Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Methods ; 65(1): 114-26, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23872058

RESUMO

The Fc variant of IgG2, designated as IgG2σ, was engineered with V234A/G237A /P238S/H268A/V309L/A330S/P331S substitutions to eliminate affinity for Fcγ receptors and C1q complement protein and consequently, immune effector functions. IgG2σ was compared to other previously well-characterized Fc 'muted' variants, including aglycosylated IgG1, IgG2m4 (H268Q/V309L/A330S/P331S, changes to IgG4), and IgG4 ProAlaAla (S228P/L234A/L235A) in its capacity to bind FcγRs and activate various immune-stimulatory responses. In contrast to the previously characterized muted Fc variants, which retain selective FcγR binding and effector functions, IgG2σ shows no detectable binding to the Fcγ receptors in affinity and avidity measurements, nor any detectable antibody-dependent cytotoxicity, phagocytosis, complement activity, or Fc-mediated cytokine release. Moreover, IgG2σ shows minimal immunogenic potential by T-cell epitope analysis. The circulating half-life of IgG2σ in monkeys is extended relative to IgG1 and IgG2, in spite of similar in vitro binding to recombinant FcRn. The three-dimensional structure of the Fc, needed for assessing the basis for the absence of effector function, was compared with that of IgG2 revealing a number of conformational differences near the hinge region of the CH2 domain that result from the amino acid substitutions. Modeling reveals that at least one of the key interactions with FcγRs is disrupted by a conformational change that reorients P329 to a position that prevents it from interacting with conserved W90 and W113 residues of the FcγRs. Inspection of the structure also indicated significant changes to the conformations of D270 and P329 in the CH2 domain that could negatively impact C1q binding. Thus, structural perturbations of the Fc provide a rationale for the loss of function. In toto, these properties of IgG2σ suggest that it is a superior alternative to previously described IgG variants of minimal effector function, for future therapeutic applications of non-immunostimulatory mAb and Fc-fusion platforms.


Assuntos
Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/química , Fatores Imunológicos/química , Substituição de Aminoácidos , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Afinidade de Anticorpos , Citotoxicidade Celular Dependente de Anticorpos , Sítios de Ligação , Cristalografia por Raios X , Citocinas/metabolismo , Células HEK293 , Meia-Vida , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/farmacologia , Imunoglobulina G/genética , Imunoglobulina G/farmacologia , Fatores Imunológicos/genética , Fatores Imunológicos/farmacologia , Macaca fascicularis , Masculino , Modelos Moleculares , Mutagênese Sítio-Dirigida , Ligação Proteica , Estrutura Secundária de Proteína , Receptor ErbB-2/imunologia , Receptores de IgG/química
2.
Mol Immunol ; 44(7): 1524-34, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17045339

RESUMO

Although it is now clear that certain Fc glycan structures on immunoglobulin G (IgG) antibodies (Abs) can have a dramatic influence on binding to selected Fcgamma receptors (FcgammaR) and on Fc-mediated immune functions, the effects of all known Fc glycan structures still have not been exhaustively studied. We report that in vitro analyses of pairs of monoclonal human IgG Abs that differ in the amount of sialic acid in their Fc glycans revealed that, for each of the three Ab pairs we examined, higher levels of sialylation were associated with reduced activity in Ab-dependent cellular cytotoxicity (ADCC) assays. This relationship between sialylation and ADCC activity was observed regardless of whether the differences in the extent of sialylation were derived by different Ab production processes, use of a lectin column to separate monoclonal Ab preparations into differentially sialylated fractions, or use of direct in vitro glycoengineering methods to convert a lesser sialylated Ab into a highly sialylated Ab. Subsequent investigations revealed that, depending on the individual Ab and how the differences in sialylation were derived, the lower ADCC potency of the more sialylated variants was apparently due to lower-affinity binding to FcgammaRIIIa on natural killer (NK) cells and/or, more interestingly, lower-affinity binding to cell-surface antigen. Our data provide the first example of an Fc glycan structure impacting antigen binding and suggest that avoiding Fc glycan sialylation can offer another means of optimizing ADCC activity of Abs.


