicle dosing was initiated following two OVA challenges when disease was established. At this time, mice exhibited airway hyperresponsiveness (AHR), increased mucus production, inflammation, and initiation of subepithelial fibrosis compared with saline-challenged mice. Mice received four additional OVA challenges. Treatment with anti-IL-13 mAb inhibited AHR and prevented the further development of subepithelial fibrosis and progression of inflammation. Furthermore, mAb treatment reversed the mucus hyperplasia to basal levels. These effects were associated with an inhibition of cytokines, chemokines, and matrix metalloproteinase-9. These data demonstrate that neutralization of IL-13 can inhibit the progression of established disease in the presence of repeated allergen exposures.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Asma/tratamento farmacológico , Interleucina-13/imunologia , Animais , Asma/patologia , Hiper-Reatividade Brônquica/fisiopatologia , Líquido da Lavagem Broncoalveolar/citologia , Quimiocinas/biossíntese , Citocinas/biossíntese , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Mediadores da Inflamação/fisiologia , Interleucina-13/fisiologia , Pulmão/patologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Muco/fisiologia , Ovalbumina/imunologia , Fibrose Pulmonar/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...