Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Protein Sci ; 33(5): e4990, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38607241

RESUMEN

The antigen-binding sites in conventional antibodies are formed by hypervariable complementarity-determining regions (CDRs) from both heavy chains (HCs) and light chains (LCs). A deviation from this paradigm is found in a subset of bovine antibodies that bind antigens via an ultra-long CDR. The HCs bearing ultra-long CDRs pair with a restricted set of highly conserved LCs that convey stability to the antibody. Despite the importance of these LCs, their specific features remained unknown. Here, we show that the conserved bovine LC found in antibodies with ultra-long CDRs exhibits a distinct combination of favorable physicochemical properties such as good secretion from mammalian cells, strong dimerization, high stability, and resistance to aggregation. These physicochemical traits of the LCs arise from a combination of the specific sequences in the germline CDRs and a lambda LC framework. In addition to understanding the molecular architecture of antibodies with ultra-long CDRs, our findings reveal fundamental insights into LC characteristics that can guide the design of antibodies with improved properties.


Asunto(s)
Regiones Determinantes de Complementariedad , Cadenas Ligeras de Inmunoglobulina , Animales , Bovinos , Cadenas Ligeras de Inmunoglobulina/genética , Anticuerpos , Dimerización , Fenotipo , Mamíferos
2.
Mol Pharm ; 20(6): 2951-2965, 2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37146162

RESUMEN

Therapeutic proteins can be challenging to develop due to their complexity and the requirement of an acceptable formulation to ensure patient safety and efficacy. To date, there is no universal formulation development strategy that can identify optimal formulation conditions for all types of proteins in a fast and reliable manner. In this work, high-throughput characterization, employing a toolbox of five techniques, was performed on 14 structurally different proteins formulated in 6 different buffer conditions and in the presence of 4 different excipients. Multivariate data analysis and chemometrics were used to analyze the data in an unbiased way. First, observed changes in stability were primarily determined by the individual protein. Second, pH and ionic strength are the two most important factors determining the physical stability of proteins, where there exists a significant statistical interaction between protein and pH/ionic strength. Additionally, we developed prediction methods by partial least-squares regression. Colloidal stability indicators are important for prediction of real-time stability, while conformational stability indicators are important for prediction of stability under accelerated stress conditions at 40 °C. In order to predict real-time storage stability, protein-protein repulsion and the initial monomer fraction are the most important properties to monitor.


Asunto(s)
Anticuerpos Monoclonales , Quimiometría , Humanos , Estabilidad Proteica , Anticuerpos Monoclonales/química , Desplegamiento Proteico , Conformación Proteica , Estabilidad de Medicamentos
3.
Commun Biol ; 6(1): 386, 2023 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-37031320

RESUMEN

The angiotensin-converting enzyme 2 (ACE2) is a viral receptor used by sarbecoviruses to infect cells. Fusion proteins comprising extracellular ACE2 domains and the Fc part of immunoglobulins exhibit high virus neutralization efficiency, but the structure and stability of these molecules are poorly understood. We show that although the hinge between the ACE2 and the IgG4-Fc is highly flexible, the conformational dynamics of the two ACE2 domains is restricted by their association. Interestingly, the conformational stability of the ACE2 moiety is much lower than that of the Fc part. We found that chemical compounds binding to ACE2, such as DX600 and MLN4760, can be used to strongly increase the thermal stability of the ACE2 by different mechanisms. Together, our findings reveal a general concept for stabilizing the labile receptor segments of therapeutic antiviral fusion proteins by chemical compounds.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Antivirales/química , Enzima Convertidora de Angiotensina 2/metabolismo , Unión Proteica
4.
Biotechnol Adv ; 64: 108120, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36764335

RESUMEN

In contrast to other species, cattle possess exceptional antibodies with ultra-long complementarity-determining regions (ulCDRs) that can consist of 40-70 amino acids. The bovine ulCDR is folded into a stalk and a disulfide-rich knob domain. The binding to the antigen is via the 3-6 kDa knob. There exists an immense sequence and structural diversity in the knob that enables binding to different antigens. Here we summarize the current knowledge of the ulCDR structure and provide an overview of the approaches to discover ulCDRs against novel antigens. Furthermore, we outline protein engineering approaches inspired by the natural ulCDRs. Finally, we discuss the enormous potential of using isolated bovine knobs, also named picobodies, as the smallest antigen-binding domains derived from natural antibodies.


Asunto(s)
Anticuerpos , Regiones Determinantes de Complementariedad , Bovinos , Animales , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/metabolismo , Anticuerpos/genética , Anticuerpos/química , Antígenos , Aminoácidos
5.
MAbs ; 15(1): 2164459, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36629855

RESUMEN

Antibody drugs should exhibit not only high-binding affinity for their target antigens but also favorable physicochemical drug-like properties. Such drug-like biophysical properties are essential for the successful development of antibody drug products. The traditional approaches used in antibody drug development require significant experimentation to produce, optimize, and characterize many candidates. Therefore, it is attractive to integrate new methods that can optimize the process of selecting antibodies with both desired target-binding and drug-like biophysical properties. Here, we summarize a selection of techniques that can complement the conventional toolbox used to de-risk antibody drug development. These techniques can be integrated at different stages of the antibody development process to reduce the frequency of physicochemical liabilities in antibody libraries during initial discovery and to co-optimize multiple antibody features during early-stage antibody engineering and affinity maturation. Moreover, we highlight biophysical and computational approaches that can be used to predict physical degradation pathways relevant for long-term storage and in-use stability to reduce the need for extensive experimentation.


Asunto(s)
Anticuerpos Monoclonales , Descubrimiento de Drogas , Anticuerpos Monoclonales/química , Descubrimiento de Drogas/métodos , Desarrollo de Medicamentos
6.
Structure ; 31(1): 2-3, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36608664

RESUMEN

In this issue of Structure, Ge et al. report an epitope-directed strategy to select antibodies specific for Frizzled subtypes. Structural and biochemical analyses provide mechanistic insights into the target binding of the isolated antibodies that could guide the design of reagents and therapeutics targeting distinct Frizzled receptors.


Asunto(s)
Anticuerpos , Receptores Frizzled , Epítopos/química
7.
FEBS J ; 290(6): 1398-1419, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35122394

RESUMEN

Light chain amyloidosis (AL) is a systemic disease in which abnormally proliferating plasma cells secrete large amounts of mutated antibody light chains (LCs) that eventually form fibrils. The fibrils are deposited in various organs, most often in the heart and kidney, and impair their function. The prognosis for patients diagnosed with AL is generally poor. The disease is set apart from other amyloidoses by the huge number of patient-specific mutations in the disease-causing and fibril-forming protein. The molecular mechanisms that drive the aggregation of mutated LCs into fibrils have been enigmatic, which hindered the development of efficient diagnostics and therapies. In this review, we summarize our current knowledge on AL amyloidosis and discuss open issues.


Asunto(s)
Amiloidosis , Humanos , Amiloidosis/genética , Amiloidosis/metabolismo , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/metabolismo , Pronóstico , Células Plasmáticas/metabolismo , Anticuerpos , Amiloide/genética , Amiloide/metabolismo
8.
J Pharm Sci ; 112(2): 404-410, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36257338

RESUMEN

Surfactants are commonly used in biopharmaceutical formulations to stabilize proteins against aggregation. However, the choice of a suitable surfactant for a particular protein is decided mostly empirically, and their mechanism of action on molecular level is largely unknown. Here we show that a straightforward label-free method, saturation transfer difference (STD) nuclear magnetic resonance (NMR) spectroscopy, can be used to detect protein-surfactant interactions in formulations of a model protein, interferon alpha. We find that polysorbate 20 binds with its fatty acid to interferon, and that the binding is stronger at pH closer to the isoelectric point of the protein. In contrast, we did not detect interactions between poloxamer 407 and interferon alpha. Neither of the two surfactants affected the tertiary structure and the thermal stability of the protein as evident from circular dichroism and nanoDSF measurements. Interestingly, both surfactants inhibited the formation of subvisible particles during long-term storage, but only polysorbate 20 reduced the amount of small soluble aggregates detected by size-exclusion chromatography. This proof-of-principle study demonstrates how STD-NMR can be employed to quickly assess surfactant-protein interactions and support the choice of surfactant in protein formulation.


Asunto(s)
Polisorbatos , Tensoactivos , Tensoactivos/química , Polisorbatos/química , Interferón-alfa , Espectroscopía de Resonancia Magnética/métodos , Proteínas/química
9.
Commun Biol ; 5(1): 1237, 2022 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-36371561

RESUMEN

Coronavirus infections are a world-wide threat to human health. A promising strategy to develop a broadly active antiviral is the use of fusion proteins consisting of an antibody IgG Fc region and a human ACE2 domain to which the viral spike proteins bind. Here we create antiviral fusion proteins based on IgM scaffolds. The hexameric ACE2-IgM-Fc fusions can be efficiently produced in mammalian cells and they neutralize the infectious virus with picomolar affinity thus surpassing monomeric ACE2-IgM-Fc by up to 96-fold in potency. In addition, the ACE2-IgM fusion shows increased neutralization efficiency for the highly infectious SARS-CoV-2 omicron variant in comparison to prototypic SARS-CoV-2. Taken together, these multimeric IgM fusions proteins are a powerful weapon to fight coronavirus infections.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Animales , Humanos , SARS-CoV-2/genética , Enzima Convertidora de Angiotensina 2/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Antivirales/farmacología , Peptidil-Dipeptidasa A , Unión Proteica , Inmunoglobulina M , Mamíferos
10.
J Pharm Sci ; 111(8): 2149-2157, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35598781

RESUMEN

Monoclonal antibodies (mAbs) have been immensely successful as biological drugs. However, the treatment of some diseases requires combinations of antibodies that bind to different pharmacological targets. An elegant approach to delivering the therapeutic potential of antibody combinations is to develop drug products based on fixed-dose combinations (FDCs) of co-formulated mAbs. Since the first FDA approval of two co-formulated mAbs in 2020, the interest in antibody FDCs is increasing. However, there are different strategies to develop co-formulated antibodies and unique challenges related to their analytical characterization. In this review, we summarize the recent progress on antibody FDCs with a focus on important considerations during drug development and the analytical toolbox for co-formulated mAbs.


Asunto(s)
Anticuerpos Monoclonales , Desarrollo de Medicamentos , Anticuerpos Monoclonales/uso terapéutico
11.
Antiviral Res ; 196: 105197, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34774603

RESUMEN

SARS-CoV-2 enters host cells after binding through its spike glycoprotein to the angiotensin-converting enzyme 2 (ACE2) receptor. Soluble ACE2 ectodomains bind and neutralize the virus, yet their short in vivo half-live limits their therapeutic use. This limitation can be overcome by fusing the fragment crystallizable (Fc) part of human immunoglobulin G (IgG) to the ACE2 ectodomain, but this bears the risk of Fc-receptor activation and antibody-dependent cellular cytotoxicity. Here, we describe optimized ACE2-IgG4-Fc fusion constructs that avoid Fc-receptor activation, preserve the desired ACE2 enzymatic activity and show promising pharmaceutical properties. The engineered ACE2-IgG4-Fc fusion proteins neutralize the original SARS-CoV, pandemic SARS-CoV-2 as well as the rapidly spreading SARS-CoV-2 alpha, beta and delta variants of concern. Importantly, these variants of concern are inhibited at picomolar concentrations proving that ACE2-IgG4 maintains - in contrast to therapeutic antibodies - its full antiviral potential. Thus, ACE2-IgG4-Fc fusion proteins are promising candidate anti-antivirals to combat the current and future pandemics.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Antivirales/síntesis química , Tratamiento Farmacológico de COVID-19 , Inmunoglobulina G , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/química , Enzima Convertidora de Angiotensina 2/uso terapéutico , Antivirales/uso terapéutico , Humanos , Unión Proteica , SARS-CoV-2/efectos de los fármacos
12.
Nat Commun ; 12(1): 6737, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795299

RESUMEN

Antibodies bind antigens via flexible loops called complementarity-determining regions (CDRs). These are usually 6-20 residues long. However, some bovine antibodies have ultra-long CDRs comprising more than 50 residues organized in a stalk and a disulfide-rich knob. The design features of this structural unit and its influence on antibody stability remained enigmatic. Here, we show that the stalk length is critical for the folding and stability of antibodies with an ultra-long CDR and that the disulfide bonds in the knob do not contribute to stability; they are important for organizing the antigen-binding knob structure. The bovine ultra-long CDR can be integrated into human antibody scaffolds. Furthermore, mini-domains from de novo design can be reformatted as ultra-long CDRs to create unique antibody-based proteins neutralizing SARS-CoV-2 and the Alpha variant of concern with high efficiency. Our findings reveal basic design principles of antibody structure and open new avenues for protein engineering.


Asunto(s)
Regiones Determinantes de Complementariedad/genética , SARS-CoV-2/genética , Animales , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , COVID-19/genética , Bovinos
13.
Mol Pharm ; 18(6): 2242-2253, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33928776

RESUMEN

The efficient development of new therapeutic antibodies relies on developability assessment with biophysical and computational methods to find molecules with drug-like properties such as resistance to aggregation. Despite the many novel approaches to select well-behaved proteins, antibody aggregation during storage is still challenging to predict. For this reason, there is a high demand for methods that can identify aggregation-resistant antibodies. Here, we show that three straightforward techniques can select the aggregation-resistant antibodies from a dataset with 13 molecules. The ReFOLD assay provided information about the ability of the antibodies to refold to monomers after unfolding with chemical denaturants. Modulated scanning fluorimetry (MSF) yielded the temperatures that start causing irreversible unfolding of the proteins. Aggregation was the main reason for poor unfolding reversibility in both ReFOLD and MSF experiments. We therefore performed temperature ramps in molecular dynamics (MD) simulations to obtain partially unfolded antibody domains in silico and used CamSol to assess their aggregation potential. We compared the information from ReFOLD, MSF, and MD to size-exclusion chromatography (SEC) data that shows whether the antibodies aggregated during storage at 4, 25, and 40 °C. Contrary to the aggregation-prone molecules, the antibodies that were resistant to aggregation during storage at 40 °C shared three common features: (i) higher tendency to refold to monomers after unfolding with chemical denaturants, (ii) higher onset temperature of nonreversible unfolding, and (iii) unfolding of regions containing aggregation-prone sequences at higher temperatures in MD simulations.


Asunto(s)
Anticuerpos Monoclonales/química , Desnaturalización Proteica , Anticuerpos Monoclonales/uso terapéutico , Rastreo Diferencial de Calorimetría , Química Farmacéutica/métodos , Cromatografía en Gel , Almacenaje de Medicamentos , Dispersión Dinámica de Luz , Calor/efectos adversos , Concentración de Iones de Hidrógeno , Simulación de Dinámica Molecular , Conformación Proteica , Pliegue de Proteína , Desplegamiento Proteico
14.
Mol Pharm ; 17(8): 2971-2986, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32687367

RESUMEN

Coformulations containing two therapeutic monoclonal antibodies (mAbs) could offer various benefits like enhanced therapeutic efficacy and better patient compliance. However, there are very few published studies on coformulations and binary mixtures of mAbs. It remains unclear to what extent mAbs with different physicochemical properties can be combined in solution without detrimental effects on protein stability. Here, we present a study including six model mAbs of the IgG1 subclass that are commercially available. In silico and biophysical characterization shows that the proteins have different physicochemical properties. Thus, their combinations represent various scenarios for coformulation development. We prepared all possible binary mixtures of the six mAbs and determined several biophysical parameters that are assessed during early-stage protein drug product development. The measured biophysical parameters are indicative of the conformational protein stability (inflection points of the thermal protein unfolding transitions) and the colloidal protein stability (aggregation onset temperatures and interaction parameter kD from dynamic light scattering). Remarkably, all 15 binary mAb mixtures do not exhibit biophysical parameters that indicate inferior conformational or colloidal stability compared to the least stable mAb in the mixture. Our findings suggest that the coformulation of some therapeutic monoclonal antibodies of the IgG1 subclass could be possible in a straightforward way as severe detrimental effects on the stability of these proteins in binary mixtures were not observed.


Asunto(s)
Anticuerpos Monoclonales/química , Preparaciones Farmacéuticas/química , Biofisica/métodos , Inmunoglobulina G/química , Estabilidad Proteica/efectos de los fármacos , Desplegamiento Proteico/efectos de los fármacos
15.
Mol Pharm ; 17(7): 2638-2647, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32401526

RESUMEN

Determining the temperature at which the thermal unfolding of a protein starts becoming irreversible is relevant for many areas of protein research. Until now, published methods cannot determine, within a reasonable time frame and with moderate sample consumption, the exposure temperature that starts causing irreversible protein unfolding. We present modulated scanning fluorimetry (MSF) and share a software (MSF Analyzer), which can be used to derive nonreversibility curves of thermal protein unfolding from a series of incremental temperature cycles performed on only 10 µL samples, consuming as low as a few micrograms of protein. Further processing of the data can yield the onset temperature that starts causing nonreversible protein unfolding. The MSF method is based on the hardware of the already existing nanoDSF technology and can be applied to dozens of samples simultaneously. Here, we use MSF to study how solution pH affects the reversibility of thermal protein unfolding of several model proteins to show that the nonreversibility onset temperature (Tnr) is a unique biophysical parameter, providing orthogonal information from thermal protein denaturation data and insights into the validity of thermal unfolding analysis in the context of equilibrium thermodynamics. We also show that MSF can be used to study enzyme stability after exposure to high temperatures. Besides, we demonstrate that protein thermal unfolding and nonreversibility can be affected in different ways upon modifications like PEG-ylation or labeling with fluorescent dyes. Finally, we show that MSF can be used to study the effect of various protein interactions on thermal protein unfolding reversibility. With the diverse examples in this work, we reveal how MSF can provide orthogonal information from thermal denaturation experiments that can bring benefits to various areas of protein research. The MSF Analyzer software is available at https://github.com/CoriolisPharmaResearch/MSFAnalyser.


Asunto(s)
Fluorometría/métodos , Pliegue de Proteína , Desplegamiento Proteico , Proteínas/química , Rastreo Diferencial de Calorimetría , Colorantes Fluorescentes/química , Calor , Concentración de Iones de Hidrógeno , Cinética , Muramidasa/química , Ovalbúmina/química , Polietilenglicoles/química , Desnaturalización Proteica , Programas Informáticos , Termodinámica , Trastuzumab/química , Ubiquitina/química
16.
Eur J Pharm Biopharm ; 151: 127-136, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32283214

RESUMEN

One of the major challenges in formulation development of biopharmaceuticals is improving long-term storage stability, which is often achieved by addition of excipients to the final formulation. Finding the optimal excipient for a given protein is usually done using a trial-and-error approach, due to the lack of general understanding of how excipients work for a particular protein. Previously, preferential interactions (binding or exclusion) of excipients with proteins were postulated as a mechanism explaining diversity in the stabilisation effects. Weak preferential binding is however difficult to quantify experimentally, and the question remains whether the formulation process should seek excipients which preferentially bind with proteins, or not. Here, we apply solution NMR spectroscopy to comprehensively evaluate protein-excipient interactions between therapeutically relevant proteins and commonly used excipients. Additionally, we evaluate the effect of excipients on thermal and colloidal protein stability, on aggregation kinetics and protein storage stability at elevated temperatures. We show that there is a weak negative correlation between the strength of protein-excipient interactions and effect on enhancing protein thermal stability. We found that the overall protein-excipient binding per se can be a poor criterion for choosing excipients enhancing formulation stability. Experiments on a diverse set of excipients and test proteins reveal that while excipients affect all of the different aspects of protein stability, the effects are very much protein specific, and care must be taken to avoid apparent generalisations if a smaller dataset is being used.


Asunto(s)
Productos Biológicos/química , Excipientes/química , Unión Proteica/fisiología , Proteínas/química , Química Farmacéutica/métodos , Estabilidad de Medicamentos , Cinética , Estabilidad Proteica
17.
J Pharm Sci ; 109(6): 2048-2058, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32194093

RESUMEN

Understanding the formulation features that ensure sufficient stability during long-term storage is critical for developing next-generation therapeutic proteins. In this work, we investigate the physical stability of a bispecific antibody (Bis-mAb) in 12 different formulation conditions. Isothermal chemical denaturation with urea indicates a higher resistance to denaturant-induced unfolding when pH is increased from 5.0 to 6.5 but shows minor influence from the buffer type and ionic strength. Dynamic and static light scattering are used to derive the interaction parameter (kD) and second virial coefficient (A2), respectively. These two parameters indicate that Bis-mAb exhibits highest colloidal stability in formulations containing 10 mM histidine buffer without added sodium chloride. Further, we observe that the highest relative monomer yield (RMY) after isothermal refolding, that is the highest refoldability, from urea is measured for the low ionic strength histidine formulations. Finally, we show long-term stability data on all 12 Bis-mAb formulations after storage at 4 °C and 25 °C for 12 months. The least amount of soluble aggregates and subvisible particles were detected in the Bis-mAb formulations with the highest colloidal stability and refoldability from urea. We suggest that the optimization of these two features is crucial for obtaining physically stable formulations of Bis-mAb.


Asunto(s)
Anticuerpos Biespecíficos , Agregado de Proteínas , Anticuerpos Monoclonales , Concentración Osmolar , Estabilidad Proteica
18.
J Pharm Sci ; 109(1): 584-594, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31689429

RESUMEN

Understanding the effects of additives on therapeutic protein stability is of paramount importance for obtaining stable formulations. In this work, we apply several high- and medium-throughput methods to study the physical stability of a model monoclonal antibody at pH 5.0 and 6.5 in the presence of sucrose, arginine hydrochloride, and arginine glutamate. In low ionic strength buffer, the addition of salts reduces the antibody colloidal and thermal stability, attributed to screening of electrostatic interactions. The presence of glutamate ion in the arginine salt partially reduces the damaging effect of ionic strength increase. The addition of 280 mM sucrose shifts the thermal protein unfolding to a higher temperature. Arginine salts in the used concentration reduce the relative monomer yield after refolding from urea, whereas sucrose has a favorable effect on antibody refolding. In addition, we show 12-month long-term stability data and observe correlations between thermal protein stability, relative monomer yield after refolding, and monomer loss during storage. The monomer loss during storage is related to protein aggregation and formation of subvisible particles in some of the formulations. This study shows that the effect of commonly used additives on the long-term antibody physical stability can be predicted using orthogonal biophysical measurements.


Asunto(s)
Anticuerpos Monoclonales/química , Arginina/química , Dipéptidos/química , Sacarosa/química , Tampones (Química) , Coloides , Composición de Medicamentos , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Concentración de Iones de Hidrógeno , Agregado de Proteínas , Estabilidad Proteica , Desplegamiento Proteico , Temperatura , Factores de Tiempo
19.
Mol Pharm ; 17(2): 426-440, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31790599

RESUMEN

Therapeutic protein candidates should exhibit favorable properties that render them suitable to become drugs. Nevertheless, there are no well-established guidelines for the efficient selection of proteinaceous molecules with desired features during early stage development. Such guidelines can emerge only from a large body of published research that employs orthogonal techniques to characterize therapeutic proteins in different formulations. In this work, we share a study on a diverse group of proteins, including their primary sequences, purity data, and computational and biophysical characterization at different pH and ionic strength. We report weak linear correlations between many of the biophysical parameters. We suggest that a stability comparison of diverse therapeutic protein candidates should be based on a computational and biophysical characterization in multiple formulation conditions, as the latter can largely determine whether a protein is above or below a certain stability threshold. We use the presented data set to calculate several stability risk scores obtained with an increasing level of analytical effort and show how they correlate with protein aggregation during storage. Our work highlights the importance of developing combined risk scores that can be used for early stage developability assessment. We suggest that such scores can have high prediction accuracy only when they are based on protein stability characterization in different solution conditions.


Asunto(s)
Anticuerpos Monoclonales/química , Descubrimiento de Drogas/métodos , Inmunoglobulina G/química , Interferón alfa-2/química , Desplegamiento Proteico , Albúmina Sérica Humana/química , Transferrina/química , Secuencia de Aminoácidos , Almacenaje de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Concentración Osmolar , Agregado de Proteínas , Estabilidad Proteica , Proyectos de Investigación , Solubilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...