Assuntos
Citotoxicidade Imunológica , Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Ácido N-Acetilneuramínico/análise , Polissacarídeos/química , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Antígenos/imunologia , Sequência de Carboidratos , Células Cultivadas , Testes Imunológicos de Citotoxicidade , Humanos , Fragmentos Fc das Imunoglobulinas/química , Imunoglobulina G/química , Dados de Sequência Molecular , Engenharia de Proteínas , Receptores de IgG/imunologia
3.
Int Immunopharmacol ; 7(6): 761-72, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17466910

RESUMO

Although it has been shown that functions of immunoglobulin G (IgG) antibodies (Abs) that depend on binding to certain Fc gamma receptors (Fc gamma R) can be influenced by Fc glycan fucosylation, quantitative in vivo analyses comparing the effects of different levels of fucose are still lacking. We used a simple mouse model to compare Fc gamma R-dependent T cell activation induced by different fucosylation variants of a hamster/human IgG1 chimeric version of anti-mouse CD3 monoclonal Ab, 145-2C11 (2C11). Initial studies supported the expectation that this agonist activity by 2C11 was a reflection of Fc gamma R binding, including comparisons of human IgG1 and IgG4 variants of 2C11 that showed the IgG4 to be dramatically less active at inducing T cell activation. Dose-response analyses in mice then showed that a sample of the human IgG1 version of 2C11 Ab in which 40% of the Fc glycans in the population of Ab molecules were fucosylated was 3-5 times more potent than a sample with 90% of its Fc glycans fucosylated. A sample with 10% fucosylation showed the same activity as the 40% fucosylated sample, revealing that complete absence of fucose was not necessary to achieve maximal Fc function in this model. In vitro binding to recombinant mouse Fc gamma Rs by the 2C11 variants revealed interesting relationships between fucose content and receptor affinity, and suggested the involvement of Fc gamma RIV in mediating 2C11 activity in vivo. These analyses showed that low-fucose human IgG1 Abs indeed show greater Fc gamma R-dependent activities in mice, but that Abs with moderate levels of fucose may be just as potent as Abs with very low or no fucose.


Assuntos
Anticorpos Monoclonais/farmacologia , Fucose/imunologia , Imunoglobulina G/imunologia , Receptores de IgG/imunologia , Animais , Complexo CD3/efeitos dos fármacos , Complexo CD3/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia , Baço/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
4.
Biotechnol Prog ; 23(4): 964-71, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17571902

RESUMO

Glycosylation in the CH2 domain of Fc is required for immunoglobulins G (IgGs) to exhibit immune effector functions including complement-dependent cytotoxicity (CDC) and antibody-dependent (Ab-dependent) cellular cytotoxicity (ADCC). We recently established that glycosylated Abs are more resistant to papain digestion than non-glycosylated IgGs (Biochem. Biophys. Res. Commun. 2006, 341, 797-803). To test whether specific Fc glycan structures affect Ab resistance to papain, we used in vitro glycoengineering methods to prepare homogeneous Ab glycoforms terminated with either sialic acid (G2S2), beta-galactose (G2), or N-acetylglucosamine (G0) and subjected them to papain digestions. Analyses of aliquots taken at different times during the digestions by matrix-assisted laser desorption-time-of-flight-mass spectroscopy (MALDI-TOF-MS) and high-performance liquid chromatography (HPLC) methods showed that the G0 glycoform was at least two times more resistant to papain digestion than the G2 and G2S2 glycoforms. The increased resistance of the G0 glycoform over the G2 and G2S2 glycoforms was independent of the specific Ab analyzed. A mouse/human chimeric version of Ab1, a fully human version of Ab2, and a humanized version of Ab3 exhibited a similar pattern of glycoform-dependent resistance. These data suggest that terminal sugars of Fc glycans may play important roles in Ab stability and affect resistance to proteases in addition to impacting Ab effector functions.


Assuntos
Acetilglucosamina/química , Imunoglobulina G/química , Polissacarídeos/química , Animais , Carboidratos/química , Cromatografia/métodos , Cromatografia Líquida de Alta Pressão , Glicosilação , Humanos , Camundongos , Modelos Químicos , Oligossacarídeos/química , Papaína/química , Engenharia de Proteínas , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
5.
Antibodies (Basel) ; 6(3)2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31548527

RESUMO

Engineering of fragment crystallizable (Fc) domains of therapeutic immunoglobulin (IgG) antibodies to eliminate their immune effector functions while retaining other Fc characteristics has numerous applications, including blocking antigens on Fc gamma (Fcγ) receptor-expressing immune cells. We previously reported on a human IgG2 variant termed IgG2σ with barely detectable activity in antibody-dependent cellular cytotoxicity, phagocytosis, complement activity, and Fcγ receptor binding assays. Here, we extend that work to IgG1 and IgG4 antibodies, alternative subtypes which may offer advantages over IgG2 antibodies. In several in vitro and in vivo assays, the IgG1σ and IgG4σ variants showed equal or even lower Fc-related activities than the corresponding IgG2σ variant. In particular, IgG1σ and IgG4σ variants demonstrate complete lack of effector function as measured by antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, antibody-dependent cellular phagocytosis, and in vivo T-cell activation. The IgG1σ and IgG4σ variants showed acceptable solubility and stability, and typical human IgG1 pharmacokinetic profiles in human FcRn-transgenic mice and cynomolgus monkeys. In silico T-cell epitope analyses predict a lack of immunogenicity in humans. Finally, crystal structures and simulations of the IgG1σ and IgG4σ Fc domains can explain the lack of Fc-mediated immune functions. These variants show promise for use in those therapeutic antibodies and Fc fusions for which the Fc domain should be immunologically "silent".

6.
Mol Immunol ; 41(1): 73-80, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15140577

RESUMO

The functional valency of a monoclonal antibody (mAb) has important influences on such things as antigen avidity, Fc-mediated immune effector functions, and clearance of immune complexes. cV1q, a neutralizing rat/mouse chimeric anti-mouse tumor necrosis factor (TNF) monoclonal antibody (mAb), and Rt108, a neutralizing mouse anti-rat TNF (anti-raTNF) mAb, appear to be functionally monovalent for TNF-binding despite containing two antigen binding sites. The functional monovalency of these two independent anti-rodent TNF mAbs is presumably a result of steric hindrance from one TNF molecule binding to one Fab arm that prevents binding of a second TNF molecule to the other Fab arm. To test whether this steric hindrance could be overcome by introducing extra space and flexibility between the Fab arms, these mAbs were engineered to contain an extra CH1 immunoglobulin domain between the CH1 and hinge domains of their heavy chains. In vitro binding data showed that, compared to the original mAbs, the modified mAbs (S-mAbs) had greater capability of binding two TNF molecules simultaneously. In vitro activity assays showed that, compared to the original mAbs, the S-mAbs had significantly greater TNF-neutralization potency, with the S-mAb version of cV1q (S-cV1q) being 200-fold more effective at blocking mouse TNF (muTNF) and the S-mAb version of Rt108 (S-Rt108) being 20-fold more effective at blocking raTNF. Similar results were observed in vivo, where S-cV1q was between 100- and 500-fold more protective than cV1q in mice challenged with endotoxin. These data reveal that introduction of another constant region immunoglobulin domain into two unrelated mAbs dramatically enhanced their neutralization potency. Other mAbs may also show more potent activity using this engineering approach, particularly mAbs that recognize homopolymeric antigens.


Assuntos
Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Imunoglobulinas/química , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacocinética , Sítios de Ligação de Anticorpos , Linhagem Celular , Camundongos , Testes de Neutralização , Engenharia de Proteínas , Estrutura Terciária de Proteína , Ratos , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/toxicidade
7.
Mol Cancer Ther ; 2(5): 445-51, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12748306

RESUMO

The proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) was originally considered to have activity against malignant disease. However, recent studies suggest TNF-alpha may also act as an endogenous tumor promoter. In the present work, mice deficient in TNF-alpha either genetically (TNF-alpha(-/-)) or after blockade with a neutralizing antibody (cV1q) were used to investigate the role of TNF-alpha in skin tumor development. Papillomas were induced in wild-type (wt) mice after treatment of skin with the initiating agent 9,10-dimethyl-1,2-benzanthracene followed by promotion with 12-O-tetradecanoylphorbol-13-acetate (TPA) for 15 weeks. TNF-alpha(-/-) mice were resistant to papilloma development when compared with wt mice on C57Bl/6J, 129/SvEv, and BALB/c genetic backgrounds. Primary murine keratinocytes (newborn keratinocytes) and skin homogenates were used to characterize TPA-stimulated TNF-alpha expression. TPA induced TNF-alpha protein in newborn keratinocytes in vitro and epidermis in vivo. Neutralization of TNF-alpha protein with cV1q in vivo for 0-15 weeks of promotion significantly decreased skin tumor development after 9,10-dimethyl-1,2-benzanthracene/TPA treatment. cV1q treatment during the early stages of tumor promotion (0-6 weeks) was equally effective. These data suggest that early induction of TNF-alpha is critical for skin tumor promotion. cV1q also reduced TPA-stimulated expression of matrix metalloproteinase 9 and granulocyte macrophage colony-stimulating factor, proteins that are differentially regulated in wt and TNF-alpha(-/-) epidermis. Treatment of the 410.4 transplantable breast carcinoma with cV1q reduced tumor growth in vivo, illustrating that inhibition of tumor growth through neutralization of TNF-alpha is not limited to skin carcinogenesis. These results provide further evidence for procancer actions of TNF-alpha and give some rationale for use of TNF-alpha antagonists in cancer prevention and treatment.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Papiloma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Fator de Crescimento Transformador alfa/imunologia , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Carcinógenos/toxicidade , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Imunidade Inata , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estadiamento de Neoplasias , Papiloma/induzido quimicamente , Papiloma/metabolismo , Ratos , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/metabolismo , Acetato de Tetradecanoilforbol/toxicidade
8.
Influenza Other Respir Viruses ; 8(5): 596-604, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25074755

RESUMO

BACKGROUND: Ferrets have long been used as a disease model for the study of influenza vaccines, but a more recent use has been for the study of human monoclonal antibodies directed against influenza viruses. Published data suggest that human antibodies are cleared unusually quickly from the ferret and that immune responses may be partially responsible. This immunogenicity increases variability within groups and may present an obstacle to long-term studies. OBJECTIVE: Our aim was to identify an antibody design with reduced immunogenicity and longer circulating half-life in ferrets. METHODS: The constant region coding sequences for ferret immunoglobulin G were cloned, and chimeric human/ferret antibodies were expressed and purified. Some of the chimeric antibodies included substitutions that have been shown to extend the half-life of human IgG antibodies. These chimeric antibodies were tested for binding to recombinant ferret FcRn receptor and then evaluated in pharmacokinetic studies in ferrets. RESULTS: A one-residue substitution in the ferret Fc domain, S252Y, was identified that increased binding affinity to the ferret neonatal receptor by 24-fold and extended half-life from 65 ± 27 to 206 ± 28 hours or ~9 days. Ferrets dosed twice with this surrogate antibody showed no indications of an immune response. CONCLUSION: Expressing the variable region of a candidate human therapeutic antibody with ferret constant regions containing the S252Y substitution can offer long half-life and limit immunogenicity.


Assuntos
Furões/imunologia , Imunoglobulina G/química , Imunoglobulina G/imunologia , Sequência de Aminoácidos , Animais , Afinidade de Anticorpos , Furões/sangue , Furões/genética , Meia-Vida , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/genética , Masculino , Dados de Sequência Molecular , Alinhamento de Sequência
9.
MAbs ; 5(3): 397-405, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23549129

RESUMO

Transgenic mice expressing human neonatal Fc receptor (FcRn) instead of mouse FcRn are available for IgG antibody pharmacokinetic (PK) studies. Given the interest in a rodent model that offers reliable predictions of antibody PK in monkeys and humans, we set out to test whether the PK of IgG antibodies in such mice correlated with the PK of the same antibodies in primates. We began by using a single research antibody to study the influence of: (1) different transgenic mouse lines that differ in FcRn transgene expression; (2) homozygous vs. hemizygous FcRn transgenic mice; (3) the presence vs. absence of coinjected high-dose human intravenous immunoglobulin (IVIG), and (4) the presence vs. absence of coinjected high-dose human serum albumin (HSA). Results of those studies suggested that use of hemizygous Tg32 mice (Tg32 hemi) not treated with IVIG or HSA offered potential as a predictive model for PK in humans. Mouse PK studies were then done under those conditions with a panel of test antibodies whose PK in mice and primates is not significantly affected by target binding, and for which monkey or human PK data were readily available. Results from the studies revealed significant correlations between terminal half-life or clearance values observed in the mice and the corresponding values reported in humans. A significant relationship in clearance values between mice and monkeys was also observed. These correlations suggest that the Tg32 hemi mouse model, which is both convenient and cost-effective, can offer value in predicting antibody half-life and clearance in primates.


Assuntos
Antígenos de Histocompatibilidade Classe I/genética , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Imunoglobulina G/administração & dosagem , Receptores Fc/genética , Vírus Sinciciais Respiratórios/imunologia , Animais , Ensaios Clínicos como Assunto , Feminino , Meia-Vida , Haplorrinos , Heterozigoto , Homozigoto , Humanos , Imunoglobulinas Intravenosas/administração & dosagem , Taxa de Depuração Metabólica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Valor Preditivo dos Testes , Albumina Sérica/administração & dosagem , Proteínas Virais de Fusão/imunologia
10.
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
11.
MAbs ; 2(5): 519-27, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20716959

RESUMO

Covalently-linked glycans on proteins have many functional roles, some of which are still not completely understood. Antibodies have a very specific glycan modification in the Fc region that is required for mediating immune effector functions. These Fc glycans are typically highly heterogeneous in structure, and this heterogeneity is influenced by many factors, such as type of cellular host and rate of Ab secretion. Glycan heterogeneity can affect the Fc-dependent activities of antibodies. It has been shown recently that increased Fc sialylation can result in decreased binding to immobilized antigens and some Fcγ receptors, as well as decreased antibody-dependent cell-mediated cytotoxicity (ADCC) activity. In contrast, increased Fc sialylation enhances the anti-inflammatory activity of antibodies. To produce antibodies with increased effector functions, we developed host cell lines that would limit the degree of sialylation of recombinantly-expressed antibodies. Towards this end, the catalytic domain of the Arthrobacter ureafaciens sialidase (sialidase A) was engineered for secreted expression in mammalian cell lines. Expression of this sialidase A gene in mammalian cells resulted in secreted expression of soluble enzyme that was capable of removing sialic acid from antibodies secreted into the medium. Purified antibodies secreted from these cells were found to possess very low levels of sialylation compared with the same antibodies purified from unmodified host cells. The low sialylated antibodies exhibited similar binding affinity to soluble antigens, improved ADCC activity, and they possessed pharmacokinetic properties comparable to their more sialylated counterparts. Further, it was observed that the amount of sialidase A expressed was sufficient to thoroughly remove sialic acid from Abs made in high-producing cell lines. Thus, engineering host cells to express sialidase A enzyme can be used to produce recombinant antibodies with very low levels of sialylation.


Assuntos
Anticorpos/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neuraminidase/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/genética , Anticorpos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Arthrobacter/enzimologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Glicosilação , Células HEK293 , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Neuraminidase/genética , Plasmídeos/genética , Polissacarídeos/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Transfecção
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.
Biochem Biophys Res Commun ; 341(3): 797-803, 2006 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-16442075

RESUMO

IgG antibodies (Abs) and fragments of IgG Abs are becoming major biotherapeutics to treat an assortment of human diseases. Commonly prepared fragments of IgGs include Fc, Fab, and F(ab')2 fragments, all of which can be made using the sulfhydryl protease papain, although prolonged digestion times and/or excessive amounts of papain typically result in further cleavage of the Fc domain into smaller fragments. During our attempts to use papain to isolate Fc fragments from different IgG monoclonal Abs, it was observed that prior removal of Fc glycans resulted in a faster rate of papain-mediated degradation of the Fc domain. Subsequent time-course experiments comparing glycosylated and deglycosylated versions of IgG antibodies showed that the majority of molecules in a deglycosylated IgG sample were converted into Fab, Fc, and smaller Fc fragments in less than one hour, whereas the original glycosylated IgG required more than two hours to convert into a comparable amount of Fab and Fc fragments. Furthermore, whereas papain digestion converted almost all of a deglycosylated Fc fragment into smaller fragments of approximately 10 and approximately 12 kDa within 4 h, more than 40% of a glycosylated Fc fragment remained intact even after 24 h of digestion. These results indicate that the presence of CH(2) domain glycans in either IgGs or purified Fc fragments increases resistance to papain digestion. Increased sensitivity of non-glycosylated Fc domains to papain is consistent with the Fc domains lacking a defined structure, as exemplified by their inability to bind Fcgamma receptors, since misfolded proteins are often degraded by proteases because of increased accessibility of their proteolytic cleavage sites. Based on these observations it is possible to use papain sensitivity as a means of assessing proper Fc structure of IgG molecules.


Assuntos
Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/metabolismo , Papaína/metabolismo , Animais , Configuração de Carboidratos , Glicosilação , Humanos , Camundongos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA