Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
PDA J Pharm Sci Technol ; 77(3): 197-210, 2023.
Article in English | MEDLINE | ID: mdl-36241213

ABSTRACT

Container closure systems that are used for packaging pharmaceutical products are required to satisfy numerous safety requirements. Maximum permitted limits on the concentrations of numerous toxic elemental impurities that potentially leach from the packaging are one such requirement. The implementation of ICH-Q3D Guideline for Elemental Impurities, in conjunction with the 2018 publication of USP <232> Elemental Impurities-Limits and USP <233> Elemental Impurities-Procedures, requires a critical risk assessment of all container closure systems to evaluate their contribution of certain elemental impurities to the enclosed drug product. ICH-Q3D has established limits for each specific elemental impurity that considers relevant toxicological data and administration route (oral, parenteral, or inhalation) and presents them as permitted daily exposures based on the maximum daily dosage of the final drug product. A study was undertaken to assess the degree of elemental impurity leaching from one type of pharmaceutical glass vial under specific, fixed environmental controls. Multiple buffer systems representing a broad spectrum of possible parenteral drug product formulations were used in the study. Resulting buffer solutions that had been in contact with a single type of glass vial under specific conditions were subsequently analyzed using an inductively coupled plasma mass spectrometry (ICP-MS) method developed and validated specifically for the purpose of quantifying elemental impurity leachables in a variety of parenteral formulations. Results indicated that the degree of elemental impurity leachables imparted by the specific type of glass vial evaluated during this study posed no risk to patient safety, regardless of the drug product buffer formulation. Following this evaluation, the ICP-MS method developed for the determination of elemental impurities leachables has been successfully applied to the assessment of elemental impurities in a number of different biological parenteral drug product formulations currently under development. These data can be leveraged for inclusion in elemental impurities component ICH-Q3D risk assessments to satisfy the container closure system contribution.


Subject(s)
Drug Contamination , Drug Packaging , Humans , Pharmaceutical Preparations , Spectrum Analysis , Risk Assessment/methods , Mass Spectrometry/methods , Drug Contamination/prevention & control
2.
J Pharm Innov ; 15(2)2020 Jun.
Article in English | MEDLINE | ID: mdl-32582399
3.
Biotechnol Bioeng ; 116(10): 2632-2639, 2019 10.
Article in English | MEDLINE | ID: mdl-31286487

ABSTRACT

Histidine is a frequently used buffer in the final formulation of many commercialized monoclonal antibodies (mAbs), with histidine helping to stabilize the antibody during storage in addition to its buffering function. The objective of this study was to examine the stereospecificity of any histidine-antibody interactions using a combination of experimental studies and molecular dynamics simulations. Isothermal titration calorimetry provided evidence of weak stereospecific interactions, with the antibody showing approximately two to four additional interaction sites for d- versus l-histidine. The greater interactions with d-histidine were confirmed by measurements of the net protein charge using electrophoretic light scattering. The reduction in the net negative charge of the antibody in d-histidine led to significantly different behavior during diafiltration due to Donnan exclusion effects. Molecular dynamics simulations corroborated the presence of additional d-histidine interaction sites. These results provide the first demonstration of weak stereospecific interactions between l- and d-histidine and a mAb and the implications of these interactions for antibody formulation.


Subject(s)
Antibodies, Monoclonal/chemistry , Histidine/chemistry , Molecular Dynamics Simulation
4.
J Phys Chem B ; 123(27): 5709-5720, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31241333

ABSTRACT

Nonspecific protein-protein interactions of a monoclonal antibody were quantified experimentally using light scattering from low to high protein concentrations (c2) and compared with prior work for a different antibody that yielded qualitatively different behavior. The c2 dependence of the excess Rayleigh ratio (Rex) provided the osmotic second virial coefficient (B22) at low c2 and the static structure factor (Sq=0) at high c2, as a function of solution pH, total ionic strength (TIS), and sucrose concentration. Net repulsive interactions were observed at pH 5, with weaker repulsions at higher TIS. Conversely, attractive electrostatic interactions were observed at pH 6.5, with weaker attractions at higher TIS. Refined coarse-grained models were used to fit model parameters using experimental B22 versus TIS data. The parameters were used to predict high-c2 Rex values via Monte Carlo simulations and separately with Mayer-sampling calculations of higher-order virial coefficients. For both methods, predictions for repulsive to mildly attractive conditions were quantitatively accurate. However, only qualitatively accurate predictions were practical for strongly attractive conditions. An alternative, higher resolution model was used to show semiquantitatively and quantitatively accurate predictions of strong electrostatic attractions at low c2 and low ionic strength.


Subject(s)
Antibodies, Monoclonal/chemistry , Hydrogen-Ion Concentration , Models, Molecular , Osmolar Concentration , Protein Binding , Solutions
5.
Curr Opin Biotechnol ; 60: 159-167, 2019 12.
Article in English | MEDLINE | ID: mdl-30861476

ABSTRACT

Development of a robust biologic drug product is accomplished by extensive formulation and process development screening studies; however, even in the most optimal formulation, a protein can undergo spontaneous degradation during manufacture, storage, and clinical use. Chemical changes to amino acid residues, such as oxidation of methionine or tryptophan, or changes in charge such as deamidation or carbonylation, can induce conformational changes in the overall protein structure, potentially leading to changes in physical - in addition to chemical - stability. Oxidation is often caused by light exposure or the presence of metal ions or peroxides. Asparagine deamidation is more likely to occur at higher pH and/or elevated temperature. Mechanical and interfacial stresses during manufacturing can lead to physical instabilities (i.e. various forms of aggregation). A well-defined manufacturing process and effective in-process controls are essential in minimizing chemical and physical instabilities, enabling robust production and distribution of a safe and efficacious drug product. In this work, the authors provide a review of developments in these areas over the past two years, with emphasis on manufacturability of therapeutically relevant proteins and protein-based drug products.


Subject(s)
Proteins/chemistry , Amino Acids , Methionine , Oxidation-Reduction
6.
Curr Pharm Biotechnol ; 20(3): 232-244, 2019.
Article in English | MEDLINE | ID: mdl-30767738

ABSTRACT

BACKGROUND: The presence of subvisible particles (SVPs) in parenteral formulations of biologics is a major challenge in the development of therapeutic protein formulations. Distinction between proteinaceous and non-proteinaceous SVPs is vital in monitoring formulation stability. METHODS: The current compendial method based on light obscuration (LO) has limitations in the analysis of translucent/low refractive index particles. A number of attempts have been made to develop an unambiguous method to characterize SVPs, albeit with limited success. RESULTS: Herein, we describe a robust method that characterizes and distinguishes both potentially proteinaceous and non-proteinaceous SVPs in protein formulations using Microflow imaging (MFI) in conjunction with the MVAS software (MFI View Analysis Suite), developed by ProteinSimple. The method utilizes two Intensity parameters and a morphological filter that successfully distinguishes proteinaceous SVPs from non-proteinaceous SVPs and mixed aggregates. CONCLUSION: The MFI generated raw data of a protein sample is processed through Lumetics LINK software that applies an in-house developed filter to separate proteinaceous from the rest of the particulates.


Subject(s)
Biological Products/analysis , Biological Products/chemistry , Particle Size , Protein Aggregates , Discriminant Analysis , Protein Aggregates/physiology
7.
Biophys Chem ; 185: 79-87, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24374388

ABSTRACT

Proper disulfide formation can be essential for the conformational stability of natively folded proteins. For proteins that must unfold in order to aggregate, disruption of native disulfides may therefore promote aggregation. This study characterizes differences in the aggregation process for wild-type (WT) α-chymostrypsinogen A (aCgn) and the same molecule with one of its native disulfides (C191-C220) reduced to free thiols (aCgnSH) at acidic pH, where WT aCgn forms semi-flexible amyloid polymers. Loss of the disulfide leads to no discernable differences in folded monomer secondary or tertiary structure based on circular dichroism (CD) or intrinsic fluorescence (FL), and causes a small decrease in the free energy change upon unfolding. After unfolding-mediated aggregation, the resulting amyloid morphology and structure are similar or indistinguishable for aCgn and aCgnSH by CD, FL, ThT binding, multi-angle laser light scattering, and transmission electron microscopy. Aggregates of aCgn and aCgnSH are also able to cross-seed with monomers of the other species. However, aggregates of aCgnSH are more resistive than aCgn aggregates to urea-mediated dissociation, suggesting some degree of structural differences in the aggregated species that was not resolvable in detail without higher resolution methods. Mechanistic analyses of aggregation kinetics indicate that the initiation or nucleation of new aggregates from aCgnSH involves a mono-molecular rate limiting step, possibly the unfolding step. In contrast, that for aCgn involves an oligomeric intermediate, suggesting native disulfide linkages help to hinder non-native protein aggregation by providing conformational barriers to key nucleation event(s).


Subject(s)
Amyloid/chemistry , Chymotrypsinogen/chemistry , Disulfides/chemistry , Hydrogen-Ion Concentration , Models, Molecular , Oxidation-Reduction , Protein Conformation , Protein Unfolding
8.
J Phys Chem B ; 117(50): 16013-28, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24289039

ABSTRACT

Reversible protein cluster formation is an important initial step in the processes of native and non-native protein aggregation, but involves relatively long time and length scales for detailed atomistic simulations and extensive mapping of free energy landscapes. A coarse-grained (CG) model is presented to semiquantitatively characterize the thermodynamics and key configurations involved in the landscape for protein oligomerization, as well as experimental measures of interactions such as the osmotic second virial coefficient (B22). Based on earlier work (Grüenberger et al., J. Phys. Chem. B 2013, 117, 763), this CG model treats proteins as rigid bodies composed of one bead per amino acid, with each amino acid having specific parameters for its size, hydrophobicity, and charge. The net interactions are a combination of steric repulsions, short-range attractions, and screened long-range charge-charge interactions. Model parametrization was done by fitting simulation results against experimental value of B22 as a function of solution ionic strength for α-chymotrypsinogen A and γD-Crystallin (gD-Crys). The CG model is applied to characterize the pairwise interactions and dimerization of gD-Crys and the dependence on temperature, protein concentration, and ionic strength. The results illustrate that at experimentally relevant conditions where stable dimers do not form, the entropic contributions are predominant in the free-energy of protein cluster formation and colloidal protein interactions, arguing against interpretations that treat B22 primarily from energetic considerations alone. Additionally, the results suggest that electrostatic interactions help to modulate the population of the different stable configurations for protein nearest-neighbor pairs, while short-range attractions determine the relative orientations of proteins within these configurations. Finally, simulation results are combined with Principal Component Analysis to identify those amino-acids/surface patches that form interprotein contacts at conditions that favor dimerization of gD-Crys. The resulting regions agree with previously found aggregation-prone sites, as well as suggesting new ones that may be important.


Subject(s)
Colloids/chemistry , Models, Chemical , Proteins/chemistry , Amino Acid Sequence , Molecular Sequence Data , Osmolar Concentration , Principal Component Analysis , Sequence Homology, Amino Acid , Thermodynamics
9.
AAPS PharmSciTech ; 14(4): 1360-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24002823

ABSTRACT

A high-throughput thermal-scanning method to rank-order formulation conditions for therapeutic proteins is described. Apparent transition temperatures for unfolding and aggregation of four different proteins are determined using the dyes SYPRO Orange and thioflavin T (ThT) under a variety of buffer conditions. The results indicate that the ThT-based thermal scanning method offers several advantages over the previously described SYPRO Orange-based thermal scanning and allows rapid rank ordering of solution conditions relevant toward long-term storage of therapeutic molecules. The method is also amenable to high protein concentration and does not require sample dilution or extensive preparation. Additionally, this parallel use of SYPRO Orange and ThT can be readily applied to the screening of mutants for their unfolding and aggregation propensities.


Subject(s)
Chemistry, Pharmaceutical/methods , High-Throughput Screening Assays/methods , Proteins/chemistry , Algorithms , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Benzothiazoles , Buffers , Chymotrypsinogen , Peptides/administration & dosage , Peptides/chemistry , Protein Conformation , Protein Structure, Secondary , Real-Time Polymerase Chain Reaction , Solubility , Spectrometry, Fluorescence , Temperature , Thiazoles
10.
J Pharm Sci ; 101(10): 3651-60, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22806414

ABSTRACT

Visible and subvisible particle formation during the storage of protein solutions is of increasing concern for pharmaceutical products. Previous work (Li Y, Ogunnaike BA, Roberts CJ. 2010. J Pharm Sci 99:645-662) showed that the model protein, alpha-chymotrypsinogen A (aCgn), forms non-native aggregates under accelerated (heated) conditions, but the size and morphology of the resulting aggregates depended sensitively on pH and NaCl. Here, it is shown that aggregates created as high-molecular-weight soluble aggregates undergo a pH- and salt-dependent reversible phase transition to a condensed or insoluble phase of suspended microparticles, whereas monomers remain completely soluble in the same regime. The location of the phase boundary is quantitatively consistent with the different regimes of kinetic behavior observed previously for aCgn. This suggests that the while kinetics is important for controlling the rates of monomer loss during non-native aggregation, it may be possible to tune solution thermodynamics and phase behavior to suppress otherwise soluble aggregates from propagating to form visible or large subvisible particles. Interestingly, the aggregate phase boundary is sensitive to the identity of salt anions in solution, highlighting the importance of electrostatics and preferential salt interactions in mediating aggregate condensation and particle formation.


Subject(s)
Chymotrypsinogen/chemistry , Sodium Chloride/chemistry , Anions/chemistry , Drug Storage , Hydrogen-Ion Concentration , Kinetics , Phase Transition , Protein Binding , Solutions/chemistry , Thermodynamics
11.
Methods Mol Biol ; 899: 403-23, 2012.
Article in English | MEDLINE | ID: mdl-22735967

ABSTRACT

In this chapter, application of size exclusion chromatography with inline multi-angle light scattering (SEC-MALS) to protein systems is reviewed, in particular for its use in elucidating mechanistic details of net-irreversible aggregation processes. After motivating why SEC-MALS or analogous techniques are natural choices to interrogate such aggregating systems, the individual techniques (SEC and MALS) are reviewed briefly, as needed for the context of the remainder of the chapter. Illustrative examples are provided to highlight when and how SEC-MALS can be applied to test mass-action kinetic models for protein aggregation. Limitations of the technique, as well as recommendations for troubleshooting and potential errors in data interpretation are also provided.


Subject(s)
Chromatography, Gel/methods , Protein Biosynthesis , Proteins/chemistry , Kinetics , Light , Molecular Weight , Proteins/isolation & purification , Scattering, Radiation
12.
J Pharm Sci ; 101(5): 1678-87, 2012 May.
Article in English | MEDLINE | ID: mdl-22246657

ABSTRACT

Monomer unfolding and thermally accelerated aggregation kinetics to produce soluble oligomers or insoluble macroscopic aggregates were characterized as a function of pH for an IgG2 antibody using differential scanning calorimetry (DSC) and size-exclusion chromatography (SEC). Aggregate size was quantified via laser light scattering, and aggregate solubility via turbidity and visual inspection. Interestingly, nonnative oligomers were soluble at pH 5.5 above approximately 15°C, but converted reversibly to visible/insoluble particles at lower temperatures. Lower pH values yielded only soluble aggregates, whereas higher pH resulted in insoluble aggregates, regardless of the solution temperature. Unlike the growing body of literature that supports the three-endotherm model of IgG1 unfolding in DSC, the results here also illustrate limitations of that model for other monoclonal antibodies. Comparison of DSC with monomer loss (via SEC) from samples during thermal scanning indicates that the least conformationally stable domain is not the most aggregation prone, and that a number of the domains remain intact within the constituent monomers of the resulting aggregates. This highlights continued challenges with predicting a priori which domain(s) or thermal transition(s) is(are) most relevant for product stability with respect to aggregation.


Subject(s)
Antibodies/immunology , Hydrogen-Ion Concentration , Immunoglobulin G/immunology , Temperature , Calorimetry, Differential Scanning , Kinetics , Solubility
13.
J Chem Phys ; 134(22): 225103, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21682538

ABSTRACT

The classic analysis of Rayleigh light scattering (LS) is re-examined for multi-component protein solutions, within the context of Kirkwood-Buff (KB) theory as well as a more generalized canonical treatment. Significant differences arise when traditional treatments that approximate constant pressure and neglect concentration fluctuations in one or more (co)solvent/co-solute species are compared with more rigorous treatments at constant volume and with all species free to fluctuate. For dilute solutions, it is shown that LS can be used to rigorously and unambiguously obtain values for the osmotic second virial coefficient (B(22)), in contrast with recent arguments regarding protein interactions deduced from LS experiments. For more concentrated solutions, it is shown that conventional analysis over(under)-estimates the magnitude of B(22) for significantly repulsive(attractive) conditions, and that protein-protein KB integrals (G(22)) are the more relevant quantity obtainable from LS. Published data for α-chymotrypsinogen A and a series of monoclonal antibodies at different pH and salt concentrations are re-analyzed using traditional and new treatments. The results illustrate that while traditional analysis may be sufficient if one is interested in only the sign of B(22) or G(22), the quantitative values can be significantly in error. A simple approach is illustrated for determining whether protein concentration (c(2)) is sufficiently dilute for B(22) to apply, and for correcting B(22) values from traditional LS regression at higher c(2) values. The apparent molecular weight M(2, app) obtained from LS is shown to generally not be equal to the true molecular weight, with the differences arising from a combination of protein-solute and protein-cosolute interactions that may, in principle, also be determined from LS.


Subject(s)
Light , Proteins/chemistry , Protein Binding , Scattering, Radiation , Solutions , Solvents/chemistry , Thermodynamics
14.
Int J Pharm ; 418(2): 318-33, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21497188

ABSTRACT

Non-native aggregation is a common concern during therapeutic protein product development and manufacturing, particularly for liquid dosage forms. Because aggregates are often net irreversible under the conditions that they form, controlling aggregate levels requires control of aggregation rates across a range of solution conditions. Rational design of product formulation(s) would therefore benefit greatly from methods to accurately predict aggregation rates. This article focuses on the principles underlying current rate-prediction approaches for non-native aggregation, the limitations and strengths of different approaches, and illustrative examples from the authors' laboratories. The analysis highlights a number of reasons why accurate prediction of aggregation rates remains an outstanding challenge, and suggests some of the important areas for research to ultimately enable improved predictive capabilities in the future.


Subject(s)
Drug Compounding/methods , Drug Design , Pharmaceutical Preparations/chemistry , Proteins/chemistry , Drug Stability , Electromagnetic Phenomena , Humans , Models, Molecular , Osmosis , Protein Conformation , Protein Unfolding , Solutions/chemistry , Temperature
15.
Biochemistry ; 50(5): 628-39, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21184609

ABSTRACT

γD crystallin is a natively monomeric eye-lens protein that is associated with hereditary juvenile cataract formation. It is an attractive model system as a multidomain Greek-key protein that aggregates through partially folded intermediates. Point mutations M69Q and S130P were used to test (1) whether the protein-design algorithm RosettaDesign would successfully predict mutants that are resistant to aggregation when combined with informatic sequence-based predictors of peptide aggregation propensity and (2) how the mutations affected relative unfolding free energies (ΔΔG(un)) and intrinsic aggregation propensity (IAP). M69Q was predicted to have ΔΔG(un) ≫ 0, without significantly affecting IAP. S130P was predicted to have ΔΔG(un) ∼ 0 but with reduced IAP. The stability, conformation, and aggregation kinetics in acidic solution were experimentally characterized and compared for the variants and wild-type (WT) protein using circular dichroism and intrinsic fluorescence spectroscopy, calorimetric and chemical unfolding, thioflavin-T binding, chromatography, static laser light scattering, and kinetic modeling. Monomer secondary and tertiary structures of both variants were indistinguishable from WT, while ΔΔG(un) > 0 for M69Q and ΔΔG(un) < 0 for S130P. Surprisingly, despite being the least conformationally stable, S130P was the most resistant to aggregation, indicating a significant decrease of its IAP compared to WT and M69Q.


Subject(s)
Point Mutation , gamma-Crystallins/chemistry , gamma-Crystallins/genetics , Amino Acid Sequence , Circular Dichroism , Computer-Aided Design , Humans , Kinetics , Molecular Sequence Data , Protein Conformation , Protein Folding , Protein Stability , Thermodynamics , gamma-Crystallins/metabolism
16.
J Pharm Sci ; 99(12): 4830-48, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20821389

ABSTRACT

Changes in protein-protein interactions, protein unfolding, and nonnative aggregation were assessed for a series of human IgG1 antibodies as a function of pH and solution ionic strength (I). Unfolding transitions were characterized with differential scanning calorimetry. Protein-protein interactions were characterized with the apparent second virial coefficient (A(2)) from light scattering. Aggregation pathways were assessed using size-exclusion chromatography and multi-angle laser light scattering, aggregation kinetics, and structural changes monitored by circular dichroism spectroscopy and thioflavine T (ThT) binding. Ionic strength had relatively minor qualitative effects on unfolding, while pH had large effects for all four antibodies. A(2) was sensitive to both pH and I, and indicated that electrostatic interactions and nonuniform surface-charge distributions were important near neutral pH. Depending on solution pH and I, distinct aggregation pathways were found for each antibody, and these shared similar patterns versus pH, I, and A(2). Main differences observed across different antibodies included thermal unfolding transitions in DSC and the effects of pH and I on aggregation kinetics and pathways. These correlated strongly with whether aggregates of a given antibody bound ThT, suggesting possible differences with respect to conformational changes and/or regions of the proteins that are structurally involved in stabilizing the aggregates.


Subject(s)
Hydrogen-Ion Concentration , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Osmolar Concentration , Protein Unfolding , Benzothiazoles , Calorimetry, Differential Scanning , Chromatography, Gel , Circular Dichroism , Humans , Immunoglobulin Isotypes/chemistry , Kinetics , Lasers , Light , Protein Binding , Protein Conformation , Scattering, Radiation , Solutions , Spectrometry, Fluorescence , Static Electricity , Temperature , Thiazoles/metabolism
17.
Biomacromolecules ; 10(10): 2740-9, 2009 Oct 12.
Article in English | MEDLINE | ID: mdl-19743840

ABSTRACT

The bioconjugation of proteins and peptides with synthetic polymers is a promising method for tailoring the chemical, biological, and physical properties of both the polymeric and protein-based components. Here, we describe macromolecular assemblies of polyethylene glycol-coiled-coil alternating multiblock polymers guided by hetero- and homodimeric association of coiled-coils. High molecular weight, alternating block polymers of polyethylene glycol (PEG) and coiled-coil peptides were formed via facile NHS-activated amide bond formation under strictly anhydrous conditions. Confirmation of multiblock formation was assessed via a combination of NMR spectroscopy, size-exclusion chromatography, and electrophoretic analysis. Formation of the alternating multiblock polymers of PEG with coiled-coil peptides through the f-position on the heptads did not impair the ability of the coiled-coils to form heterodimers, as assessed via circular dichroic spectroscopy. Interestingly, the conjugation triggered homooligomer formation in one of the peptides that is monomeric in the absence of PEG. The macromolecular assembly of the homooligomer was characterized via circular dichroic spectroscopy and analytical ultracentrifugation, as well as via dynamic and static light scattering. The assembled structures formed in phosphate buffered saline even at very dilute concentrations of multiblock polymer and exhibited controlled sizes relevant in applications such as drug delivery and controlled release.


Subject(s)
Biopolymers/chemistry , Peptides/chemistry , Proteins/chemistry , Chromatography, Gel , Circular Dichroism , Nuclear Magnetic Resonance, Biomolecular , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL
...