Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
MAbs ; 16(1): 2379560, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39028186

RESUMO

The self-association of therapeutic antibodies can result in elevated viscosity and create problems in manufacturing and formulation, as well as limit delivery by subcutaneous injection. The high concentration viscosity of some antibodies has been reduced by variable domain mutations or by the addition of formulation excipients. In contrast, the impact of Fc mutations on antibody viscosity has been minimally explored. Here, we studied the effect of a panel of common and clinically validated Fc mutations on the viscosity of two closely related humanized IgG1, κ antibodies, omalizumab (anti-IgE) and trastuzumab (anti-HER2). Data presented here suggest that both Fab-Fab and Fab-Fc interactions contribute to the high viscosity of omalizumab, in a four-contact model of self-association. Most strikingly, the high viscosity of omalizumab (176 cP) was reduced 10.7- and 2.2-fold by Fc modifications for half-life extension (M252Y:S254T:T256E) and aglycosylation (N297G), respectively. Related single mutations (S254T and T256E) each reduced the viscosity of omalizumab by ~6-fold. An alternative half-life extension Fc mutant (M428L:N434S) had the opposite effect in increasing the viscosity of omalizumab by 1.5-fold. The low viscosity of trastuzumab (8.6 cP) was unchanged or increased by ≤2-fold by the different Fc variants. Molecular dynamics simulations provided mechanistic insight into the impact of Fc mutations in modulating electrostatic and hydrophobic surface properties as well as conformational stability of the Fc. This study demonstrates that high viscosity of some IgG1 antibodies can be mitigated by Fc mutations, and thereby offers an additional tool to help design future antibody therapeutics potentially suitable for subcutaneous delivery.


Assuntos
Fragmentos Fc das Imunoglobulinas , Imunoglobulina G , Mutação , Omalizumab , Trastuzumab , Humanos , Trastuzumab/química , Viscosidade , Omalizumab/química , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/genética , Imunoglobulina G/genética , Imunoglobulina G/química , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética
2.
Biomed Pharmacother ; 169: 115848, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37976893

RESUMO

Analytical and functional comparison is key for substantiating the level of convergence (essential sameness) or divergence between versions or variants of a given biological medicine. Accordingly, an overlapping biological activity between products meant to be equal probably reflects a highly similar structure and anticipates a comparable pharmacodynamic behavior. We developed an orthogonal approach to compare the human IgE binding features of different lots and versions of Xolair® (omalizumab), an anti-human IgE monoclonal antibody. The IgE binding affinity and kinetics were measured by surface plasmon resonance. Ability to prevent mast cell activity was assessed in vitro and in vivo in mast cell-based models. The variability of monoclonal antibodies with identical amino acid sequences produced either in Chinese hamster ovarian cells or in human HEK293 cells, was compared. Monoclonal antibodies from the two sources exhibited slightly different human IgE binding and neutralizing features. A known variant exhibiting a three amino acid replacement in the Fab region had lower IgE binding affinity than the original omalizumab. The lower binding affinity translated into reduced IgE neutralizing capacity and, in turn, a difference in the ability to prevent mast cell activation in vitro and in vivo. The proposed set of analytical and functional assays was sensitive enough to detect Fab-linked differences between anti-IgE antibody versions exhibiting an identical aminoacid sequence. In addition to add value to the comparative assessment of biosimilar candidates bearing omalizumab, these methods can aid pre-assessments of new anti-IgE agents that aim to improve therapeutic performance.


Assuntos
Medicamentos Biossimilares , Omalizumab , Humanos , Omalizumab/farmacologia , Omalizumab/química , Omalizumab/metabolismo , Anticorpos Monoclonais Humanizados/farmacologia , Células HEK293 , Imunoglobulina E , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Imunossupressores
3.
J Pharm Biomed Anal ; 209: 114483, 2022 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-34864596

RESUMO

Omalizumab, a glycoprotein based biotherapeutics, is one of the most frequently used targeted antibody biopharmaceutical to reduce asthma exacerbations, improve lung function and reduce oral corticosteroid use. The effector function and clearance time of such glycoprotein drugs is affected by their N-glycosylation, that defines the required administration frequency to improve the quality of life in appropriately selected patients. Therefore, the glycosylation of biologics is an important critical quality attribute (CQA). The profile of asparagine linked carbohydrates is greatly dependent on the manufacturing process. Even a small deviation may have a major effect on the structure and therefore the function of the biotherapeutic product. For this reason, comprehensive N-glycosylation analysis is of high importance during production and release. Capillary electrophoresis (CE) is one of the frequently used tools to characterize protein therapeutics and utilized by the biopharmaceutical industry for protein and glycan level analysis, which are key parts both for drug development and quality control. To reveal important structure - function relationships, characterization of omalizumab is presented using capillary SDS gel electrophoresis with UV detection at the protein level and capillary gel electrophoresis with laser induced fluorescent detection at the N-linked carbohydrate level. This latter technique was also used for oligosaccharide sequencing for glycan structure validation. The results suggested no ADCC function - structure relationship due to the mostly core fucosylated biantennary glycans found. However, the presence of the high mannose structures probably affects the clearance rate of the drug.


Assuntos
Antiasmáticos , Omalizumab , Antiasmáticos/química , Glicosilação , Manose , Omalizumab/química , Polissacarídeos
4.
Front Immunol ; 11: 596908, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329588

RESUMO

The critical role of IgE in allergic diseases is well-documented and clinically proven. Omalizumab, a humanized anti-IgE antibody, was the first approved antibody for the treatment of allergic diseases. Nevertheless, omalizumab still has some limitations, such as product instability and dosage restriction in clinical application. In this study, we attempted to develop an omalizumab biobetter antibody with the potential to overcome its limitations. We removed two aspartic acid isomerization hotspots in CDRs of omalizumab to improve antibody candidate's stability. Meanwhile, several murine amino acids in the framework region of omalizumab were replaced with human source to reduce the potential immunogenicity. Yeast display technology was then applied to screen antibody candidates with high binding affinity to IgE. Moreover, YTE mutation in Fc fragment was introduced into the candidates for extending their serum half-life. A lead candidate, AB1904Am15, was screened out, which showed desired biophysical properties and improved stability, high binding affinity and elevated potency in vitro, prolonged half-life in human FcRn transgenic mouse, and enhanced in vivo efficacy in cynomolgus monkey asthma model. Overall, our study developed a biobetter antibody of omalizumab, AB1904Am15, which has the potential to show improved clinical benefit in the treatment of allergic diseases.


Assuntos
Antialérgicos/farmacologia , Antialérgicos/uso terapêutico , Anticorpos Anti-Idiotípicos/farmacologia , Anticorpos Anti-Idiotípicos/uso terapêutico , Hipersensibilidade/tratamento farmacológico , Omalizumab/farmacologia , Omalizumab/uso terapêutico , Antialérgicos/química , Anticorpos Anti-Idiotípicos/química , Afinidade de Anticorpos/imunologia , Fenômenos Biofísicos , Cromatografia Líquida , Monitoramento de Medicamentos , Estabilidade de Medicamentos , Citometria de Fluxo , Humanos , Hipersensibilidade/diagnóstico , Hipersensibilidade/imunologia , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Omalizumab/química , Ligação Proteica , Espectrometria de Massas em Tandem , Resultado do Tratamento
5.
Anal Chem ; 92(5): 4023-4028, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32023038

RESUMO

The temperature-dependent migration of molecular weight protein size standards and several biotherapeutic proteins were studied in sodium dodecyl sulfate capillary gel electrophoresis (SDS-CGE) in the interval from 15 to 60 °C using borate cross-linked dextran sieving matrix. Arrhenius plots were generated to calculate the respective activation energy values for the various solute molecules. SDS-CGE analysis of the biotherapeutic protein test mixture revealed no correlation between the activation energy requirement of the different species and their molecular weights, emphasizing the importance of separation temperature optimization to obtain high resolution between the solute molecules of interest. In contrast, the molecular weight protein size ladder ranging from 10 to 225 kDa, built from the same polypeptide blocks with no post-translational and other modifications, showed predictable activation energy requirement. The electrophoretic mobility of the SDS-protein complexes was found to be the function of the reciprocal sixth root of the molecular weight (Mw-1/6), implying cylindrical conformation.


Assuntos
Produtos Biológicos/química , Eletroforese em Gel de Poliacrilamida/métodos , Eletroforese Capilar , Peso Molecular , Omalizumab/química , Anticorpos de Cadeia Única/química , Temperatura
6.
Handb Exp Pharmacol ; 261: 131-151, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32076895

RESUMO

The management of atopic diseases such as severe asthma, severe atopic dermatitis, and severe food allergy in childhood is challenging. In particular, there are safety concerns regarding the use of high-dose corticosteroids. The recent development of biologicals and their approval for the treatment of children offer a new, very promising, and more personalized therapy option. Omalizumab, mepolizumab, and dupilumab are currently approved as add-on treatments of severe asthma in children and have been shown to be effective in improvement of asthma control and reduction of exacerbations. Dupilumab is the only biological approved for the treatment of atopic dermatitis in adolescents so far. It has been demonstrated to significantly improve symptoms of atopic dermatitis.However, safety data for biologicals used in atopic diseases in children and adolescents are still very limited. Biologicals are generally considered to be safe in adults. These data are often extrapolated to children. Additionally, data for long-term use are lacking. Thus, the safety profiles of those biologicals cannot yet be conclusively assessed.


Assuntos
Asma , Produtos Biológicos , Dermatite Atópica , Adolescente , Corticosteroides/química , Corticosteroides/genética , Corticosteroides/metabolismo , Adulto , Criança , Humanos , Omalizumab/química
7.
Pharm Res ; 37(2): 29, 2020 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-31915934

RESUMO

PURPOSE: To speed up the drug development process in the biopharmaceutical industry, high throughput methods are indispensable for assessing drug candidates and potential lead formulations, in particular during late stages of discovery and early phases of development. This study aimed to establish a bio-layer-interferometry based high throughput assay for assessing formulation dependent mAb self-interaction (SI-BLI) and to compare the results with kD values obtained by dynamic light scattering (DLS). METHODS: Self-interaction of proprietary and commercially available mAbs was analyzed by SI-BLI and dynamic light scattering (DLS). RESULTS: We found significant correlations of the SI-BLI results and kD-values obtained by DLS for both, different mAbs in one platform formulation and for mAbs formulated in several buffer compositions. In total, we assessed self-interaction propensity of different mAbs in 58 formulations and found significant Pearson correlation (p < 0.05) between kD and results of SI-BLI. CONCLUSIONS: The SI-BLI results correlate with kD and enable fast ranking of both different drug candidates and potential lead formulations. Thus, SI-BLI might decrease the risk to lose potent mAb candidates during transition from discovery to development, and help to accelerate the development of high concentration liquid formulations.


Assuntos
Adalimumab/química , Omalizumab/química , Composição de Medicamentos , Ensaios de Triagem em Larga Escala , Humanos , Interferometria/métodos , Cinética , Ligação Proteica , Conformação Proteica , Multimerização Proteica
8.
Nat Commun ; 11(1): 165, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31913280

RESUMO

Targeting of immunoglobulin E (IgE) represents an interesting approach for the treatment of allergic disorders. A high-affinity monoclonal anti-IgE antibody, ligelizumab, has recently been developed to overcome some of the limitations associated with the clinical use of the therapeutic anti-IgE antibody, omalizumab. Here, we determine the molecular binding profile and functional modes-of-action of ligelizumab. We solve the crystal structure of ligelizumab bound to IgE, and report epitope differences between ligelizumab and omalizumab that contribute to their qualitatively distinct IgE-receptor inhibition profiles. While ligelizumab shows superior inhibition of IgE binding to FcεRI, basophil activation, IgE production by B cells and passive systemic anaphylaxis in an in vivo mouse model, ligelizumab is less potent in inhibiting IgE:CD23 interactions than omalizumab. Our data thus provide a structural and mechanistic foundation for understanding the efficient suppression of FcεRI-dependent allergic reactions by ligelizumab in vitro as well as in vivo.


Assuntos
Antialérgicos/administração & dosagem , Anticorpos Anti-Idiotípicos/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Hipersensibilidade/tratamento farmacológico , Omalizumab/administração & dosagem , Animais , Antialérgicos/química , Anticorpos Anti-Idiotípicos/química , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Basófilos/efeitos dos fármacos , Basófilos/imunologia , Humanos , Hipersensibilidade/imunologia , Imunoglobulina E/química , Imunoglobulina E/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Omalizumab/química , Receptores de IgE/imunologia
9.
J Allergy Clin Immunol ; 144(2): 514-523.e5, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30995457

RESUMO

BACKGROUND: Variable heavy chain (VH) family frameworks (FWRs) have been reported to affect antibody receptor and superantigen binding; however, such effects in IgE remain largely unknown. Given that VH family biases have been previously reported in IgE of certain allergies, there is a need to investigate this phenomenon for biotechnological and therapeutic purposes. OBJECTIVE: We sought to investigate the effects of VH families on IgE interaction with FcεRIα, anti-IgE omalizumab, antigen, and superantigen protein A (spA) by using the pertuzumab and trastuzumab IgE models. METHODS: Pertuzumab VH1-VH7 family variants of IgE with the same complementarity-determining regions were investigated with regard to their binding interactions to FcεRIα, Her2, omalizumab, and spA. Notable FcεRIα-IgE observations were cross-checked against appropriate trastuzumab IgE VH variants. Computational structural modeling and simulations were also performed for insight into the mechanism of interactions with various VH FWRs. RESULTS: The pertuzumab VH5 IgE variant, but not the trastuzumab VH5 IgE, was found to interact with FcεRIα significantly longer than the respective VH family variants within each model antibody. No significant differences in interaction were found between IgE and omalizumab for the pertuzumab VH variants. Although trastuzumab VH3 interacted with spA, none of our pertuzumab VH variants, including VH3, associated with spA. CONCLUSION: We found unexpected varying allosteric communications caused by the VH family FWRs to the FcεRIα-, Her2-, and spA-binding regions of pertuzumab IgE, with implications for use of IgE/anti-IgE therapeutics to treat allergy and IgE therapeutics in allergo-oncology.


Assuntos
Antígenos de Bactérias/química , Imunoglobulina E/química , Cadeias Pesadas de Imunoglobulinas/química , Região Variável de Imunoglobulina/imunologia , Receptores de IgE/química , Superantígenos/química , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/imunologia , Antígenos de Bactérias/imunologia , Humanos , Hipersensibilidade/imunologia , Hipersensibilidade/terapia , Imunoglobulina E/imunologia , Imunoglobulina E/uso terapêutico , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/uso terapêutico , Imunoterapia , Omalizumab/química , Omalizumab/imunologia , Receptores de IgE/imunologia , Superantígenos/imunologia , Trastuzumab/química , Trastuzumab/imunologia
11.
Anal Chem ; 89(15): 7818-7822, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28707471

RESUMO

Therapeutic antibodies are prone to degradation via a variety of pathways during each stage of the manufacturing process. Hence, a low-cost, rapid, and broadly applicable tool that is able to identify when and how antibodies degrade would be highly desirable to control the quality of therapeutic antibody products. With this goal in mind, we have developed signature-based sensing system to discriminate differently degraded therapeutic antibodies. The use of arrays consisting of conjugates between nanographene oxide and fluorophore-modified single-stranded DNAs under acidic pH conditions generated unique fluorescence signatures for each state of the antibodies. Multivariate analyses of the thus obtained signatures allowed identifying (i) common features of native, denatured, and visibly aggregated antibodies, (ii) complicated degradation pathways of therapeutic omalizumab upon time-course heat-treatment, and (iii) the individual compositions of differently degraded omalizumab mixtures. As the signature-based sensing has the potential to identify a broad range of degraded antibodies formed by different kinds of realistic stress types, this system may serve as the basis for high-throughput assays for the screening of antibody manufacturing processes.


