Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Pharm Biomed Anal ; 251: 116452, 2024 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-39217700

RESUMO

Adeno-associated virus (AAV)-based gene therapy is experiencing a rapid growth in the field of medicine and holds great promise in combating a wide range of human diseases. For successful development of AAV-based products, comprehensive thermal stability studies are often required to establish storage conditions and shelf life. However, as a relatively new modality, limited studies have been reported to elucidate the chemical degradation pathways of AAV products under thermal stress conditions. In this study, we first presented an intriguing difference in charge profile shift between thermally stressed AAV8 and AAV1 capsids when analyzed by anion exchange chromatography. Subsequently, a novel and robust peptide mapping protocol was developed and applied to elucidate the underlying chemical degradation pathways of thermally stressed AAV8 and AAV1. Compared to the conventional therapeutic proteins, the unique structure of AAV capsids also led to some key differences in how modifications at specific sites may impact the overall charge properties. Finally, despite the high sequency identity, the analysis revealed that the opposite charge profile shifts between thermally stressed AAV8 and AAV1 could be mainly attributed to a single modification unique to each serotype.


Assuntos
Dependovirus , Mapeamento de Peptídeos , Dependovirus/genética , Dependovirus/química , Cromatografia por Troca Iônica/métodos , Mapeamento de Peptídeos/métodos , Capsídeo/química , Temperatura Alta , Humanos , Proteínas do Capsídeo/química , Vetores Genéticos/química , Terapia Genética/métodos , Cromatografia Líquida/métodos , Espectrometria de Massa com Cromatografia Líquida
2.
Cell Rep Med ; : 101747, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39326410

RESUMO

The clinical use of interleukin-2 (IL-2) for cancer immunotherapy is limited by severe toxicity. Emerging IL-2 therapies with reduced IL-2 receptor alpha (IL-2Rα) binding aim to mitigate toxicity and regulatory T cell (Treg) expansion but have had limited clinical success. Here, we show that IL-2Rα engagement is critical for the anti-tumor activity of systemic IL-2 therapy. A "non-α" IL-2 mutein induces systemic expansion of CD8+ T cells and natural killer (NK) cells over Tregs but exhibits limited anti-tumor efficacy. We develop a programmed cell death protein 1 (PD-1)-targeted, receptor-masked IL-2 immunocytokine, PD1-IL2Ra-IL2, which attenuates systemic IL-2 activity while maintaining the capacity to engage IL-2Rα on PD-1+ T cells. Mice treated with PD1-IL2Ra-IL2 show no systemic toxicities observed with unmasked IL-2 treatment yet achieve robust tumor growth control. Furthermore, PD1-IL2Ra-IL2 can be effectively combined with other T cell-mediated immunotherapies to enhance anti-tumor responses. These findings highlight the therapeutic potential of PD1-IL2Ra-IL2 as a targeted, receptor-masked, and "α-maintained" IL-2 therapy for cancer.

3.
MAbs ; 14(1): 2133674, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36224723

RESUMO

Therapeutic monoclonal antibodies (mAbs) have a propensity to host a large number of chemical and enzymatical modifications that need to be properly assessed for their potential impact on target binding. Traditional strategies of assessing the criticality of these attributes often involve a laborious and low-throughput variant enrichment step prior to binding affinity measurement. Here, we developed a novel competitive binding-based enrichment strategy followed by mass spectrometry analysis (namely, competitive binding-MS) to achieve high-throughput evaluation of potential critical quality attributes in therapeutic mAbs. Leveraging the differences in target binding capability under competitive binding conditions, the criticality of multiple mAb attributes can be simultaneously evaluated by quantitative mass spectrometry analysis. The utility of this new workflow was demonstrated in three mAb case studies, where different post-translational modifications occurring within the complementarity-determining regions were successfully interrogated for their impact on antigen binding. As this workflow does not require prior enrichment (e.g., by forced degradation or liquid chromatography fractionation) of the variants, it is particularly valuable during the mAb candidate developability assessment, where fast turn-around time is highly desired to assist candidate selection.Abbreviations: ACN: acetonitrile; ADCC: antibody-dependent cell-mediated cytotoxicity; AEX: anion exchange chromatography; bsAb: bispecific antibody; CDC: complement-dependent cytotoxicity; CDR: complementarity-determining region; CML: carboxymethylation; CQA: critical quality attribute; DDA: data-dependent acquisition; DMSO: dimethyl sulfoxide; DTT: dithiothreitol; FA: formic acid; Fab: Fragment antigen-binding; FcRn: neonatal Fc receptor; HC: heavy chain; HIC: hydrophobic interaction chromatography; IAA: iodoacetamide; IEX: ion exchange chromatography; LC: light chain; mAb monoclonal antibody; msAb: monospecific antibody; MS: mass spectrometry; PBS: phosphate-buffered saline; pI: isoelectric point; PTM: post-translational modification; SCX: strong cation exchange chromatography; SEC: size exclusion chromatography; SPR: surface plasmon resonance; XIC: extracted ion chromatography.


Assuntos
Anticorpos Monoclonais , Antineoplásicos Imunológicos , Acetonitrilas , Anticorpos Monoclonais/química , Ligação Competitiva , Cátions , Regiões Determinantes de Complementaridade/química , Dimetil Sulfóxido , Ditiotreitol , Iodoacetamida , Espectrometria de Massas/métodos , Fosfatos
4.
J Pharm Biomed Anal ; 189: 113481, 2020 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-32750536

RESUMO

To support adeno-associated virus (AAV)-based gene therapy development, characterization of the three capsid viral proteins (VP; VP1/VP2/VP3) from recombinant AAV can offer insights on capsid identity, heterogeneity, and product and process consistency. Intact protein mass analysis is a rapid, reliable, and sensitive method to confirm AAV serotypes based on accurate mass measurement of the constituent capsid proteins. Compared to commonly applied reversed-phase liquid chromatography (RPLC) methods, we demonstrated that, using a wide-pore amide-bonded column, hydrophilic interaction chromatography (HILIC) could achieve improved separation of VPs from a variety of AAV serotypes using a generic method prior to MS detection. Moreover, HILIC-based separation was shown to be particularly sensitive in detecting capsid protein variants resulting from different post-translational modifications (PTMs) (e.g. phosphorylation and oxidation) and protein backbone clippings, making it ideally suited for capsid heterogeneity characterization. To overcome the challenges associated with low protein concentrations of AAV samples, as well as the trifluoroacetic acid (TFA)-induced ion suppression during HILIC-MS analysis, different strategies were implemented to improve method sensitivity, including increasing the HILIC column loading and the application of a desolvation gas modification device. Finally, we demonstrated that this integrated HILIC-FLR-MS method can be generically applied to characterize a variety of AAV serotype samples at low concentrations without any sample treatment to achieve unambiguous serotype identification, stoichiometry assessment, and PTM characterization.


Assuntos
Proteínas do Capsídeo , Dependovirus , Proteínas do Capsídeo/genética , Cromatografia de Fase Reversa , Dependovirus/genética , Interações Hidrofóbicas e Hidrofílicas , Espectrometria de Massas
5.
Anal Chem ; 91(4): 3156-3162, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30682238

RESUMO

LC-MS based analysis of protein biopharmaceuticals could benefit from improved data quality, which can subsequently lead to improved drug characterization with higher confidence and less ambiguity. In this study, we created a simple device to modify the desolvation gas on a Q-Exactive mass spectrometer and to demonstrate the utility in improving both peptide mapping analysis and intact mass analysis, the two most routinely and widely applied LC-MS techniques in protein biopharmaceutical characterization. By modifying the desolvation gas with acid vapor from propionic acid (PA) and isopropanol (IPA), the ion suppression effects from trifluoroacetic acid (TFA) in a typical peptide mapping method can be effectively mitigated, thus leading to improved MS sensitivity. By modifying the desolvation gas with base vapor from triethylamine (TEA), the charge reduction effect can be achieved and utilized to improve the spectral quality from intact mass analysis of protein biopharmaceuticals. The approach and device described in this work suggests a low-cost and practical solution to improve the LC-MS characterization of protein biopharmaceuticals, which has the potential to be widely implemented in biopharmaceutical analytical laboratories.


Assuntos
Anticorpos Monoclonais/análise , Produtos Biológicos/análise , Cromatografia Líquida de Alta Pressão , Gases/química , Humanos , Espectrometria de Massas em Tandem
6.
Biochemistry ; 54(34): 5322-8, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26266572

RESUMO

The type 2 L-serine dehydratase from Legionella pneumophila (lpLSD) contains a [4Fe-4S](2+) cluster that acts as a Lewis acid to extract the hydroxyl group of L-serine during the dehydration reaction. Surprisingly, the crystal structure shows that all four of the iron atoms in the cluster are coordinated with protein cysteinyl residues and that the cluster is buried and not exposed to solvent. If the crystal structure of lpLSD accurately reflects the structure in solution, then substantial rearrangement at the active site is necessary for the substrate to enter. Furthermore, repair of the oxidized protein when the cluster has degraded would presumably entail exposure of the buried cysteine ligands. Thus, the conformation required for the substrate to enter may be similar to those required for a new cluster to enter the active site. To address this, hydrogen-deuterium exchange combined with mass spectrometry (HDX MS) was used to probe the conformational changes that occur upon oxidative degradation of the Fe-S cluster. The regions that show the most significant differential HDX are adjacent to the cluster location in the holoenzyme or connect regions that are adjacent to the cluster. The observed decrease in flexibility upon cluster binding provides direct evidence that the "tail-in-mouth" conformation observed in the crystal structure also occurs in solution and that the C-terminal peptide is coordinated to the [4Fe-4S] cluster in a precatalytic conformation. This observation is consistent with the requirement of an activation step prior to catalysis and the unusually high level of resistance to oxygen-induced cluster degradation. Furthermore, peptide mapping of the apo form under nonreducing conditions revealed the formation of disulfide bonds between C396 and C485 and possibly between C343 and C385. These observations provide a picture of how the cluster loci are stabilized and poised to receive the cluster in the apo form and the requirement for a reduction step during cluster formation.


Assuntos
Proteínas de Bactérias/química , L-Serina Desidratase/química , Legionella pneumophila/enzimologia , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Medição da Troca de Deutério , Holoenzimas/química , Holoenzimas/genética , Holoenzimas/metabolismo , Proteínas Ferro-Enxofre/química , L-Serina Desidratase/genética , L-Serina Desidratase/metabolismo , Legionella pneumophila/genética , Espectrometria de Massas , Modelos Moleculares , Dados de Sequência Molecular , Mapeamento de Peptídeos , Ligação Proteica , Conformação Proteica
7.
J Am Soc Mass Spectrom ; 25(12): 2084-92, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25267085

RESUMO

Epitope mapping is an important tool for the development of monoclonal antibodies, mAbs, as therapeutic drugs. Recently, a class of therapeutic mAb alternatives, adnectins, has been developed as targeted biologics. They are derived from the 10th type III domain of human fibronectin ((10)Fn3). A common approach to map the epitope binding of these therapeutic proteins to their binding partners is X-ray crystallography. Although the crystal structure is known for Adnectin 1 binding to human epidermal growth factor receptor (EGFR), we seek to determine complementary binding in solution and to test the efficacy of footprinting for this purpose. As a relatively new tool in structural biology and complementary to X-ray crystallography, protein footprinting coupled with mass spectrometry is promising for protein-protein interaction studies. We report here the use of fast photochemical oxidation of proteins (FPOP) coupled with MS to map the epitope of EGFR-Adnectin 1 at both the peptide and amino-acid residue levels. The data correlate well with the previously determined epitopes from the crystal structure and are consistent with HDX MS data, which are presented in an accompanying paper. The FPOP-determined binding interface involves various amino-acid and peptide regions near the N terminus of EGFR. The outcome adds credibility to oxidative labeling by FPOP for epitope mapping and motivates more applications in the therapeutic protein area as a stand-alone method or in conjunction with X-ray crystallography, NMR, site-directed mutagenesis, and other orthogonal methods.


Assuntos
Mapeamento de Epitopos/métodos , Receptores ErbB/química , Receptores ErbB/metabolismo , Fibronectinas/química , Fibronectinas/metabolismo , Epitopos , Humanos , Modelos Moleculares , Oxirredução , Ligação Proteica , Estrutura Terciária de Proteína
8.
Biochemistry ; 53(19): 3248-60, 2014 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-24786965

RESUMO

The Sec pathway mediates translocation of protein across the inner membrane of bacteria. SecA is a motor protein that drives translocation of preprotein through the SecYEG channel. SecA reversibly dimerizes under physiological conditions, but different dimer interfaces have been observed in SecA crystal structures. Here, we have used biophysical approaches to address the nature of the SecA dimer that exists in solution. We have taken advantage of the extreme salt sensitivity of SecA dimerization to compare the rates of hydrogen-deuterium exchange of the monomer and dimer and have analyzed the effects of single-alanine substitutions on dimerization affinity. Our results support the antiparallel dimer arrangement observed in one of the crystal structures of Bacillus subtilis SecA. Additional residues lying within the preprotein binding domain and the C-terminus are also protected from exchange upon dimerization, indicating linkage to a conformational transition of the preprotein binding domain from an open to a closed state. In agreement with this interpretation, normal mode analysis demonstrates that the SecA dimer interface influences the global dynamics of SecA such that dimerization stabilizes the closed conformation.


Assuntos
Adenosina Trifosfatases/química , Proteínas de Bactérias/química , Escherichia coli/química , Proteínas de Membrana Transportadoras/química , Multimerização Proteica/fisiologia , Adenosina Trifosfatases/genética , Substituição de Aminoácidos , Bacillus subtilis/química , Bacillus subtilis/genética , Proteínas de Bactérias/genética , Cristalografia por Raios X , Escherichia coli/genética , Proteínas de Membrana Transportadoras/genética , Estrutura Quaternária de Proteína , Canais de Translocação SEC , Proteínas SecA
9.
Anal Chim Acta ; 644(1-2): 36-41, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19463559

RESUMO

A sensitive, rapid and simple method for analysis of protein was proposed by introducing an electric field-driven technique to the incubation process of immunoassay and immobilizing horseradish peroxidase (HRP) labeled antibody in a newly designed gel matrix to construct an immunosensor. With an electric field-driven technique, the incubation for immuno-recognition could be completed within 2 min. The immobilized HRP showed excellent direct electrochemistry in this new gel matrix, and the detection procedure was greatly simplified by directly monitoring the sensitive electrochemical signal of HRP upon the immunoreaction. Using alpha-fetoprotein as a model the linear detection range was from 0.02 to 2.0 ng mL(-1) with a detection limit down to 138 amol mL(-1). The immunosensor and the proposed detection method showed good specificity and acceptable stability and accuracy.


Assuntos
Técnicas Eletroquímicas/métodos , Imunoensaio/métodos , Proteínas/análise , Anticorpos Imobilizados/química , Anticorpos Imobilizados/imunologia , Biomarcadores Tumorais/análise , Eletrodos , Peroxidase do Rábano Silvestre/química , Peroxidase do Rábano Silvestre/metabolismo , Fatores de Tempo , alfa-Fetoproteínas/análise
10.
Anal Chem ; 80(15): 6072-7, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18593191

RESUMO

A fast, simple, sensitive, and low-cost method for electrochemical multianalyte immunoassay was developed by combining newly designed electric field-driven incubation with a screen-printed reagentless immunosensor array. The disposable array was prepared by immobilizing respectively horseradish peroxidase (HRP)-labeled antibodies modified gold nanoparticles in biopolymer/sol-gel modified electrodes to obtain direct electrochemical responses of HRP. Upon the formation of immunocomplexes, the responses decreased due to increasing spatial blocking and impedance. At a driving potential of 0.5 V, the incubation process could be accomplished within 2 min. Under optimal conditions, this method could simultaneously detect carbohydrate antigens 153, 125, and 199 and carcinoembryonic antigens ranging from 0.084 to 16, 0.11 to 13, and 0.16 to 15 U mL(-1) and 0.16 to 9.2 ng mL(-1) with a detection time of less than 5 min, and the detection limits corresponding to the signals of 3SD were 0.06, 0.03, and 0.10 U mL(-1) and 0.04 ng mL(-1), respectively. The disposable immunosensor array and simple detection system for fast measurement of panels of tumor markers show significant clinical value for application in cancer screening and provide great potential for convenient point-of-care testing and commercial application.


Assuntos
Eletroquímica/métodos , Imunoensaio/métodos , Proteínas/análise , Animais , Anticorpos , Biomarcadores/análise , Antígeno Carcinoembrionário/análise , Eletrodos , Glicoproteínas/análise , Ouro , Peroxidase do Rábano Silvestre , Humanos , Nanopartículas Metálicas , Sistemas Automatizados de Assistência Junto ao Leito
11.
Clin Chem ; 54(9): 1481-8, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18606631

RESUMO

BACKGROUND: A reagentless sensor array for simultaneous multianalyte testing (SMAT) may enable accurate diagnosis and be applicable for point-of-care testing. We developed a disposable reagentless immunosensor array for simple immunoassay of panels of tumor markers. METHODS: We carried out SMAT with a direct capture format, in which colloidal gold nanoparticles with bound horseradish peroxidase (HRP)-labeled antibodies were immobilized on screen-printed carbon electrodes with biopolymer/sol-gel to trap their corresponding antigens from sample solution. Upon formation of immunocomplex, the direct electrochemical signal of the HRP decreased owing to increasing spatial blocking, and the analytes could be simultaneously determined by monitoring the signal changes. RESULTS: The proposed reagentless immunosensor array allowed simultaneous detection of carcinoma antigen 153, carcinoma antigen 125, carbohydrate antigen 199, and carcinoembryonic antigen in clinical serum samples in the ranges of 0.4-140 kU/L, 0.5-330 kU/L, 0.8-190 kU/L, and 0.1-44 microg/L, respectively, with detection limits of 0.2 kU/L, 0.5 kU/L, 0.3 kU/L, and 0.1 microg/L corresponding to the signals 3 SD above the mean of a zero standard. The interassay imprecision of the arrays was <9.5%, and they were stable for 35 days. The positivity detection rate of panels of tumor markers was >95.5% for 95 cases of cancer-positive sera. CONCLUSIONS: The immunosensor array provides a SMAT with short analytical time, small sampling volume, no need for substrate, and, no between-electrode cross-talk. This method not only proved the capability of the array in point-of-care testing, but also allowed simultaneous testing of several tumor markers.


Assuntos
Biomarcadores Tumorais/sangue , Imunoensaio/métodos , Transição de Fase , Biopolímeros/química , Eletroquímica , Humanos , Indicadores e Reagentes , Microscopia Eletrônica , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA