Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Anal Biochem ; 694: 115617, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39019206

ABSTRACT

Data are presented demonstrating that absorbance detection can be used during high-speed sedimentation velocity analytical ultracentrifugation (hs-SV-AUC) experiments to characterize the size distribution of adeno-associated virus (AAV) drug products accurately. Advantages and limitations of being able to use this detector in this specific type of SV-AUC experiment are discussed.

2.
Anal Biochem ; 689: 115482, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38342199

ABSTRACT

Simulated SV-AUC data for an adeno-associated virus (AAV) sample consisting of four components having closely spaced sedimentation coefficients were used to develop a high-speed protocol that optimized the size distribution analysis resolution. The resulting high speed (45K rpm) SV-AUC (hs-SV-AUC) protocol poses several experimental challenges: 1) the need for rapid data acquisition, 2) increased potential for optical artifacts from steep and fast moving boundaries and 3) the increased potential for convection. To overcome these challenges the protocol uses interference detection at low temperatures and data that are confined to a limited radial-time window. In addition to providing higher resolution AAV SV-AUC data and very short run times (<20 min after temperature equilibration), the need to match the sample and reference solvent composition and meniscus positions is relaxed making interference detection as simple to employ as absorbance detection. Finally, experimental data comparing hs-SV-AUC (at 45K rpm) with standard low-speed (15K rpm) SV-AUC on the same AAV sample demonstrate the size distribution resolution improvement. These experiments also validate the use of a radial-time window and show how quickly data can be acquired using the hs-SV-AUC protocol.


Subject(s)
Cold Temperature , Dependovirus , Dependovirus/genetics , Area Under Curve , Ultracentrifugation/methods , Temperature
3.
Anal Biochem ; 631: 114306, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34274312

ABSTRACT

Analytical ultracentrifugation (AUC) provides the most widely applicable, precise, and accurate means for characterizing solution hydrodynamic and thermodynamic properties. While generally useful, boundary sedimentation velocity AUC (SV-AUC) analysis has become particularly important in assessing protein aggregation, fragmentation and conformational variants in the same solvents used during drug development and production. In early 2017 the only manufacturer of the analytical ultracentrifuge released its newest analytical ultracentrifuge, the Optima, to replace the aging second-generation XLA/I series ultracentrifuges. However, SV-AUC data from four Optima units used in the characterization of adeno-associated virus (AAV) have shown evidence of sample convection. Further investigation reveals this problem arises from the design of the temperature control system, which makes it prone to producing destabilizing temperature-induced density gradients that can lead to density inversions. The problem is intermittent and variable in severity within a given Optima unit and between Optima units. This convection appears to be associated mainly with low rotor speeds and dilute concentration of solvent components, i.e., AAV analysis conditions. Data features diagnostic for this problem and strategies for its elimination or minimization are provided.


Subject(s)
Ultracentrifugation/instrumentation , Artifacts , Buffers , Convection , Dependovirus , Equipment Design , Solvents , Temperature
4.
J Am Soc Mass Spectrom ; 28(5): 840-849, 2017 05.
Article in English | MEDLINE | ID: mdl-28194741

ABSTRACT

Introduction of a chemical change to one or more amino acids in a protein's polypeptide chain can result in various effects on its higher-order structure (HOS) and biophysical behavior (or properties). These effects range from no detectable change to significant structural or conformational alteration that can greatly affect the protein's biophysical properties and its resulting biological function. The ability to reliably detect the absence or presence of such changes is essential to understanding the structure-function relationship in a protein and in the successful commercial development of protein-based drugs (biopharmaceuticals). In this paper, we focus our attention on the latter by specifically elucidating the impact of oxidation on the HOS, structural dynamics, and biophysical properties of interferon beta-1a (IFNß-1a). Oxidation is a common biochemical modification that occurs in many biopharmaceuticals, specifically in two naturally-occurring sulfur-containing amino acids, methionine and cysteine. To carry out this work, we used combinations of hydrogen peroxide and pH to differentially oxidize IFNß-1a (to focus on only methionine oxidation versus methionine and cysteine oxidation). We then employed several analytical and biophysical techniques to acquire information about the differential impact of these two oxidation scenarios on IFNß-1a. In particular, the use of MS-based techniques, especially HDX-MS, play a dominant role in revealing the differential effects. Graphical Abstract ᅟ.


Subject(s)
Adjuvants, Immunologic/chemistry , Antiviral Agents/chemistry , Cysteine/chemistry , Interferon beta-1a/chemistry , Methionine/chemistry , Animals , CHO Cells , Chromatography, High Pressure Liquid , Cricetulus , Humans , Hydrogen Peroxide/chemistry , Hydrogen-Ion Concentration , Mass Spectrometry , Models, Molecular , Oxidation-Reduction , Peptide Mapping , Protein Conformation
5.
J Pharm Sci ; 102(12): 4315-29, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24136070

ABSTRACT

To determine how structural changes in antibodies are connected with aggregation, the structural areas of an antibody prone to and/or impacted by aggregation must be identified. In this work, the higher-order structure and biophysical properties of two different monoclonal antibody (mAb) monomers were compared with their simplest aggregated form, that is, dimers that naturally occurred during normal production and storage conditions. A combination of hydrogen/deuterium exchange mass spectrometry and other biophysical measurements was used to make the comparison. The results show that the dimerization process for one of the mAb monomers (mAb1) displayed no differences in its deuterium uptake between monomer and dimer forms. However, the other mAb monomer (mAb2) showed subtle changes in hydrogen/deuterium exchange as compared with its dimer form. In this case, differences observed were located in specific functional regions of the CH 2 domain and the hinge region between CH 1 and CH 2 domains. The importance and the implications of these changes on the antibody structure and mechanism of aggregation are discussed.


Subject(s)
Antibodies, Monoclonal/chemistry , Animals , CHO Cells , Calorimetry, Differential Scanning , Cricetulus , Disulfides/analysis , Electrophoresis, Polyacrylamide Gel , Mass Spectrometry , Models, Molecular , Protein Conformation , Protein Multimerization , Protein Stability , Scattering, Small Angle , X-Ray Diffraction
6.
J Biol Chem ; 287(46): 39050-60, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-22977246

ABSTRACT

Synthesis of the leading DNA strand requires the coordinated activity of DNA polymerase and DNA helicase, whereas synthesis of the lagging strand involves interactions of these proteins with DNA primase. We present the first structural model of a bacteriophage T7 DNA helicase-DNA polymerase complex using a combination of small angle x-ray scattering, single-molecule, and biochemical methods. We propose that the protein-protein interface stabilizing the leading strand synthesis involves two distinct interactions: a stable binding of the helicase to the palm domain of the polymerase and an electrostatic binding of the carboxyl-terminal tail of the helicase to a basic patch on the polymerase. DNA primase facilitates binding of DNA helicase to ssDNA and contributes to formation of the DNA helicase-DNA polymerase complex by stabilizing DNA helicase.


Subject(s)
Bacteriophage T7/genetics , DNA Helicases/chemistry , DNA-Directed DNA Polymerase/chemistry , Virus Replication , Catalysis , DNA Replication , DNA, Single-Stranded/genetics , Kinetics , Microscopy, Electron/methods , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Scattering, Radiation , Surface Plasmon Resonance , Ultracentrifugation , Viral Proteins/chemistry , X-Rays
7.
Nat Rev Drug Discov ; 11(7): 527-40, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22743980

ABSTRACT

Biologics such as monoclonal antibodies are much more complex than small-molecule drugs, which raises challenging questions for the development and regulatory evaluation of follow-on versions of such biopharmaceutical products (also known as biosimilars) and their clinical use once patent protection for the pioneering biologic has expired. With the recent introduction of regulatory pathways for follow-on versions of complex biologics, the role of analytical technologies in comparing biosimilars with the corresponding reference product is attracting substantial interest in establishing the development requirements for biosimilars. Here, we discuss the current state of the art in analytical technologies to assess three characteristics of protein biopharmaceuticals that regulatory authorities have identified as being important in development strategies for biosimilars: post-translational modifications, three-dimensional structures and protein aggregation.


Subject(s)
Biosimilar Pharmaceuticals/chemistry , Chemistry Techniques, Analytical/trends , Animals , Biosimilar Pharmaceuticals/therapeutic use , Chemistry Techniques, Analytical/methods , Humans , Peptides/chemistry , Protein Processing, Post-Translational/physiology , Signal Transduction/physiology
8.
J Pharm Sci ; 101(5): 1688-700, 2012 May.
Article in English | MEDLINE | ID: mdl-22271461

ABSTRACT

A long lasting recombinant factor IX -Fc fusion protein (rFIX-Fc) is being developed for the treatment of hemophilia B and is currently in late stage clinical investigation. By limiting injection frequency and maintaining efficacy, rFIX-Fc shows promise as a new therapeutic option for hemophilia B patients. However, before gaining regulatory approval, rFIX-Fc must undergo rigorous analytical and biological testing, in addition to clinical trials. Included in this testing is the need to understand this protein's higher-order structure and dynamics. In this study, we investigated and compared the biophysical properties of rFIX-Fc, rFIX, and Fc using hydrogen/deuterium exchange mass spectrometry and differential scanning calorimetry. Within the limits of these techniques, our results show that structural comparability exists between rFIX and the FIX region of rFIX-Fc. In addition, changes in the structure and dynamics of both proteins, in response to calcium binding, a requirement for FIX function, are also highly comparable. In the case of Fc and Fc region of rFIX-Fc, conformational comparability is also established. These biophysical results further support the conclusion that fusing an immunoglobulin gamma 1 Fc to rFIX does not significantly alter the higher-order structure of FIX or Fc, Ca binding to FIX, or Fc functionality.


Subject(s)
Calcium/chemistry , Factor IX/chemistry , Immunoglobulin Fc Fragments/chemistry , Calorimetry, Differential Scanning , Mass Spectrometry , Protein Conformation
9.
J Pharm Sci ; 100(11): 4597-606, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21678428

ABSTRACT

There is significant scope for more meaningful evaluation of higher-order structure in defining the quality of biopharmaceutical products [Bush L. 2010. Biopharm Int 23(4):14]. We have used isothermal titration calorimetry (ITC) to characterize the Ca(2+) -binding isotherm of a recombinant human factor IX Fc fusion protein (rFIXFc) and the parent recombinant human factor IX molecule (rFIX). Circular dichroism, intrinsic fluorescence, and Fourier transform infrared spectroscopies detected characteristic spectral changes that appear qualitatively consistent with the previously characterized behavior of the factor IX molecule. Sedimentation velocity and dynamic light scattering measurements were recorded in the presence and absence of Ca(2+) over the protein concentration range 1-10 mg/mL. ITC of Ca(2+) binding to rFIXFc reveals a distinctive exothermic-binding isotherm, which is interpreted as consistent with two high-affinity and approximately 14 lower-affinity Ca(2+) sites reported in the literature for human factor IX (Schmidt AE, Bajaj SP. 2003. Trends Cardiovasc Med 13(1):39-45). Analysis of accelerated degradation samples showed significant alterations in Ca(2+) binding, which correlates with significant loss of biopotency and fragmentation by gel chip capillary electrophoresis. Collectively, these data establish a close correspondence in the Ca(2+) -binding characteristics of rFIXFc and its parent rFIX molecule. The utility of ITC to provide a highly pertinent and selective biophysical signature of structure-function for a therapeutic factor protein is discussed.


Subject(s)
Calcium/metabolism , Factor IX/metabolism , Recombinant Fusion Proteins/metabolism , Calorimetry , Chromatography, Gel , Circular Dichroism , Electrophoresis, Capillary , Factor IX/chemistry , Models, Molecular , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemistry , Scattering, Radiation , Spectrometry, Fluorescence , Spectroscopy, Fourier Transform Infrared , Ultracentrifugation
10.
J Pharm Sci ; 100(6): 2071-86, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21491437

ABSTRACT

The function, efficacy, and safety of protein biopharmaceuticals are tied to their three-dimensional structure. The analysis and verification of this higher-order structure are critical in demonstrating manufacturing consistency and in establishing the absence of structural changes in response to changes in production. It is, therefore, essential to have reliable, high-resolution and high sensitivity biophysical tools capable of interrogating protein structure and conformation. Here, we demonstrate the use of hydrogen/deuterium exchange mass spectrometry (H/DX-MS) in biopharmaceutical comparability studies. H/DX-MS measurements can be conducted with good precision, consume only picomoles of protein, interrogate nearly the entire molecule with peptide level resolution, and can be completed in a few days. Structural comparability or lack of comparability was monitored for different preparations of interferon-ß-1a. We present specific graphical formats for the display of H/DX-MS data that aid in rapidly making both the qualitative (visual) and quantitative assessment of comparability. H/DX-MS is capable of making significant contributions in biopharmaceutical characterization by providing more informative and confidant comparability assessments of protein higher-order structures than are currently available within the biopharmaceutical industry.


Subject(s)
Biopharmaceutics/methods , Deuterium Exchange Measurement , Mass Spectrometry , Proteins/chemistry , Technology, Pharmaceutical/methods , Biopharmaceutics/standards , Interferons/chemistry , Models, Molecular , Protein Conformation , Technology, Pharmaceutical/standards
11.
Protein Sci ; 19(5): 954-66, 2010 May.
Article in English | MEDLINE | ID: mdl-20198683

ABSTRACT

Monoclonal antibodies (Mabs) are a favorite drug platform of the biopharmaceutical industry. Currently, over 20 Mabs have been approved and several hundred others are in clinical trials. The anti-LINGO-1 Mab Li33 was selected from a large panel of antibodies by Fab phage display technology based on its extraordinary biological activity in promoting oligodendrocyte differentiation and myelination in vitro and in animal models of remyelination. However, the Li33 Fab had poor solubility when converted into a full antibody in an immunoglobulin G1 framework. A detailed analysis of the biochemical and structural features of the antibody revealed several possible reasons for its propensity to aggregate. Here, we successfully applied three molecular approaches (isotype switching, targeted mutagenesis of complementarity determining region residues, and glycosylation site insertion mutagenesis) to address the solubility problem. Through these efforts we were able to improve the solubility of the Li33 Mab from 0.3 mg/mL to >50 mg/mL and reduce aggregation to an acceptable level. These strategies can be readily applied to other proteins with solubility issues.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin G/chemistry , Membrane Proteins/immunology , Mutagenesis, Site-Directed/methods , Nerve Tissue Proteins/immunology , Protein Engineering/methods , Amino Acid Sequence , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Area Under Curve , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Immunoglobulin Class Switching , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Stability , Solubility , Temperature
12.
Mol Cell Proteomics ; 9(8): 1716-28, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20103567

ABSTRACT

Post-translational modifications (PTMs) can have profound effects on protein structure and protein dynamics and thereby can influence protein function. To understand and connect PTM-induced functional differences with any resulting conformational changes, the conformational changes must be detected and localized to specific parts of the protein. We illustrate these principles here with a study of the functional and conformational changes that accompany modifications to a monoclonal immunoglobulin gamma1 (IgG1) antibody. IgG1s are large and heterogeneous proteins capable of incorporating a multiplicity of PTMs both in vivo and in vitro. For many IgG1s, these PTMs can play a critical role in affecting conformation, biological function, and the ability of the antibody to initiate a potential adverse biological response. We investigated the impact of differential galactosylation, methionine oxidation, and fucosylation on solution conformation using hydrogen/deuterium exchange mass spectrometry and probed the effects of IgG1 binding to the FcgammaRIIIa receptor. The results showed that methionine oxidation and galactosylation both impact IgG1 conformation, whereas fucosylation appears to have little or no impact to the conformation. FcgammaRIIIa binding was strongly influenced by both the glycan structure/composition (namely galactose and fucose) and conformational changes that were induced by some of the modifications.


Subject(s)
Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Protein Processing, Post-Translational , Receptors, Fc/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Deuterium , Glycosylation , Humans , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protons
13.
J Am Soc Mass Spectrom ; 21(3): 323-37, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19963397

ABSTRACT

Mass spectrometry plays a very visible role in biopharmaceutical industry, although its use in development, characterization, and quality control of protein drugs is mostly limited to the analysis of covalent structure (amino acid sequence and post-translational modifications). Despite the centrality of protein conformation to biological activity, stability, and safety of biopharmaceutical products, the expanding arsenal of mass spectrometry-based methods that are currently available to probe higher order structure and conformational dynamics of biopolymers did not, until recently, enjoy much attention in the industry. This is beginning to change as a result of recent work demonstrating the utility of these experimental tools for various aspects of biopharmaceutical product development and manufacturing. In this work, we use a paradigmatic protein drug interferon beta-1a as an example to illustrate the utility of mass spectrometry as a powerful tool not only to assess the integrity of higher order structure of a protein drug, but also to predict consequences of its degradation at a variety of levels.


Subject(s)
Biological Products/analysis , Biological Products/chemistry , Mass Spectrometry/methods , Mass Spectrometry/trends , Protein Conformation , Proteins/analysis , Proteins/chemistry , Alkylation , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/trends , Industry , Technology Transfer , Universities
14.
Anal Chem ; 80(19): 7473-81, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18729476

ABSTRACT

Unlike small-molecule drugs, the conformational properties of protein biopharmaceuticals in solution are influenced by a variety of factors that are not solely defined by their covalent chemical structure. Since the conformation (or higher order structure) of a protein is a major modulator of its biological activity, the ability to detect changes in both the higher order structure and conformational dynamics of a protein, induced by an array of extrinsic factors, is of central importance in producing, purifying, and formulating a commercial biopharmaceutical with consistent therapeutic properties. In this study we demonstrate that two complementary mass spectrometry-based approaches (analysis of ionic charge-state distribution and hydrogen/deuterium exchange) can be a potent tool in monitoring conformational changes in protein biopharmaceuticals. The utility of these approaches is demonstrated by detecting and characterizing conformational changes in the biopharmaceutical product interferon beta-1a (IFN-beta-1a). The protein degradation process was modeled by inducing a single chemical modification of IFN-beta1a (alkylation of its only free cysteine residue with N-ethylmaleimide), which causes significant reduction in its antiviral activity. Analysis of IFN-beta1a ionic charge-state distributions unequivocally reveals a significant decrease of conformational stability in the degraded protein, while hydrogen/deuterium exchange measurements provide a clear indication that the higher order structure is affected well beyond the covalent modification site. Importantly, neither technique required that the location or indeed the nature of the chemical modification be known prior to or elucidated in the process of the analysis. In contrast, application of the standard armamentarium of biophysical tools, which are commonly employed for quality control of protein pharmaceuticals, met with very limited success in detection and characterization of conformational changes in the modified IFN-beta1a. This work highlights the role mass spectrometry can and should play in the biopharmaceutical industry beyond the presently assigned task of primary structure analysis.


Subject(s)
Interferon-beta/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Amino Acid Sequence , Cardiovirus Infections/drug therapy , Cardiovirus Infections/virology , Cell Line, Tumor , Cytopathogenic Effect, Viral/drug effects , Encephalomyocarditis virus/physiology , Ethylmaleimide/analogs & derivatives , Ethylmaleimide/chemistry , Humans , Interferon beta-1a , Interferon-beta/analysis , Interferon-beta/pharmacology , Lung/drug effects , Lung/virology , Models, Molecular , Molecular Sequence Data , Protein Conformation , Recombinant Proteins/analysis , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
15.
Anal Biochem ; 380(1): 152-4, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18539129

ABSTRACT

A concentration of 6.4 +/- 0.2 (at 1 SD) x 10(11) virus particles (vp) per milliliter was determined for the adenovirus reference material (ARM) by averaging analytical ultracentrifugation concentration data obtained with refractometric detection with the completely independent concentration reported by the Adenoviral Reference Material Working Group (ARMWG). Using this concentration, the ARM absorptivity factor (in sodium dodecyl sulfate [SDS] at 260 nm) of 1.2+/-0.1 (at 1 SD) x 10(12) vp/ml per OD(260 nm) per centimeter was obtained.


Subject(s)
Adenoviridae , Sodium Dodecyl Sulfate/chemistry , Ultracentrifugation/standards , Absorption , Reference Standards , Refractometry
16.
Anal Biochem ; 362(1): 16-37, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17223062

ABSTRACT

This study explores the capability of modern analytical ultracentrifugation (AUC) to characterize the homogeneity, under product formulation conditions, of preparations of adenovirus vectors used in gene therapy and to assess the lot-to-lot consistency of this unique drug product. We demonstrate that a single sedimentation velocity run on an adenovirus sample can detect and accurately quantify a number of different forms of virus particles and subvirus particles. These forms include (a) intact virus monomer particles, (b) virus aggregates, (c) empty capsids (ECs), and (d) smaller assembly intermediates or subparticles formed during normal or aberrant virus assembly (or as a result of damage to the intact adenovirus or EC material during all phases of virus production). This information, which is collected on adenovirus samples under the exact formulation conditions that exist in the adenovirus vial, is obtained by direct boundary modeling of the AUC data generated from refractometric and/or UV detection systems using the computer program SEDFIT developed by Peter Schuck. Although both detectors are useful, refractometric detection using the Rayleigh interferometer offers a key advantage for providing accurate concentration information due to the similar response factors for both protein and DNA and its insensitivity to light scattering effects. Additional AUC data obtained from analytical band sedimentation velocity and density gradient sedimentation equilibrium experiments in CsCl with UV detection were also generated. These results further support conclusions concerning the solution properties of adenovirus, the identity of the different virus species, and the overall capability of boundary sedimentation velocity analysis.


Subject(s)
Adenoviridae/isolation & purification , Genetic Vectors/isolation & purification , Ultracentrifugation/methods , Adenoviridae/genetics , Cesium , Chlorides , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Kinetics
17.
AAPS J ; 8(3): E590-605, 2006 Sep 22.
Article in English | MEDLINE | ID: mdl-17025277

ABSTRACT

In developing and manufacturing protein biopharmaceuticals, aggregation is a parameter that needs careful monitoring to ensure the quality and consistency of the final biopharmaceutical drug product. The analytical method of choice used to perform this task is size-exclusion chromatography (SEC). However, it is becoming more and more apparent that considerable care is required in assessing the accuracy of SEC data. One old analytical tool that is now reappearing to help in this assessment is analytical ultracentrifugation (AUC). Developments in AUC hardware and, more importantly, recent developments in AUC data analysis computer programs have converged to provide this old biophysical tool with the ability to extract very high resolution size information about the molecules in a given sample from a simple sedimentation velocity experiment. In addition, AUC allows sample testing to be conducted in the exact or nearly exact liquid formulation or reconstituted liquid formulation of the biopharmaceutical in the vial, with minimal surface area contact with extraneous materials. As a result, AUC analysis can provide detailed information on the aggregation of a biopharmaceutical, while avoiding many of the major problems that can plague SEC, thus allowing AUC to be used as an orthogonal method to verify SEC aggregation information and the associating properties of biopharmaceuticals.


Subject(s)
Biopharmaceutics , Proteins/chemistry , Ultracentrifugation/methods , Chromatography, Gel/methods , Molecular Weight
18.
J Chromatogr A ; 1079(1-2): 254-65, 2005 Jun 24.
Article in English | MEDLINE | ID: mdl-16038312

ABSTRACT

A simple high-resolution capillary zone electrophoresis (CZE) method capable of rapidly assessing the micro-heterogeneity of a 24 kDa molecular weight glycoprotein, has been developed. Separation is carried out using a bare silica capillary at a pH of 2.5 in a commercially available electrophoresis buffer system composed of triethanolamine and phosphoric acid. Over 30 peaks were detected within a run time of 15 min using a 27 cm capillary and approximately 60 peaks were detected using a 77 cm capillary. Although most of the peaks arise from differences in the oligosaccharide structures present on the one glycosylation site on this molecule, other forms of micro-heterogeneity due to the presence of the nonglycosylated form of this glycoprotein and various types of chemical degradation, e.g., deamidation, are also responsible for the multitude of peaks observed. Although the exact chemical identity of each peak in the resulting electropherogram of this glycoprotein is not known, useful information can be obtained for assessing comparability, stability, and batch consistency. Factors impacting the resolution, precision, accuracy, and robustness of the assay are also discussed along with inherent advantages and limitations associated with measuring the micro-heterogeneity of intact glycoproteins.


Subject(s)
Electrophoresis, Capillary/methods , Glycoproteins/analysis , Antiporters , Buffers , Ethanolamines/chemistry , Glycoproteins/isolation & purification , Glycoproteins/metabolism , Glycosylation , Hydrogen-Ion Concentration , Monitoring, Physiologic , Neuraminidase/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Phosphoric Acids/chemistry , Recombinant Proteins/analysis , Reproducibility of Results , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...