Assuntos
Anticorpos Monoclonais/metabolismo , DNA de Cadeia Simples/química , Anticorpos Monoclonais/química , Análise Discriminante , Corantes Fluorescentes/química , Grafite/química , Concentração de Íons de Hidrogênio , Análise em Microsséries , Omalizumab/química , Omalizumab/metabolismo , Agregados Proteicos , Desnaturação Proteica , Espectrometria de Fluorescência , Temperatura
12.
J Immunol Methods ; 449: 28-36, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28647457

RESUMO

Targeting plasma IgE by therapeutic mABs like Omalizumab (Xolair®) is current clinical practice for severe allergic conditions or other IgE related diseases like chronic urticaria. As an alternative to soluble IgE targeting, IgE supply can be lowered by targeting the Extracellular Membrane Proximal Domain (EMPD) of the IgE B cell receptor (BCR) present on IgE switched B cells. This ultimately leads to apoptosis of these cells upon IgE BCR crosslinking. Since tools to selectively assess the efficacy of IgE BCR crosslinking by IgE targeting antibodies are limited, a readily quantifiable cell model was developed that allows to specifically address IgE BCR crosslinking activity in vitro. The new cell model allowed for a direct quantitative comparison of anti-EMPD IgE therapeutic prototype antibody 47H4 with anti-IgE(Ce3) directed therapeutic antibody Omalizumab and with a newly selected anti-human EMPD IgE monoclonal antibody, designated mAB 15cl12. Furthermore, a complementing mouse model was developed that allows for in vivo validation of antibodies addressing human EMPD IgE. It carries a targetable humanized EMPD IgE sequence that has been introduced by seamless genomic replacement of the endogenous EMPD encoding sequence. The model allowed to directly compare IgE lowering activity of two anti-human EMPD IgE therapeutic antibodies in vivo. Our tools provide the means for quantitative assessment of IgE BCR crosslinking activity which is increasingly gaining attention with respect to forthcoming second generation anti-IgE clinical candidates such as Ligelizumab or other clinical candidates featuring additional effector functions such as IgE BCR crosslinking activity.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Imunoglobulina E/química , Imunoglobulina E/imunologia , Receptores de Antígenos de Linfócitos B/química , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Antialérgicos/química , Antialérgicos/metabolismo , Anticorpos Anti-Idiotípicos/química , Anticorpos Anti-Idiotípicos/metabolismo , Reagentes de Ligações Cruzadas , Humanos , Imunoglobulina E/biossíntese , Imunoglobulina E/metabolismo , Camundongos , Omalizumab/química , Omalizumab/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo
13.
J Biol Chem ; 292(24): 9975-9987, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28438838

RESUMO

Immunoglobulin E and its interactions with receptors FcϵRI and CD23 play a central role in allergic disease. Omalizumab, a clinically approved therapeutic antibody, inhibits the interaction between IgE and FcϵRI, preventing mast cell and basophil activation, and blocks IgE binding to CD23 on B cells and antigen-presenting cells. We solved the crystal structure of the complex between an omalizumab-derived Fab and IgE-Fc, with one Fab bound to each Cϵ3 domain. Free IgE-Fc adopts an acutely bent structure, but in the complex it is only partially bent, with large-scale conformational changes in the Cϵ3 domains that inhibit the interaction with FcϵRI. CD23 binding is inhibited sterically due to overlapping binding sites on each Cϵ3 domain. Studies of omalizumab Fab binding in solution demonstrate the allosteric basis for FcϵRI inhibition and, together with the structure, reveal how omalizumab may accelerate dissociation of receptor-bound IgE from FcϵRI, exploiting the intrinsic flexibility and allosteric potential of IgE.


Assuntos
Antiasmáticos/farmacologia , Imunoglobulina E/metabolismo , Modelos Moleculares , Omalizumab/farmacologia , Receptores de IgE/antagonistas & inibidores , Sítio Alostérico , Substituição de Aminoácidos , Cristalografia por Raios X , Transferência Ressonante de Energia de Fluorescência , Humanos , Imunoglobulina E/química , Imunoglobulina E/genética , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/metabolismo , Fragmentos Fab das Imunoglobulinas/farmacologia , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Fragmentos Fc das Imunoglobulinas/farmacologia , Omalizumab/química , Omalizumab/genética , Omalizumab/metabolismo , Maleabilidade , Mutação Puntual , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Redobramento de Proteína , Receptores de IgE/química , Receptores de IgE/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Solubilidade , Ressonância de Plasmônio de Superfície
14.
Biochem Biophys Res Commun ; 485(2): 284-289, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28223218

RESUMO

Acute lung injury (ALI) is an early pathophysiologic change in acute respiratory distress syndrome and its management can be challenging. Omalizumab (Xolair™) is a recombinant DNA-derived, humanized antibody. OMZ-SPT is a polypeptide on the heavy chain of omalizumab monoclonal antibody. Here, we found that intramuscular administration of OMZ-SPT significantly improved survival and attenuated lung inflammation in female C57BL/6 mice suffering from lipopolysaccharide (LPS)-induced ALI. We also demonstrated that OMZ-SPT can inhibit expression of the inflammatory cytokines tumor necrosis factor-α, interleukin-1ß and interleukin-6 by ELISA in mice suffering from LPS-induced ALI and a mouse macrophage line (RAW264.7 cells). In addition, we showed that OMZ-SPT inhibited LPS-induced activation of nuclear factor-kappa B (NF-κB) signaling and total expression of NF-κB by western blotting. These data suggest that OMZ-SPT could be a novel therapeutic choice for ALI.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Anti-Inflamatórios/uso terapêutico , Pulmão/efeitos dos fármacos , NF-kappa B/imunologia , Omalizumab/uso terapêutico , Pneumonia/tratamento farmacológico , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Anti-Inflamatórios/química , Feminino , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/patologia , Camundongos Endogâmicos C57BL , Omalizumab/química , Peptídeos/química , Peptídeos/uso terapêutico , Pneumonia/imunologia , Pneumonia/patologia , Transdução de Sinais/efeitos dos fármacos
15.
Nat Commun ; 7: 11610, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27194387

RESUMO

Omalizumab is a widely used therapeutic anti-IgE antibody. Here we report the crystal structure of the omalizumab-Fab in complex with an IgE-Fc fragment. This structure reveals the mechanism of omalizumab-mediated inhibition of IgE interactions with both high- and low-affinity IgE receptors, and explains why omalizumab selectively binds free IgE. The structure of the complex also provides mechanistic insight into a class of disruptive IgE inhibitors that accelerate the dissociation of the high-affinity IgE receptor from IgE. We use this structural data to generate a mutant IgE-Fc fragment that is resistant to omalizumab binding. Treatment with this omalizumab-resistant IgE-Fc fragment, in combination with omalizumab, promotes the exchange of cell-bound full-length IgE with omalizumab-resistant IgE-Fc fragments on human basophils. This combination treatment also blocks basophil activation more efficiently than either agent alone, providing a novel approach to probe regulatory mechanisms underlying IgE hypersensitivity with implications for therapeutic interventions.


Assuntos
Antialérgicos/farmacologia , Imunoglobulina E/efeitos dos fármacos , Omalizumab/farmacologia , Receptores de IgE/antagonistas & inibidores , Animais , Antialérgicos/química , Basófilos/efeitos dos fármacos , Linhagem Celular , Sinergismo Farmacológico , Humanos , Imunoglobulina E/química , Imunoglobulina E/genética , Mutação , Omalizumab/química , Conformação Proteica
16.
Sheng Wu Gong Cheng Xue Bao ; 32(4): 497-506, 2016 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-28853271

RESUMO

We compared the similarity of Omalizumab (Xolair; a humanized anti-immunoglobulin E monoclonal antibody) and it's biosimilar CMAB007. An in depth characterization of a candidate biosimilar was carried out using a systematic approach, the approach provides a set of routine tools that combine accurate intact mass measurement, peptide mapping, and released glycan profiling. CMAB007 and Omalizumab had the same primary structure and exhibited almost the same content of C-terminal lysine variants. The types of detected free oligosaccharides were very similar, such as sialylation, fucosylation and high mannose types. CMAB007 could be considered as a highly similar molecular to Omalizumab and expected to be the first humanized anti-immunoglobulin E monoclonal antibody drug in China.


Assuntos
Medicamentos Biossimilares/química , Omalizumab/química , Cromatografia Líquida , Glicosilação , Humanos , Manose/química , Espectrometria de Massas , Mapeamento de Peptídeos , Polissacarídeos/química
17.
J Pharm Sci ; 104(8): 2457-63, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26036204

RESUMO

The stabilization of antibodies in aqueous solution against physical stress remains a problematic issue for pharmaceutical applications. Recently, protein-polyelectrolyte complex (PPC) formation using poly(amino acids) was proposed to prepare antibody formulation in a salt-dissociable precipitated state without protein denaturation. Here, we investigated the stabilization effect of PPC of therapeutic antibodies with poly-l-glutamic acid on agitation and thermal stress as forms of mechanical and non-mechanical stress, respectively. The precipitated state of PPC prevented the inactivation and aggregation induced by agitation. Similar results were obtained using the suspension state of PPC, but the stabilizing effects were slightly inferior to those of the PPC precipitate. PPC precipitate and PPC suspension prevented heat-induced inactivation of the antibodies, but showed little effect on heat-induced aggregation. Thus, PPC is a new candidate as a simple storage method for antibodies in aqueous solution, as an alternative state for freeze-drying.


Assuntos
Anticorpos Monoclonais/química , Modelos Químicos , Preparações Farmacêuticas/química , Ácido Poliglutâmico/química , Conservantes Farmacêuticos/química , Adalimumab/química , Adalimumab/metabolismo , Animais , Antiasmáticos/química , Antiasmáticos/metabolismo , Anticorpos Monoclonais/metabolismo , Antirreumáticos/química , Antirreumáticos/metabolismo , Precipitação Química , Química Farmacêutica , Estabilidade de Medicamentos , Armazenamento de Medicamentos , Temperatura Alta/efeitos adversos , Humanos , Omalizumab/química , Omalizumab/metabolismo , Tamanho da Partícula , Preparações Farmacêuticas/metabolismo , Ácido Poliglutâmico/metabolismo , Conservantes Farmacêuticos/metabolismo , Agregados Proteicos , Estabilidade Proteica , Solubilidade , Estresse Mecânico , Suspensões
18.
Sci Rep ; 5: 11581, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-26113483

RESUMO

Omalizumab, an anti-IgE antibody, used to treat severe allergic asthma and chronic idiopathic urticaria, binds to IgE in blood or membrane-bound on B lymphocytes but not to IgE bound to its high (FcεRI) or low (CD23) affinity receptor. Mutagenesis studies indicate overlapping FcεRI and omalizumab-binding sites in the Cε3 domain, but crystallographic studies show FcεRI and CD23-binding sites that are far apart, so how can omalizumab block IgE from binding both receptors? We report a 2.42-Šomalizumab-Fab structure, a docked IgE-Fc/omalizumab-Fab structure consistent with available experimental data, and the free energy contributions of IgE residues to binding omalizumab, CD23, and FcεRI. These results provide a structural and physical basis as to why omalizumab cannot bind receptor-bound IgE and why omalizumab-bound IgE cannot bind to CD23/FcεRI. They reveal the key IgE residues and their roles in binding omalizumab, CD23, and FcεRI.


Assuntos
Anticorpos Anti-Idiotípicos/química , Imunoglobulina E/química , Omalizumab/química , Sequência de Aminoácidos , Antiasmáticos/imunologia , Antiasmáticos/metabolismo , Antiasmáticos/uso terapêutico , Anticorpos Anti-Idiotípicos/metabolismo , Anticorpos Anti-Idiotípicos/uso terapêutico , Asma/tratamento farmacológico , Asma/imunologia , Sítios de Ligação/genética , Cristalografia por Raios X , Humanos , Imunoglobulina E/metabolismo , Modelos Moleculares , Mutação , Omalizumab/metabolismo , Omalizumab/uso terapêutico , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de IgE/química , Receptores de IgE/metabolismo
19.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 4): 419-26, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25849503

RESUMO

Omalizumab is a humanized anti-IgE antibody that inhibits the binding of IgE to its receptors on mast cells and basophils, thus blocking the IgE-mediated release of inflammatory mediators from these cells. Omalizumab binds to the Fc domains of IgE in proximity to the binding site of the high-affinity IgE receptor FcℇRI, but the epitope and the mechanisms and conformations governing the recognition remain unknown. In order to elucidate the molecular mechanism of its anti-IgE activity, the aim was to analyse the interaction of omalizumab with human IgE. Therefore, IgE Fc Cℇ2-4 was recombinantly produced in mammalian HEK-293 cells. Functionality of the IgE Fc was proven by ELISA and mediator-release assays. Omalizumab IgG was cleaved with papain and the resulting Fab was purified by ion-exchange chromatography. The complex of IgE Fc with omalizumab was prepared by size-exclusion chromatography. However, crystals containing the complex were not obtained, suggesting that the process of crystallization favoured the dissociation of the two proteins. Instead, two structures of the omalizumab Fab with maximum resolutions of 1.9 and 3.0 Šwere obtained. The structures reveal the arrangement of the CDRs and the position of omalizumab residues known from prior functional studies to be involved in IgE binding. Thus, the structure of omalizumab provides the structural basis for understanding the function of omalizumab, allows optimization of the procedure for complex crystallization and poses questions about the conformational requirements for anti-IgE activity.


Assuntos
Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/metabolismo , Omalizumab/química , Omalizumab/metabolismo , Anticorpos Anti-Idiotípicos/química , Anticorpos Anti-Idiotípicos/metabolismo , Cristalização , Células HEK293 , Humanos , Imunoglobulina E/química , Imunoglobulina E/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores de IgE/química , Receptores de IgE/metabolismo
20.
J Pharm Sci ; 104(6): 1929-1937, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25807887

RESUMO

Development of high-concentration antibody formulations for subcutaneous administration remains challenging. Recently, a precipitation-redissolution method was proposed to prepare suspensions or precipitates of salt-dissociable protein-poly(amino acid) complexes. To elucidate the utility of this method for protein therapy, we investigated the feasibility of a precipitation-redissolution method using poly(amino acid) for high-concentration antibody formulation. Omalizumab and adalimumab formulations of 150 mg/mL could be prepared using poly-l-glutamic acid (polyE) from low-concentration stock solutions. Enzyme-linked immunosorbent assay, circular dichroism, and size-exclusion chromatography revealed that the formation of antibody-polyE complex and precipitation-redissolution process did not significantly affect the immunoreactivity or secondary structure of the antibodies. The precipitation-redissolution method was less time-consuming and more effective than lyophilization-redissolution, evaporation-redissolution, and ultrafiltration from the viewpoint of final yield. Scalability was confirmed from 400 µL to 1.0 L. The general toxicity and pharmacokinetic profiles of the antibody-polyE complex formulations were similar to those of conventional antibody formulations. These results suggested that the precipitation-redissolution method using poly(amino acid) has great potential as a concentration method for antibody formulation and medicinal use.


Assuntos
Adalimumab/química , Antiasmáticos/química , Anti-Inflamatórios/química , Omalizumab/química , Ácido Poliglutâmico/química , Adalimumab/sangue , Adalimumab/farmacologia , Animais , Antiasmáticos/sangue , Antiasmáticos/farmacologia , Anti-Inflamatórios/sangue , Anti-Inflamatórios/farmacologia , Precipitação Química , Humanos , Omalizumab/sangue , Omalizumab/farmacologia , Ácido Poliglutâmico/sangue , Ácido Poliglutâmico/farmacologia , Estrutura Secundária de Proteína , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA