Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Chromatogr A ; 1716: 464632, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38219623

RESUMEN

Recombinant adeno-associated virus (AAV) has emerged as one of the most promising systems for therapeutic gene delivery and has demonstrated clinical success in a wide range of genetic disorders. However, manufacturing of high-quality AAV in large amounts still remains a challenge. A significant difficulty for downstream processing is the need to remove empty capsids that are generated in all currently utilized expression systems and that represent product-related impurities that adversely affect safety and efficacy of AAV vectors. Empty and full capsids exhibit only subtle differences in surface charge and size, making chromatography-based separations highly challenging. Here, we present a rapid methodology for the systematic process development of the crucial AAV full/empty capsid separation on ion-exchange media based on high-throughput screening and mechanistic modeling. Two of the most commonly employed serotypes, AAV8 and AAV9, are used as case studies. First, high-throughput studies in filter-plate format are performed that allow the rapid and comprehensive study of binding and elution behavior of AAV on different resins, using different buffer systems, pH, salt conditions, and solution additives. Small amounts of separated empty and full AAV capsids are generated by iodixanol gradient centrifugation that allow studying the binding and elution behavior of the two vector species separately in miniaturized format. Process conditions that result in maximum differences in elution behavior between empty and full capsids are then transferred to benchtop chromatography systems that are used to generate calibration data for the estimation of steric mass-action isotherm and mass transport parameters for process simulation. The resulting column models are employed for in-silico process development that serves to enhance understanding of separation constraints and to identify optimized conditions for the removal of empty particles. Finally, optimized separation conditions are verified experimentally. The methodology presented in this work provides a systematic framework that affords mechanistic understanding of the crucial empty/full capsid separation and accelerates the development of a scalable AAV downstream process.


Asunto(s)
Cápside , Dependovirus , Cápside/química , Cápside/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Ensayos Analíticos de Alto Rendimiento , Vectores Genéticos , Proteínas de la Cápside/genética , Proteínas de la Cápside/análisis
2.
3.
Front Bioeng Biotechnol ; 11: 1123842, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37082211

RESUMEN

The manufacturing of antibody-drug conjugates (ADCs) involves the addition of a cytotoxic small-molecule linker-drug (= payload) to a solution of functionalized antibodies. For the development of robust conjugation processes, initially small-scale reaction tubes are used which requires a lot of manual handling. Scale-up to larger reaction vessels is often knowledge-driven and scale-comparability is solely assessed based on final product quality which does not account for the dynamics of the reaction. In addition, information about the influence of process parameters, such as stirrer speed, temperature, or payload addition rates, is limited due to high material costs. Given these limitations, there is a need for a modeling-based approach to investigate conjugation scale-up. In this work, both experimental kinetic studies and computational fluid dynamics (CFD) conjugation simulations were performed to understand the influence of scale and mixing parameters. In the experimental part, conjugation kinetics in small-scale reaction tubes with different mixing types were investigated for two ADC systems and compared to larger bench-scale reactions. It was demonstrated that more robust kinetics can be achieved through internal stirrer mixing instead of external mixing devices, such as orbital shakers. In the simulation part, 3D-reactor models were created by coupling CFD-models for three large-scale reaction vessels with a kinetic model for a site-specific conjugation reaction. This enabled to study the kinetics in different vessels, as well as the effect of process parameter variations in silico. Overall, it was found that for this conjugation type sufficient mixing can be achieved at all scales and the studied parameters cause only deviations during the payload addition period. An additional time-scale analysis demonstrated to aid the assessment of mixing effects during ADC process scale-up when mixing times and kinetic rates are known. In summary, this work highlights the benefit of kinetic models for enhanced conjugation process understanding without the need for large-scale experiments.

4.
J Chromatogr A ; 1653: 462378, 2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34311388

RESUMEN

In this paper, a methodology for the development of a multimodal chromatography process is presented that is aimed at removal of under-conjugated antibody-drug conjugate (ADC) species. Two ADCs are used as case studies: One ADC results from site-directed conjugation to inserted cysteine residues and has a drug-to-antibody ratio (DAR) of two, the other is the product of conjugation to interchain disulfide bonds with a DAR of eight. First, filter plate screening studies are designed for the unconjugated antibody and the ADCs. Different metrics for the analysis of these data sets are presented and discussed. From this analysis, the selected process conditions are then carried out using a benchtop chromatography system to confirm the separations observed in the filter plate studies while simultaneously generating data to estimate steric mass-action isotherm and mass transport parameters for process simulation. This column model is then employed to develop separation processes in-silico for the removal of the unconjugated parent antibody and under-conjugated product variants. The optimized process conditions identified using the model are then verified experimentally. The methodology presented in this work utilizes multimodal chromatography for ADC purification and provides the framework for a streamlined systematic approach to process development.


Asunto(s)
Química Farmacéutica , Cromatografía Liquida , Inmunoconjugados , Química Farmacéutica/métodos , Simulación por Computador , Cisteína , Ensayos Analíticos de Alto Rendimiento , Inmunoconjugados/aislamiento & purificación
5.
MAbs ; 11(6): 1064-1076, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31198090

RESUMEN

Site-specific antibody-drug conjugates (ADCs) are designed to overcome the heterogeneity observed with first-generation ADCs that use random conjugation to surface-exposed lysine residues or conjugation to interchain disulfide bonds. Despite significantly enhanced homogeneity, however, the production of site-specific ADCs yields some process-related species heterogeneity, including stereoisomers, unconjugated antibody, underconjugated species, and overconjugated species. An elevated level of size variants, such as heavy chain-light chain species (half ADC), heavy chain-heavy chain-light chain species, and light chain species, is also observed with the final site-specific ADC product. To understand the root cause of heterogeneity generated during the ADC conjugation process, we designed time-course studies for each conjugation step, including reduction, oxidation, conjugation, and quenching. We developed both non-reduced peptide map and LabChip-based capillary electrophoresis sodium dodecyl sulfate methods for time-course sample analysis. On the basis of our time-course data, the half ADC and unconjugated antibody were generated during oxidation as a result of alternative disulfide bond arrangements. During oxidation, two hinge cysteines formed an intra-chain disulfide bond in the half ADC, and three inter-chain hinge disulfide bonds were formed in the unconjugated antibody. Time-course data also showed that the elevated level of size variants, especially heavy chain-heavy chain-light chain species and light chain species, resulted from the quenching step, where the quenching reagent engaged in a disulfide bond exchange reaction with the ADC and broke the disulfide bonds connecting the heavy chain and light chain. Underconjugated and overconjugated species arose from the equilibrium established during the conjugation reaction.


Asunto(s)
Anticuerpos Monoclonales/química , Especificidad de Anticuerpos , Inmunoconjugados/química , Cadenas Pesadas de Inmunoglobulina/química , Humanos , Oxidación-Reducción
6.
J Biotechnol ; 288: 15-22, 2018 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-30321572

RESUMEN

The conjugation reaction of monoclonal antibodies (mAbs) with small-molecule drugs is a central step during production of antibody-drug conjugates (ADCs). The ability to monitor this step in real time can be advantageous for process understanding and control. Here, we propose a method based on UV/Vis spectroscopy in conjunction with partial least squares (PLS) regression for non-invasive monitoring of conjugation reactions. In experiments, the method was applied to conjugation reactions with two surrogate drugs in microplate format as well as at 20 ml scale. All calibrated PLS models performed well in cross-validation (Q2>0.975 for all models). In microplate format, the PLS models were furthermore successfully validated with an independent prediction set (Rpred2=0.9770 resp. 0.8940). In summary, the proposed method provides a quick and easily implementable tool for reaction monitoring of ADC conjugation reactions and may in the future support the implementation of Process Analytical Technologies (PAT).


Asunto(s)
Anticuerpos Monoclonales/química , Cumarinas/química , Inmunoconjugados/química , Inmunoglobulina G/química , Maleimidas/química , Espectrofotometría Ultravioleta
7.
Biotechnol Prog ; 34(5): 1195-1204, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30094961

RESUMEN

This work presents the optimization and critical evaluation of continuous capture chromatography in the downstream process of a recombinant enzyme. For the upstream manufacturing of this molecule, a perfusion process was implemented due to benefits for product quality and productivity. This process is, however, characterized by low titer and significant changes over the course of the harvest duration in terms of active enzyme concentration and impurity content. We evaluated the feasibility and benefits of a continuous capture operation. This case study illustrates the design approach that can be utilized to address challenges presented by a changing feedstream, and the statistical measures that can be employed to characterize and optimize the operating space under material and time constraints. Process economic modeling in conjunction with Monte Carlo simulations indicate that even for a nonaffinity capture step utilizing a relatively cheap ion-exchange resin, the smaller column volume used in a continuous set-up results in cost savings compared to the batch process. We compare this option to the scenario of repeated processing using a small capture column in batch mode. Our analysis establishes that continuous processing becomes economically attractive for processes where only a small portion of the potential column lifetime can be utilized or for column steps with slow mass transport and shallow breakthrough curves. In cases where column breakthrough is sharp and resin lifetime is relatively short, continuous processing may offer an improvement over traditional batch processing, but much of the productivity and cost savings can be realized through repeated column cycling. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 2018 © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1195-1204, 2018.


Asunto(s)
Cromatografía/métodos , Proteínas Recombinantes/metabolismo , Animales , Células CHO , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Cricetulus , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
8.
J Biotechnol ; 278: 48-55, 2018 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-29733878

RESUMEN

Antibody-drug conjugates (ADCs) form a rapidly growing class of biopharmaceuticals which attracts a lot of attention throughout the industry due to its high potential for cancer therapy. They combine the specificity of a monoclonal antibody (mAb) and the cell-killing capacity of highly cytotoxic small molecule drugs. Site-specific conjugation approaches involve a multi-step process for covalent linkage of antibody and drug via a linker. Despite the range of parameters that have to be investigated, high-throughput methods are scarcely used so far in ADC development. In this work an automated high-throughput platform for a site-specific multi-step conjugation process on a liquid-handling station is presented by use of a model conjugation system. A high-throughput solid-phase buffer exchange was successfully incorporated for reagent removal by utilization of a batch cation exchange step. To ensure accurate screening of conjugation parameters, an intermediate UV/Vis-based concentration determination was established including feedback to the process. For conjugate characterization, a high-throughput compatible reversed-phase chromatography method with a runtime of 7 min and no sample preparation was developed. Two case studies illustrate the efficient use for mapping the operating space of a conjugation process. Due to the degree of automation and parallelization, the platform is capable of significantly reducing process development efforts and material demands and shorten development timelines for antibody-drug conjugates.


Asunto(s)
Anticuerpos Monoclonales , Ensayos Analíticos de Alto Rendimiento/métodos , Inmunoconjugados , Anticuerpos Monoclonales/análisis , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/metabolismo , Automatización de Laboratorios , Resinas de Intercambio de Catión , Cromatografía de Fase Inversa , Cisteína/química , Cisteína/metabolismo , Inmunoconjugados/análisis , Inmunoconjugados/química , Inmunoconjugados/aislamiento & purificación , Inmunoconjugados/metabolismo , Modelos Químicos
9.
J Pharm Sci ; 106(11): 3280-3286, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28734785

RESUMEN

Stainless steel containers are widely used in the pharmaceutical and biopharmaceutical industry for the storage of buffers, process intermediates, and purified drug substance. They are generally held to be corrosion resistant, biocompatible, and nonreactive, although it is well established that trace amounts of metal ions can leach from stainless steel equipment into biopharmaceutical products. We report here that the use of stainless steel containers in conjunction with magnetic stirring bars leads to significantly aggravated metal contamination, consisting of both metal particles and significantly elevated metal ions in solution, the degree of which is several orders of magnitude higher than described for static conditions. Metal particles are analyzed by scanning electron microscopy with electron-dispersive X-ray spectroscopy, and metal content in solution is quantitated at different time points by inductively coupled plasma-mass spectrometry. The concentration of iron, chromium, nickel, and manganese increases with increasing stirring time and speed. We describe the impact of buffer components on the extent of metal particles and ions in solution and illustrate the effect on model proteins.


Asunto(s)
Composición de Medicamentos/métodos , Embalaje de Medicamentos/métodos , Metales/análisis , Acero Inoxidable/química , Tampones (Química) , Cromo/análisis , Corrosión , Contaminación de Medicamentos , Hierro/análisis , Magnetismo/métodos , Imanes/química , Manganeso/análisis , Níquel/análisis , Agregado de Proteínas
10.
PDA J Pharm Sci Technol ; 71(2): 115-126, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28289128

RESUMEN

The analysis and accurate quantitation of bioconjugations proves challenging in the case of oligomeric proteins, especially when the size of the molecule or the nature of the conjugate do not allow the analysis of the intact protein under native conditions. In this case, analytical methods are frequently applied that result in a dissociation of non-covalently linked subunits. This limits the analysis to a description of individual subunits, thereby obscuring the accurate characterization of the overall functionalization. This situation is frequently encountered in the biopharmaceutically important case of protein PEGylation, as the biophysical properties of the PEG polymer generally make analysis and accurate quantitation for a protein with multiple conjugation sites challenging under native conditions. In this work we present a statistical measure for deriving the overall functionalization of an oligomeric protein from the data obtained from readily accessible assays that cause non-covalently associated subunits to dissociate. This approach is broadly applicable for the characterization and optimization of bioconjugation reactions for multimeric biomolecules. It should also be highly valuable for the accurate description of composition and manufacturing consistency of conjugated biotherapeutics in regulatory filings.LAY ABSTRACT: Conjugated proteins are an important class of biopharmaceuticals. For these molecules, successful drug development requires accurate methods for the quantitative characterization of protein conjugation. This task is particularly challenging in the case of proteins consisting of several, non-covalently linked subunits, especially when the size of the protein or nature of the conjugate do not allow for analysis of the intact oligomeric molecule. Many of the analytical methods used to characterize these conjugates, such as reverse phase high-performance liquid chromatography, cause the individual subunits to dissociate, making it difficult to fully understand quality attributes at the native oligomeric level. We present a method to accurately quantify the overall conjugation of an oligomeric protein in these cases when readily available assays describe only individual subunits. This should be highly valuable for process optimization and to correctly characterize the conjugated biopharmaceutical in interactions with regulatory agencies.


Asunto(s)
Química Farmacéutica/métodos , Polietilenglicoles/química , Proteínas Recombinantes/química , Sitios de Unión , Química Farmacéutica/estadística & datos numéricos , Cromatografía Líquida de Alta Presión , Cromatografía de Fase Inversa , Maleimidas/química , Modelos Estadísticos , Unión Proteica , Conformación Proteica
11.
Biotechnol J ; 12(2)2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27677057

RESUMEN

Interest in new and diverse classes of molecules such as recombinant toxins, enzymes, and blood factors continues to grow for use a biotherapeutics. Compared to monoclonal antibodies, these novel drugs typically lack a commercially available affinity chromatography option, which leads to greater process complexity, longer development timelines, and poor platformability. To date, for both monoclonal antibodies and novel molecules, affinity chromatography has been mostly reserved for separation of process-related impurities such as host cell proteins and DNA. Reports of affinity purification of closely related product variants and modified forms are much rarer. In this work we describe custom affinity chromatography development using camelid VH H antibody fragments as "tunable" immunoaffinity ligands for separation of product-related impurities. One example demonstrates high selectivity for a recombinant immunotoxin where no binding was observed for an undesired deamidated species. Also discussed is affinity purification of a coagulation factor through specific recognition of the gamma-carboxylglutamic acid domain.


Asunto(s)
Cromatografía de Afinidad/métodos , Anticuerpos/aislamiento & purificación , Toxinas Bacterianas/aislamiento & purificación , Exotoxinas/aislamiento & purificación , Protrombina/aislamiento & purificación
12.
PLoS One ; 11(12): e0167935, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28002433

RESUMEN

Humans and higher primates are unique in that they lack uricase, the enzyme capable of oxidizing uric acid. As a consequence of this enzyme deficiency, humans have high serum uric acid levels. In some people, uric acid levels rise above the solubility limit resulting in crystallization in joints, acute inflammation in response to those crystals causes severe pain; a condition known as gout. Treatment for severe gout includes injection of non-human uricase to reduce serum uric acid levels. Krystexxa® is a hyper-PEGylated pig-baboon chimeric uricase indicated for chronic refractory gout that induces an immunogenic response in 91% of treated patients, including infusion reactions (26%) and anaphylaxis (6.5%). These properties limit its use and effectiveness. An innovative approach has been used to develop a therapeutic uricase with improved properties such as: soluble expression, neutral pH solubility, high E. coli expression level, thermal stability, and excellent activity. More than 200 diverse uricase sequences were aligned to guide protein engineering and reduce putative sequence liabilities. A single uricase lead candidate was identified, which showed low potential for immunogenicity in >200 human donor samples selected to represent diverse HLA haplotypes. Cysteines were engineered into the lead sequence for site specific PEGylation and studies demonstrated >95% PEGylation efficiency. PEGylated uricase retains enzymatic activity in vitro at neutral pH, in human serum and in vivo (rats and canines) and has an extended half-life. In canines, an 85% reduction in serum uric acid levels was observed with a single subcutaneous injection. This PEGylated, non-immunogenic uricase has the potential to provide meaningful benefits to patients with gout.


Asunto(s)
Gota/tratamiento farmacológico , Urato Oxidasa/uso terapéutico , Animales , Rastreo Diferencial de Calorimetría , Perros , Escherichia coli/metabolismo , Semivida , Humanos , Concentración de Iones de Hidrógeno , Cinética , Papio , Polietilenglicoles/química , Ratas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Especificidad por Sustrato , Porcinos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Urato Oxidasa/efectos adversos , Urato Oxidasa/inmunología
13.
Biotechnol Prog ; 30(6): 1380-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25139260

RESUMEN

We describe the development and scale-up of a novel two chain immunotoxin refolding process. This work provides a case study comparing a clinical manufacturing process and the commercial process developed to replace it. While the clinical process produced high quality material, it suffered from low yield and high yield variability. A systematic approach to process development and understanding led to a number of improvements that were implemented in the commercial process. These include a shorter inclusion body recovery process, limiting the formation of an undesired deamidated species and the implementation of fed batch dilution refolding for increased refold titers. The use of a combination of urea, arginine and DTT for capture column cleaning restored the binding capacity of the capture step column and resulted in consistent capture step yields compared to the clinical process. Scalability is shown with data from 250 L and 950 L scale refolding processes. Compared to the clinical process it replaces, the commercial process demonstrated a greater than fivefold improvement in volumetric productivity at the 950 L refolding scale.


Asunto(s)
Inmunotoxinas/química , Inmunotoxinas/metabolismo , Replegamiento Proteico , Arginina/química , Ditiotreitol/química , Escherichia coli/genética , Escherichia coli/metabolismo , Fermentación , Inmunotoxinas/inmunología , Inmunotoxinas/aislamiento & purificación , Cuerpos de Inclusión/química , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Solubilidad , Urea/química
14.
Bioconjug Chem ; 25(1): 93-101, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24320725

RESUMEN

The conjugation of biomolecules by chemoselective oxime ligation is of great interest for the site-specific modification of proteins, peptides, nucleic acids, and carbohydrates. These conjugations proceed optimally at a reaction pH of 4-5, but some biomolecules are not soluble or stable under these conditions. Aniline can be used as a nucleophilic catalyst to enhance the rate of oxime formation, but even in its presence, the reaction rate at neutral pH can be slower than desired, particularly at low reagent concentrations and/or temperature. Recently, alternative catalysts with improved properties were reported, including anthranilic acid derivatives for small molecule ligations, as well as m-phenylenediamine at high concentrations for protein conjugations. Here, we report that p-substituted anilines containing an electron-donating ring substituent are superior catalysts of oxime-based conjugations at pH 7. One such catalyst, p-phenylenediamine, was studied in greater detail. This catalyst was highly effective at neutral pH, even at the low concentration of 2 mM. In a model oxime ligation using aminooxy-functionalized PEG, catalysis at pH 7 resulted in a 120-fold faster rate of protein PEGylation as compared to an uncatalyzed reaction, and 19-fold faster than the equivalent aniline-catalyzed reaction. p-Phenylenediamine (10 mM) was also an effective catalyst under acidic conditions and was more efficient than aniline throughout the pH range 4-7. This catalyst allows efficient oxime bioconjugations to proceed under mild conditions and low micromolar concentrations, as demonstrated by the PEGylation of a small protein.


Asunto(s)
Compuestos de Anilina/química , Oximas/síntesis química , Catálisis , Concentración de Iones de Hidrógeno , Modelos Moleculares , Estructura Molecular , Oximas/química
15.
J Chromatogr A ; 1325: 171-8, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24369997

RESUMEN

Prothrombin (coagulation Factor II) is a complex multidomain glycoprotein that plays a central role in blood coagulation. It is the zymogen precursor to the protease thrombin that catalyzes the formation of the fibrin clot and regulates a multitude of other cellular responses related to coagulation and hemostasis. For the biological activity of prothrombin, the vitamin K dependent posttranslational modification of glutamic acid residues to gamma-carboxylglutamic acid is of crucial importance. Prothrombin can be recombinantly expressed using mammalian cell culture. However, the product is a heterogeneous mixture of variants with different degrees of carboxylation, requiring separation of closely related charge isoforms. A second challenge for purification is the need to remove traces of the product-related impurity thrombin, a protease, to extremely low levels. In this work, we describe a purification strategy that provides solutions to both challenges and results in an efficient and robust process for active recombinant prothrombin. We also describe the analytical characterization of recombinant prothrombin by HPLC, LC-MS/MS, and complementary biochemical assays.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Procesamiento Proteico-Postraduccional , Protrombina/aislamiento & purificación , Espectrometría de Masas en Tándem/métodos , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión/instrumentación , Humanos , Datos de Secuencia Molecular , Protrombina/química , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Espectrometría de Masas en Tándem/instrumentación
16.
RNA ; 17(9): 1688-96, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21752927

RESUMEN

Selective 2'-hydroxyl acylation analyzed by primer extension (SHAPE) is a facile technique for quantitative analysis of RNA secondary structure. In general, low SHAPE signal values indicate Watson-Crick base-pairing, and high values indicate positions that are single-stranded within the RNA structure. However, the relationship of SHAPE signals to structural properties such as non-Watson-Crick base-pairing or stacking has thus far not been thoroughly investigated. Here, we present results of SHAPE experiments performed on several RNAs with published three-dimensional structures. This strategy allows us to analyze the results in terms of correlations between chemical reactivities and structural properties of the respective nucleotide, such as different types of base-pairing, stacking, and phosphate-backbone interactions. We find that the RNA SHAPE signal is strongly correlated with cis-Watson-Crick/Watson-Crick base-pairing and is to a remarkable degree not dependent on other structural properties with the exception of stacking. We subsequently generated probabilistic models that estimate the likelihood that a residue with a given SHAPE score participates in base-pairing. We show that several models that take SHAPE scores of adjacent residues into account perform better in predicting base-pairing compared with individual SHAPE scores. This underscores the context sensitivity of SHAPE and provides a framework for an improved interpretation of the response of RNA to chemical modification.


Asunto(s)
Conformación de Ácido Nucleico , ARN/química , ARN/genética , Acilación , Secuencias de Aminoácidos , Emparejamiento Base , Cartilla de ADN , Electroforesis , Modelos Moleculares , Nucleótidos/genética , Análisis de Secuencia de ARN , Transcripción Genética
17.
Antimicrob Agents Chemother ; 54(9): 3913-21, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20547794

RESUMEN

Vinylogous ureas 2-amino-5,6,7,8-tetrahydro-4H-cyclohepta[b]thiophene-3-carboxamide and N-[3-(aminocarbonyl)-4,5-dimethyl-2-thienyl]-2-furancarboxamide (compounds 1 and 2, respectively) were recently identified to be modestly potent inhibitors of the RNase H activity of HIV-1 and HIV-2 reverse transcriptase (RT). Both compounds shared a 3-CONH(2)-substituted thiophene ring but were otherwise structurally unrelated, which prevented a precise definition of the pharmacophore. We have therefore examined a larger series of vinylogous ureas carrying amide, amine, and cycloalkane modifications of the thiophene ring of compound 1. While cycloheptane- and cyclohexane-substituted derivatives retained potency, cyclopentane and cyclooctane substitutions eliminated activity. In the presence of a cycloheptane ring, modifying the 2-NH(2) or 3-CONH(2) functions decreased the potency. With respect to compound 2, vinylogous ureas whose dimethylthiophene ring contained modifications of the 2-NH(2) and 3-CONH(2) functions were investigated. 2-NH(2)-modified analogs displayed potency equivalent to or enhanced over that of compound 2, the most active of which, compound 16, reflected intramolecular cyclization of the 2-NH(2) and 3-CONH(2) groups. Molecular modeling was used to define an inhibitor binding site in the p51 thumb subdomain, suggesting that an interaction with the catalytically conserved His539 of the p66 RNase H domain could underlie inhibition of RNase H activity. Collectively, our data indicate that multiple functional groups of vinylogous ureas contribute to their potencies as RNase H inhibitors. Finally, single-molecule spectroscopy indicates that vinylogous ureas have the property of altering the reverse transcriptase orientation on a model RNA-DNA hybrid mimicking initiation plus-strand DNA synthesis.


Asunto(s)
Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Ribonucleasa H del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Ribonucleasa H del Virus de la Inmunodeficiencia Humana/química , Dominio Catalítico , Humanos , Modelos Moleculares , Estructura Molecular , Temperatura , Termodinámica
18.
J Mol Biol ; 388(3): 462-74, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19289131

RESUMEN

Reverse transcriptase of the human immunodeficiency virus possesses DNA polymerase and ribonuclease (RNase) H activities. Although the nucleic acid binding cleft separating these domains can accommodate structurally diverse duplexes, it is currently unknown whether regular DNA/RNA hybrids can simultaneously contact both active sites. In this study, we demonstrate that ligands capable of trapping the 3'-end of the primer at the polymerase active site affect the specificity of RNase H cleavage without altering the efficiency of the reaction. Experiments under single-turnover conditions reveal that complexes with a bound nucleotide substrate show specific RNase H cleavage at template position -18, while complexes with the pyrophosphate analogue foscarnet show a specific cut at position -19. This pattern is indicative of post-translocated and pre-translocated conformations. The data are inconsistent with models postulating that the substrate toggles between both active sites, such that the primer 3'-terminus is disengaged from the polymerase active site when the template is in contact with the RNase H active site. In contrast, our findings provide strong evidence to suggest that the nucleic acid substrate can engage both active sites at the same time. As a consequence, the bound and intact DNA/RNA hybrid can restrict access of RNase H active site inhibitors. We have mapped the binding site of the recently discovered inhibitor beta-thujaplicinol between the RNase H active site and Y501 of the RNase H primer grip, and have shown that the inhibitor is unable to bind to a preformed reverse transcriptase-DNA/RNA complex. In conclusion, the bound nucleic acid substrate and in turn, active DNA synthesis can represent an obstacle to RNase H inhibition with compounds that bind to the RNase H active site.


Asunto(s)
ADN Viral/metabolismo , Transcriptasa Inversa del VIH/metabolismo , VIH-1/fisiología , ARN Viral/metabolismo , Dominio Catalítico , Unión Proteica
19.
Glycoconj J ; 26(8): 945-52, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18473163

RESUMEN

beta-Hexosaminidases (EC 3.2.1.52) are lysosomal enzymes that remove terminal beta-glycosidically bound N-acetylglucosamine and N-acetylgalactosamine residues from a number of glycoconjugates. Reliable assay systems are particularly important for the diagnosis of a family of lysosomal storage disorders, the GM2 gangliosidoses that result from inherited beta-hexosaminidase deficiency. More recently, aberrant hexosaminidase levels have also been found to be associated with a variety of inflammatory diseases. Apart from patient testing and carrier screening, practical in vitro assays are indispensable for the characterization of knock-out mice with potentially altered hexosaminidase activities, for detailed structure-function studies aimed at elucidating the enzymatic mechanism, and to characterize newly described enzyme variants from other organisms. The purpose of this article is to discuss convenient hexosaminidase assay procedures for these and other applications, using fluorogenic or chromogenic artificial substrates as well as the physiological glycolipid substrate GM2. Attempts are also made to provide an overview of less commonly used alternative techniques and to introduce recent developments enabling high-throughput screening for enzyme inhibitors.


Asunto(s)
Pruebas de Enzimas/métodos , Hexosaminidasas/metabolismo , Animales , Conformación de Carbohidratos , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes/metabolismo , Gangliósido G(M2)/química , Gangliósido G(M2)/metabolismo , Hexosaminidasas/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento , Humanos , Liposomas/metabolismo , Ratones , Micelas , Especificidad por Sustrato/efectos de los fármacos
20.
HIV Ther ; 3(1): 39-53, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-38961883

RESUMEN

Members of the ribonuclease H (RNase H) family of enzymes (EC 3.1.26.4), which are found in nearly all organisms, are endoribonucleases that specifically hydrolyze the phosphodiester bond of RNA in a RNA-DNA hybrid. In retroviruses such as HIV-1, the RNase H activity is part of reverse transcriptase, the enzyme that converts the viral ssRNA into dsDNA suitable for integration into the host cell genome. In HIV-1, RNase H plays an essential role in various stages of reverse transcription, and it has been known for 20 years that inhibiting RNase H activity renders HIV noninfectious. However, the development of potent and selective antagonists of HIV RNase H has made surprisingly slow progress, and so far no RNase H inhibitor is in clinical trial, rendering this enzyme an important, but as yet underexplored, drug target. The recently described crystal structure of human RNase H in complex with a RNA-DNA hybrid provides new insight into the mechanism of HIV RNase H activity, with the potential to unveil new niches for therapeutic intervention. The current status of RNase H screening efforts is reviewed here.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...