1
|
Bansal R, Jha SK, Jha NK. Size-based Degradation of Therapeutic Proteins - Mechanisms, Modelling and Control. Biomol Concepts 2021; 12:68-84. [PMID: 34146465 DOI: 10.1515/bmc-2021-0008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/07/2021] [Indexed: 02/02/2023] Open
Abstract
Protein therapeutics are in great demand due to their effectiveness towards hard-to-treat diseases. Despite their high demand, these bio-therapeutics are very susceptible to degradation via aggregation, fragmentation, oxidation, and reduction, all of which are very likely to affect the quality and efficacy of the product. Mechanisms and modelling of these degradation (aggregation and fragmentation) pathways is critical for gaining a deeper understanding of stability of these products. This review aims to provide a summary of major developments that have occurred towards unravelling the mechanisms of size-based protein degradation (particularly aggregation and fragmentation), modelling of these size-based degradation pathways, and their control. Major caveats that remain in our understanding and control of size-based protein degradation have also been presented and discussed.
Collapse
Affiliation(s)
- Rohit Bansal
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
2
|
Particle Detection and Characterization for Biopharmaceutical Applications: Current Principles of Established and Alternative Techniques. Pharmaceutics 2020; 12:pharmaceutics12111112. [PMID: 33228023 PMCID: PMC7699340 DOI: 10.3390/pharmaceutics12111112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022] Open
Abstract
Detection and characterization of particles in the visible and subvisible size range is critical in many fields of industrial research. Commercial particle analysis systems have proliferated over the last decade. Despite that growth, most systems continue to be based on well-established principles, and only a handful of new approaches have emerged. Identifying the right particle-analysis approach remains a challenge in research and development. The choice depends on each individual application, the sample, and the information the operator needs to obtain. In biopharmaceutical applications, particle analysis decisions must take product safety, product quality, and regulatory requirements into account. Biopharmaceutical process samples and formulations are dynamic, polydisperse, and very susceptible to chemical and physical degradation: improperly handled product can degrade, becoming inactive or in specific cases immunogenic. This article reviews current methods for detecting, analyzing, and characterizing particles in the biopharmaceutical context. The first part of our article represents an overview about current particle detection and characterization principles, which are in part the base of the emerging techniques. It is very important to understand the measuring principle, in order to be adequately able to judge the outcome of the used assay. Typical principles used in all application fields, including particle–light interactions, the Coulter principle, suspended microchannel resonators, sedimentation processes, and further separation principles, are summarized to illustrate their potentials and limitations considering the investigated samples. In the second part, we describe potential technical approaches for biopharmaceutical particle analysis as some promising techniques, such as nanoparticle tracking analysis (NTA), micro flow imaging (MFI), tunable resistive pulse sensing (TRPS), flow cytometry, and the space- and time-resolved extinction profile (STEP®) technology.
Collapse
|
3
|
Majewski J, Jones EM, Vander Zanden CM, Biernat J, Mandelkow E, Chi EY. Lipid membrane templated misfolding and self-assembly of intrinsically disordered tau protein. Sci Rep 2020; 10:13324. [PMID: 32770092 PMCID: PMC7414892 DOI: 10.1038/s41598-020-70208-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/21/2020] [Indexed: 01/21/2023] Open
Abstract
The aggregation of the intrinsically disordered tau protein into highly ordered β-sheet-rich fibrils is implicated in the pathogenesis of a range of neurodegenerative disorders. The mechanism of tau fibrillogenesis remains unresolved, particularly early events that trigger the misfolding and assembly of the otherwise soluble and stable tau. We investigated the role the lipid membrane plays in modulating the aggregation of three tau variants, the largest isoform hTau40, the truncated construct K18, and a hyperphosphorylation-mimicking mutant hTau40/3Epi. Despite being charged and soluble, the tau proteins were also highly surface active and favorably interacted with anionic lipid monolayers at the air/water interface. Membrane binding of tau also led to the formation of a macroscopic, gelatinous layer at the air/water interface, possibly related to tau phase separation. At the molecular level, tau assembled into oligomers composed of ~ 40 proteins misfolded in a β-sheet conformation at the membrane surface, as detected by in situ synchrotron grazing-incidence X-ray diffraction. Concomitantly, membrane morphology and lipid packing became disrupted. Our findings support a general tau aggregation mechanism wherein tau’s inherent surface activity and favorable interactions with anionic lipids drive tau-membrane association, inducing misfolding and self-assembly of the disordered tau into β-sheet-rich oligomers that subsequently seed fibrillation and deposition into diseased tissues.
Collapse
Affiliation(s)
- Jaroslaw Majewski
- Division of Molecular and Cellular Biology, National Science Foundation, Alexandria, VA, 22314, USA.,Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, NM, 87131, USA.,Theoretical Biology and Biophysics Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Emmalee M Jones
- Nanoscience and Microsystems Engineering Graduate Program, University of New Mexico, Albuquerque, NM, 87131, USA.,Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Crystal M Vander Zanden
- Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, NM, 87131, USA.,Department of Chemistry and Biochemistry, University of Colorado at Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Jacek Biernat
- Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany.,CAESAR Research Center, 53175, Bonn, Germany
| | - Eckhard Mandelkow
- Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany.,CAESAR Research Center, 53175, Bonn, Germany
| | - Eva Y Chi
- Department of Chemical and Biological Engineering and Center for Biomedical Engineering, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
4
|
FTIR Spectroscopy Study of the Secondary Structure Changes in Human Serum Albumin and Trypsin under Neutral Salts. Biomolecules 2020; 10:biom10040606. [PMID: 32295172 PMCID: PMC7226448 DOI: 10.3390/biom10040606] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/27/2020] [Accepted: 03/30/2020] [Indexed: 11/24/2022] Open
Abstract
The effect of neutral salts on protein conformation was first analyzed by Hofmeister in 1888, however, even today this phenomenon is not completely understood. To clarify this effect, we studied changes in the secondary structure of two proteins: human serum albumin with predominantly α-helical structure and porcine pancreas β-trypsin with the typical β-structural arrangement in aqueous solutions of neutral salts (KSCN, KCl, (NH4)2SO4). The changes in the secondary structure were studied at 23 °C and 80 °C by using the second derivative deconvolution method of the IR spectra. Our results demonstrated that the ability of the salts to stabilize/destabilize these two proteins correlates with the Hofmeister series of ions. At the same time, some exceptions were also observed. The destabilization of the native structures of both α-helical albumin and β-structural trypsin upon interaction with neutral salts leads to the formation of intermolecular β-sheets typical for amyloid fibrils or amorphous aggregates. Thus, our quantitative FTIR-spectroscopy analysis allowed us to further clarify the mechanisms and complexity of the neutral salt actions on protein structures which may lead to strategies preventing unwelcome misfolding of proteins.
Collapse
|
5
|
Bansal R, Srivastava P, Rathore AS, Chokshi P. Population balance modelling of aggregation of monoclonal antibody based therapeutic proteins. Chem Eng Sci 2020. [DOI: 10.1016/j.ces.2020.115479] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
6
|
Kendrick BS, Chi EY. Professors John F. Carpenter and Theodore W. Randolph: 2 Giants With a Special Synergy in the Field of Biopharmaceutical Science and Engineering. J Pharm Sci 2020; 109:2-5. [DOI: 10.1016/j.xphs.2019.10.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 11/26/2022]
|
7
|
Katyal N, Deep S. A computational approach to get insights into multiple faces of additives in modulation of protein aggregation pathways. Phys Chem Chem Phys 2019; 21:24269-24285. [PMID: 31670327 DOI: 10.1039/c9cp03763b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An enormous population worldwide is presently confronted with debilitating neurodegenerative diseases. The etiology of the disease is connected to protein aggregation and the events involved therein. Thus, a complete understanding of an inhibitor at different stages in the process is imperative for the formulation of a drug molecule. This review presents a detailed summary of the current status of different cosolvents. It further develops how the complex aggregation pathway can be simplified into three steps common to all proteins and the way computer simulations can be exploited to gain insights into the ways by which known inhibitors can affect all these stages. Computation of theoretical parameters in this regard and their correlation with experimental techniques is accentuated. In addition to providing an outline of the scope of different additives, this review showcases the way by which the problem of analyzing an effect of an additive can be addressed effectively via MD simulations.
Collapse
Affiliation(s)
- Nidhi Katyal
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, Delhi, India.
| | | |
Collapse
|
8
|
Jin W, Xing Z, Song Y, Huang C, Xu X, Ghose S, Li ZJ. Protein aggregation and mitigation strategy in low pH viral inactivation for monoclonal antibody purification. MAbs 2019; 11:1479-1491. [PMID: 31441367 PMCID: PMC6816434 DOI: 10.1080/19420862.2019.1658493] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/01/2019] [Accepted: 08/17/2019] [Indexed: 01/27/2023] Open
Abstract
Significant amounts of soluble product aggregates were observed during low-pH viral inactivation (VI) scale-up for an IgG4 monoclonal antibody (mAb IgG4-N1), while small-scale experiments in the same condition showed negligible aggregation. Poor mixing and product exposure to low pH were identified as the root cause. To gain a mechanistic understanding of the problem, protein aggregation properties were studied by varying critical parameters including pH, hold time and protein concentration. Comprehensive biophysical characterization of product monomers and aggregates was performed using fluorescence-size-exclusion chromatography, differential scanning fluorimetry, fluorescence spectroscopy, and dynamic light scattering. Results showed IgG4-N1 partially unfolds at about pH 3.3 where the product molecules still exist largely as monomers owing to strong inter-molecular repulsions and favorable colloidal stability. In the subsequent neutralization step, however, the conformationally changed monomers are prone to aggregation due to weaker inter-molecular repulsions following the pH transition from 3.3 to 5.5. Surface charge calculations using homology modeling suggested that intra-molecular repulsions, especially between CH2 domains, may contribute to the IgG4-N1 unfolding at ≤ pH 3.3. Computational fluid dynamics (CFD) modeling was employed to simulate the conditions of pH titration to reduce the risk of aggregate formation. The low-pH zones during acid addition were characterized using CFD modeling and correlated to the condition causing severe product aggregation. The CFD tool integrated with the mAb solution properties was used to optimize the VI operating parameters for successful scale-up demonstration. Our research revealed the governing aggregation mechanism for IgG4-N1 under acidic conditions by linking its molecular properties and various process-related parameters to macroscopic aggregation phenomena. This study also provides useful insights into the cause and mitigation of low-pH-induced IgG4 aggregation in downstream VI operation.
Collapse
Affiliation(s)
- Weixin Jin
- Biologics Process Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, MA, USA
| | - Zizhuo Xing
- Biologics Process Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, MA, USA
| | - Yuanli Song
- Biologics Process Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, MA, USA
| | - Chao Huang
- Biologics Process Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, MA, USA
| | - Xuankuo Xu
- Biologics Process Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, MA, USA
| | - Sanchayita Ghose
- Biologics Process Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, MA, USA
| | - Zheng Jian Li
- Biologics Process Development, Global Product Development and Supply, Bristol-Myers Squibb, Devens, MA, USA
| |
Collapse
|
9
|
Wälchli R, Vermeire PJ, Massant J, Arosio P. Accelerated Aggregation Studies of Monoclonal Antibodies: Considerations for Storage Stability. J Pharm Sci 2019; 109:595-602. [PMID: 31676272 DOI: 10.1016/j.xphs.2019.10.048] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/06/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022]
Abstract
Aggregation of mAbs is a crucial concern with respect to their safety and efficacy. Among the various properties of protein aggregates, it is emerging that their size can potentially impact their immunogenicity. Therefore, stability studies of antibody formulations should not only evaluate the rate of monomer loss but also determine the size distribution of the protein aggregates, which in turn depends on the aggregation mechanism. Here, we study the aggregation behavior of different formulations of 2 monoclonal immunoglobulins (IgGs) in the temperature range from 5°C to 50°C over 52 weeks of storage. We show that the aggregation kinetics of both antibodies follow non-Arrhenius behavior and that the aggregation mechanisms change between 40°C and 5°C, leading to different types of aggregates. Specifically, for a given monomer conversion, dimer formation dominates at low temperatures, while larger aggregates are formed at higher temperatures. We further show that the stability ranking of different molecules as well as of different formulations is drastically different at 40°C and 5°C while it correlates better between 30°C and 5°C. Our findings have implications for the level of information provided by accelerated aggregation studies with respect to protein stability under storage conditions.
Collapse
Affiliation(s)
- Ruben Wälchli
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
| | - Pieter-Jan Vermeire
- UCB Pharma, BioTech Sciences, Formulation Development, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| | - Jan Massant
- UCB Pharma, BioTech Sciences, Formulation Development, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland.
| |
Collapse
|
10
|
Yang H, Wang M, Zhang Y, Liu X, Yu S, Guo Y, Yang S, Yang L. Detailed insight into the formation of protein corona: Conformational change, stability and aggregation. Int J Biol Macromol 2019; 135:1114-1122. [DOI: 10.1016/j.ijbiomac.2019.06.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/25/2019] [Accepted: 06/03/2019] [Indexed: 01/22/2023]
|
11
|
Dao Y, Han L, Wang H, Dong S. α-Selective Lysine Ligation and Application in Chemical Synthesis of Interferon Gamma. Org Lett 2019; 21:3265-3270. [DOI: 10.1021/acs.orglett.9b00980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Yuankun Dao
- State Key Laboratory of Natural and Biomimetic Drugs, and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lin Han
- State Key Laboratory of Natural and Biomimetic Drugs, and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hanxuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs, and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Suwei Dong
- State Key Laboratory of Natural and Biomimetic Drugs, and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
12
|
Evers A, Pfeiffer-Marek S, Bossart M, Heubel C, Stock U, Tiwari G, Gebauer B, Elshorst B, Pfenninger A, Lukasczyk U, Hessler G, Kamm W, Wagner M. Peptide Optimization at the Drug Discovery-Development Interface: Tailoring of Physicochemical Properties Toward Specific Formulation Requirements. J Pharm Sci 2019; 108:1404-1414. [DOI: 10.1016/j.xphs.2018.11.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/27/2018] [Indexed: 12/31/2022]
|
13
|
Gandhi AV, Randolph TW, Carpenter JF. Conjugation of Emtansine Onto Trastuzumab Promotes Aggregation of the Antibody-Drug Conjugate by Reducing Repulsive Electrostatic Interactions and Increasing Hydrophobic Interactions. J Pharm Sci 2019; 108:1973-1983. [PMID: 30735687 DOI: 10.1016/j.xphs.2019.01.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/27/2019] [Accepted: 01/31/2019] [Indexed: 12/31/2022]
Abstract
The impact of drug conjugation on intra- and intermolecular interactions of trastuzumab (TmAb) was determined by comparing the conformational and colloidal stabilities of TmAb and trastuzumab emtansine (T-DM1). In low ionic strength formulations, drug conjugation to native lysine residues of TmAb significantly reduced the repulsive electrostatic interactions between T-DM1 molecules. When these electrostatic interactions were screened in solutions with high ionic strength, intermolecular interactions between T-DM1 molecules were found to be more attractive than those between TmAb molecules. Drug conjugation lowered the colloidal stability of T-DM1 compared to TmAb, making T-DM1 more susceptible to agitation-induced aggregation. The presence of polysorbate-20 in the formulations inhibited aggregation of TmAb and T-DM1 induced by the hydrophobic air-water interface. Furthermore, the effect of increased hydrophobic interactions between T-DM1 molecules was studied by monitoring aggregation in TmAb and T-DM1 solutions that were incubated at 4°C, 25°C, and 50°C. Conjugating DM1 to TmAb increased the hydrophobicity of the molecule, and faster aggregation of T-DM1 at 50°C could be attributed to a temperature-dependent increase in hydrophobic interactions between T-DM1 molecules.
Collapse
Affiliation(s)
- Aditya V Gandhi
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology, University of Colorado, Aurora, Colorado 80045
| | - Theodore W Randolph
- Department of Chemical and Biological Engineering, Center for Pharmaceutical Biotechnology, University of Colorado Boulder, Boulder, Colorado 80309
| | - John F Carpenter
- Department of Pharmaceutical Sciences, Center for Pharmaceutical Biotechnology, University of Colorado, Aurora, Colorado 80045.
| |
Collapse
|
14
|
Gandhi AV, Arlotta KJ, Chen HN, Owen SC, Carpenter JF. Biophysical Properties and Heating-Induced Aggregation of Lysine-Conjugated Antibody-Drug Conjugates. J Pharm Sci 2018; 107:1858-1869. [DOI: 10.1016/j.xphs.2018.03.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/17/2018] [Accepted: 03/27/2018] [Indexed: 12/12/2022]
|
15
|
Torelli F, Zander S, Ellerbrok H, Kochs G, Ulrich RG, Klotz C, Seeber F. Recombinant IFN-γ from the bank vole Myodes glareolus: a novel tool for research on rodent reservoirs of zoonotic pathogens. Sci Rep 2018; 8:2797. [PMID: 29434310 PMCID: PMC5809609 DOI: 10.1038/s41598-018-21143-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/25/2018] [Indexed: 12/28/2022] Open
Abstract
Rodent species like Myodes glareolus and Microtus spp. are natural reservoirs for many zoonotic pathogens causing human diseases and are gaining increasing interest in the field of eco-immunology as candidate animal models. Despite their importance the lack of immunological reagents has hampered research in these animal species. Here we report the recombinant production and functional characterization of IFN-γ, a central mediator of host’s innate and adaptive immune responses, from the bank vole M. glareolus. Soluble dimeric recMgIFN-γ was purified in high yield from Escherichia coli. Its activity on M. glareolus and Microtus arvalis kidney cell lines was assessed by immunofluorescent detection of nuclear translocation and phosphorylation of the transcription factor STAT1. RecMgIFN-γ also induced expression of an IFN-γ-regulated innate immunity gene. Inhibition of vesicular stomatitis virus replication in vole cells upon recMgIFN-γ treatment provided further evidence of its biological activity. Finally, we established a recMgIFN-γ-responsive bank vole reporter cell line that allows the sensitive titration of the cytokine activity via a bioluminescence reporter assay. Taken together, we report valuable tools for future investigations on the immune response against zoonotic pathogens in their natural animal hosts, which might foster the development of novel animal models.
Collapse
Affiliation(s)
- Francesca Torelli
- Department of Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany
| | - Steffen Zander
- Department of Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany
| | - Heinz Ellerbrok
- Center for Biological Threats and Special Pathogens, Highly Pathogenic Viruses, Robert Koch-Institut, Berlin, Germany
| | - Georg Kochs
- Institute of Virology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rainer G Ulrich
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Christian Klotz
- Department of Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany
| | - Frank Seeber
- Department of Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany.
| |
Collapse
|
16
|
Wang Y, Wang R, Chang Y, Gao Y, Li Z, Xue C. Preparation and thermo-reversible gelling properties of protein isolate from defatted Antarctic krill (Euphausia superba) byproducts. Food Chem 2015; 188:170-6. [DOI: 10.1016/j.foodchem.2015.04.126] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/06/2015] [Accepted: 04/20/2015] [Indexed: 11/17/2022]
|
17
|
Tileva M, Krachmarova E, Ivanov I, Maskos K, Nacheva G. Production of aggregation prone human interferon gamma and its mutant in highly soluble and biologically active form by SUMO fusion technology. Protein Expr Purif 2015; 117:26-34. [PMID: 26407523 DOI: 10.1016/j.pep.2015.09.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/16/2015] [Accepted: 09/22/2015] [Indexed: 11/26/2022]
Abstract
The Escherichia coli expression system is a preferable choice for production of recombinant proteins. A disadvantage of this system is the target protein aggregation in "inclusion bodies" (IBs) that further requires solubilisation and refolding, which is crucial for the properties and the yield of the final product. In order to prevent aggregation, SUMO fusion tag technology has been successfully applied for expression of eukaryotic proteins, including human interferon gamma (hIFNγ) that was reported, however, with no satisfactory biological activity. We modified this methodology for expression and purification of both the wild type hIFNγ and an extremely prone to aggregation mutant hIFNγ-K88Q, whose recovery from IBs showed to be ineffective upon numerous conditions. By expression of the N-terminal His-SUMO fusion proteins in the E. coli strain BL21(DE3)pG-KJE8, co-expressing two chaperone systems, at 24 °C a significant increase in solubility of both target proteins (1.5-fold for hIFNγ and 8-fold for K88Q) was achieved. Two-step chromatography (affinity and ion-exchange) with on-dialysis His-SUMO-tag cleavage was applied for protein purification that yielded 6.0-7.0mg/g wet biomass for both proteins with >95% purity and native N-termini. The optimised protocol led to increased yields from 5.5 times for hIFNγ up to 100 times for K88Q in comparison to their isolation from IBs. Purified hIFNγ showed preserved thermal stability and antiproliferative activity corresponding to that of the native reference sample (3 × 10(7)IU/mg). The developed methodology represents an optimised procedure that can be successfully applied for large scale expression and purification of aggregation-prone proteins in soluble native form.
Collapse
Affiliation(s)
- M Tileva
- Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria.
| | - E Krachmarova
- Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria.
| | - I Ivanov
- Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria.
| | - K Maskos
- Proteros Biostructures, D-82152 Martinsried, Germany.
| | - G Nacheva
- Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria.
| |
Collapse
|
18
|
Cardoso AP, Gonçalves RM, Antunes JC, Pinto ML, Pinto AT, Castro F, Monteiro C, Barbosa MA, Oliveira MJ. An interferon-γ-delivery system based on chitosan/poly(γ-glutamic acid) polyelectrolyte complexes modulates macrophage-derived stimulation of cancer cell invasion in vitro. Acta Biomater 2015; 23:157-171. [PMID: 26013040 DOI: 10.1016/j.actbio.2015.05.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/20/2015] [Accepted: 05/19/2015] [Indexed: 01/01/2023]
Abstract
Macrophages represent a large component of the tumour microenvironment and are described to establish interactions with cancer cells, playing crucial roles in several stages of cancer progression. The functional plasticity of macrophages upon stimulation from the environment makes them susceptible to the influence of cancer cells and also renders them as promising therapeutic targets. In this work, we describe a drug delivery system to modulate the phenotype of macrophages, converting them from the pro-tumour M2 phenotype to the anti-tumour M1 phenotype, based on the incorporation of a pro-inflammatory cytokine (interferon-γ) in chitosan (Ch)/poly(γ-glutamic acid) (γ-PGA) complexes. Ch is a biocompatible cationic polysaccharide extensively studied and γ-PGA is a biodegradable, hydrophilic and negatively charged poly-amino acid. These components interact electrostatically, due to opposite charges, resulting in self-assembled structures that can be designed to deliver active molecules such as drugs and proteins. Ch and γ-PGA were self-assembled into polyelectrolyte multilayer films (PEMs) of 371nm thickness, using the layer-by-layer method. Interferon-γ (IFN-γ) was incorporated within the Ch layers at 100 and 500ng/mL. Ch/γ-PGA PEMs with IFN-γ were able to modulate the phenotype of IL-10-treated macrophages at the cell cytoskeleton and cytokine profile levels, inducing an increase of IL-6 and a decrease of IL-10 production. More interestingly, the pro-invasive role of IL-10-treated macrophages was hindered, as their stimulation of gastric cancer cell invasion in vitro decreased from 4 to 2-fold, upon modulation by Ch/γ-PGA PEMs with IFN-γ. This is the first report proposing Ch/γ-PGA PEMs as a suitable strategy to incorporate and release bioactive IFN-γ with the aim of modulating macrophage phenotype, counteracting their stimulating role on gastric cancer cell invasion.
Collapse
Affiliation(s)
- Ana P Cardoso
- I(3)S-Instituto de Investigação e Inovação em Saúde, Universidade doPorto, Porto, Portugal; INEB-Institute of Biomedical Engineering, University of Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal; FEUP-Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Raquel M Gonçalves
- I(3)S-Instituto de Investigação e Inovação em Saúde, Universidade doPorto, Porto, Portugal; INEB-Institute of Biomedical Engineering, University of Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal
| | - Joana C Antunes
- I(3)S-Instituto de Investigação e Inovação em Saúde, Universidade doPorto, Porto, Portugal; INEB-Institute of Biomedical Engineering, University of Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal; ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Marta L Pinto
- I(3)S-Instituto de Investigação e Inovação em Saúde, Universidade doPorto, Porto, Portugal; INEB-Institute of Biomedical Engineering, University of Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal; ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Ana T Pinto
- I(3)S-Instituto de Investigação e Inovação em Saúde, Universidade doPorto, Porto, Portugal; INEB-Institute of Biomedical Engineering, University of Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal; FEUP-Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal
| | - Flávia Castro
- I(3)S-Instituto de Investigação e Inovação em Saúde, Universidade doPorto, Porto, Portugal; INEB-Institute of Biomedical Engineering, University of Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal; ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Cátia Monteiro
- I(3)S-Instituto de Investigação e Inovação em Saúde, Universidade doPorto, Porto, Portugal; INEB-Institute of Biomedical Engineering, University of Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal
| | - Mário A Barbosa
- I(3)S-Instituto de Investigação e Inovação em Saúde, Universidade doPorto, Porto, Portugal; INEB-Institute of Biomedical Engineering, University of Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal; ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Maria José Oliveira
- I(3)S-Instituto de Investigação e Inovação em Saúde, Universidade doPorto, Porto, Portugal; INEB-Institute of Biomedical Engineering, University of Porto, Rua do Campo Alegre, 823, 4150-180 Porto, Portugal; Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
19
|
Galm L, Morgenstern J, Hubbuch J. Manipulation of lysozyme phase behavior by additives as function of conformational stability. Int J Pharm 2015; 494:370-80. [PMID: 26302861 DOI: 10.1016/j.ijpharm.2015.08.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/15/2015] [Indexed: 11/18/2022]
Abstract
Undesired protein aggregation in general and non-native protein aggregation in particular need to be inhibited during bio-pharmaceutical processing to ensure patient safety and to maintain product activity. In this work the potency of different additives, namely glycerol, PEG 1000, and glycine, to prevent lysozyme aggregation and selectively manipulate lysozyme phase behavior was investigated. The results revealed a strong pH dependency of the additive impact on lysozyme phase behavior, lysozyme solubility, crystal size and morphology. This work aims to link this pH dependent impact to a protein-specific parameter, the conformational stability of lysozyme. At pH 3 the addition of 10% (w/v) glycerol, 10% (w/v) PEG 1000, and 1 M glycine stabilized or destabilized lysozymes' native conformation resulting in a modified size of the crystallization area without influencing lysozyme solubility, crystal size and morphology. Addition of 1 M glycine even promoted non-native aggregation at pH 3 whereas addition of PEG 1000 completely inhibited non-native aggregation. At pH 5 the addition of 10% (w/v) glycerol, 10% (w/v) PEG 1000, and 1 M glycine did not influence lysozymes' native conformation, but strongly influenced the position of the crystallization area, lysozyme solubility, crystal size and morphology. The observed pH dependent impact of the additives could be linked to a differing lysozyme conformational stability in the binary systems without additives at pH 3 and pH 5. However, in any case lysozyme phase behavior could selectively be manipulated by addition of glycerol, PEG 1000 and glycine. Furthermore, at pH 5 crystal size and morphology could selectively be manipulated.
Collapse
Affiliation(s)
- Lara Galm
- Institute of Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Josefine Morgenstern
- Institute of Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Jürgen Hubbuch
- Institute of Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| |
Collapse
|
20
|
Concomitant Raman spectroscopy and dynamic light scattering for characterization of therapeutic proteins at high concentrations. Anal Biochem 2014; 472:7-20. [PMID: 25475399 DOI: 10.1016/j.ab.2014.11.016] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 11/21/2022]
Abstract
A Raman spectrometer and dynamic light scattering system were combined in a single platform (Raman-DLS) to provide concomitant higher order structural and hydrodynamic size data for therapeutic proteins at high concentration. As model therapeutic proteins, we studied human serum albumin (HSA) and intravenous immunoglobulin (IVIG). HSA concentration and temperature interval during heating did not affect the onset temperatures for conformation perturbation or aggregation. The impact of pH on thermal stability of HSA was tested at pHs 3, 5, and 8. Stability was the greatest at pH 8, but distinct unfolding and aggregation behaviors were observed at the different pHs. HSA structural transitions and aggregation kinetics were also studied in real time during isothermal incubations at pH 7. In a forced oxidation study, it was found that hydrogen peroxide (H2O2) treatment reduced the thermal stability of HSA. Finally, the structure and thermal stability of IVIG were studied, and a comprehensive characterization of heating-induced structural perturbations and aggregation was obtained. In conclusion, by providing comprehensive data on protein tertiary and secondary structures and hydrodynamic size during real-time heating or isothermal incubation experiments, the Raman-DLS system offers unique physical insights into the properties of high-concentration protein samples.
Collapse
|
21
|
|
22
|
Zhou R, Nashine V, Palm T, Gandhi R, Adams M. Utilization of Zwitterion-based solutions to dissect the relative effects of solution pH and ionic strength on the aggregation behavior and conformational stability of a fusion protein. J Pharm Sci 2014; 103:3065-74. [PMID: 25139470 DOI: 10.1002/jps.24118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 07/07/2014] [Accepted: 07/23/2014] [Indexed: 01/15/2023]
Abstract
Solution pH and ionic strength (I) have complex effects on protein stability. We developed an experimental approach based on exploitation of the zwitterionic characteristic of amino acid molecules to probe the relative contribution from each. A variety of types of amino acid solutions were adopted to investigate the effects of pH and I in a manner that allows independent evaluation of each factor. The same effect could not be achieved using conventional buffer solutions. Size-exclusion chromatography, capillary differential scanning calorimetry, and fluorescence spectroscopy were utilized to probe the protein aggregation and conformation. The results suggested that, in addition to pH, solution ionic strength as a function of ionization state of the amino acid molecules and the ions introduced by pH adjustment played an important role in the aggregation and conformation of the protein studied. This experimental approach offers a useful tool to aid fundamental understanding of the relative effects of solution pH and ionic strength on protein stability.
Collapse
Affiliation(s)
- Rong Zhou
- Drug Product Science & Technology, Bristol-Myers Squibb, New Brunswick, New Jersey, 08903
| | | | | | | | | |
Collapse
|
23
|
High-Throughput Screening and Stability Optimization of Anti-Streptavidin IgG1 and IgG2 Formulations. ACTA ACUST UNITED AC 2014; 19:1290-301. [DOI: 10.1177/1087057114542431] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Selection of a suitable formulation that provides adequate product stability is an important aspect of the development of biopharmaceutical products. Stability of proteins includes not only resistance to chemical modifications but also conformational and colloidal stabilities. While chemical degradation of antibodies is relatively easy to detect and control, propensity for conformational changes and/or aggregation during manufacturing or long-term storage is difficult to predict. In many cases, the formulation factors that increase one type of stability may significantly decrease another type under the same or different conditions. Often compromise is necessary to minimize the adverse effects of an antibody formulation by careful optimization of multiple factors responsible for overall stability. In this study, high-throughput stress and characterization techniques were applied to 96 formulations of anti-streptavidin antibodies (an IgG1 and an IgG2) to choose optimal formulations. Stress and analytical methods applied in this study were 96-well plate based using an automated liquid handling system to prepare the different formulations and sample plates. Aggregation and clipping propensity were evaluated by temperature and mechanical stresses. Multivariate regression analysis of high-throughput data was performed to find statistically significant formulation factors that alter measured parameters such as monomer percentage or unfolding temperature. The results of the regression models were used to maximize the stabilities of antibodies under different formulations and to find the optimal formulation space for each molecule. Comparison of the IgG1 and IgG2 data indicated an overall greater stability of the IgG1 molecule under the conditions studied. The described method can easily be applied to both initial preformulation screening and late-stage formulation development of biopharmaceutical products.
Collapse
|
24
|
Mehta SB, Bee JS, Randolph TW, Carpenter JF. Partial unfolding of a monoclonal antibody: role of a single domain in driving protein aggregation. Biochemistry 2014; 53:3367-77. [PMID: 24804773 DOI: 10.1021/bi5002163] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have examined the effect of incubating a monoclonal antibody (mAb) in low (0-2.0 M) concentrations of guanidine hydrochloride (GdnHCl) on the protein's conformation and aggregation during isothermal incubation. In GdnHCl solutions at concentrations from 1.2 to 1.6 M, the mAb was partially unfolded. As demonstrated by fluorescence and circular dichroism spectroscopy, the partially unfolded state of the antibody had perturbed tertiary structure but retained native secondary structure. Furthermore, partial unfolding of the antibody was documented by analytical ultracentrifugation, dynamic light scattering, and limited proteolysis. Subsequent aggregation of the antibody was characterized using size-exclusion chromatography, analytical ultracentrifugation, and dynamic light scattering. Over the entire concentration range (0-2.0 M) of GdnHCl, protein-protein interactions were attractive, as quantified by negative osmotic second virial coefficients measured with static light scattering. However, during isothermal incubation at 37 °C, the aggregation of the antibody was detected only in solutions that induced partial unfolding. Differential scanning calorimetry studies showed that the antibody's CH2 domains were unfolded in antibody molecules that had been incubated in 1.2 M and higher concentrations of GdnHCl. These results suggest that unfolding of the CH2 domains leads to aggregation.
Collapse
Affiliation(s)
- Shyam B Mehta
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, Colorado 80045, United States
| | | | | | | |
Collapse
|
25
|
Kim NA, Lim DG, Lim JY, Kim KH, Jeong SH. Comprehensive evaluation of etanercept stability in various concentrations with biophysical assessment. Int J Pharm 2014; 460:108-18. [DOI: 10.1016/j.ijpharm.2013.11.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/14/2013] [Accepted: 11/13/2013] [Indexed: 10/26/2022]
|
26
|
Reversible and non-reversible thermal denaturation of lysozyme with varying pH at low ionic strength. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2064-70. [DOI: 10.1016/j.bbapap.2013.06.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 05/29/2013] [Accepted: 06/04/2013] [Indexed: 12/23/2022]
|
27
|
Malek-Sabet N, Masoumian MR, Zeinali M, Khalilzadeh R, Mousaabadi JM. PRODUCTION, PURIFICATION, AND CHEMICAL STABILITY OF RECOMBINANT HUMAN INTERFERON-γ IN LOW OXYGEN TENSION CONDITION: A FORMULATION APPROACH. Prep Biochem Biotechnol 2013; 43:586-600. [DOI: 10.1080/10826068.2012.762716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
28
|
Shivu B, Seshadri S, Li J, Oberg KA, Uversky VN, Fink AL. Distinct β-sheet structure in protein aggregates determined by ATR-FTIR spectroscopy. Biochemistry 2013; 52:5176-83. [PMID: 23837615 DOI: 10.1021/bi400625v] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Attenuated total reflectance Fourier transform infrared spectroscopy (ATR-FTIR) was used to study the conformation of aggregated proteins in vivo and in vitro. Several different protein aggregates, including amyloid fibrils from several peptides and polypeptides, inclusion bodies, folding aggregates, soluble oligomers, and protein extracts from stressed cells, were examined in this study. All protein aggregates demonstrate a characteristic new β structure with lower-frequency band positions. All protein aggregates acquire this new β band following the aggregation process involving intermolecular interactions. The β sheets in some proteins arise from regions of the polypeptide that are helical or non β in the native conformation. For a given protein, all types of the aggregates (e.g., inclusion bodies, folding aggregates, and thermal aggregates) showed similar spectra, indicating that they arose from a common partially folded species. All of the aggregates have some nativelike secondary structure and nonperiodic structure as well as the specific new β structure. The new β could be most likely attributed to stronger hydrogen bonds in the intermolecular β-sheet structure present in the protein aggregates.
Collapse
Affiliation(s)
- Bhavana Shivu
- Department of Chemistry and Biochemistry, University of California , Santa Cruz, California 95064, United States
| | | | | | | | | | | |
Collapse
|
29
|
Belton DJ, Miller AF. Thermal aggregation of recombinant protective antigen: aggregate morphology and growth rate. JOURNAL OF BIOPHYSICS (HINDAWI PUBLISHING CORPORATION : ONLINE) 2013; 2013:751091. [PMID: 23476645 PMCID: PMC3586485 DOI: 10.1155/2013/751091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 12/18/2012] [Indexed: 11/29/2022]
Abstract
The thermal aggregation of the biopharmaceutical protein recombinant protective antigen (rPA) has been explored, and the associated kinetics and thermodynamic parameters have been extracted using optical and environmental scanning electron microscopies (ESEMs) and ultraviolet light scattering spectroscopy (UV-LSS). Visual observations and turbidity measurements provided an overall picture of the aggregation process, suggesting a two-step mechanism. Microscopy was used to examine the structure of aggregates, revealing an open morphology formed by the clustering of the microscopic aggregate particles. UV-LSS was used and developed to elucidate the growth rate of these particles, which formed in the first stage of the aggregation process. Their growth rate is observed to be high initially, before falling to converge on a final size that correlates with the ESEM data. The results suggest that the particle growth rate is limited by rPA monomer concentration, and by obtaining data over a range of incubation temperatures, an approach was developed to model the aggregation kinetics and extract the rate constants and the temperature dependence of aggregation. In doing so, we quantified the susceptibility of rPA aggregation under different temperature and environmental conditions and moreover demonstrated a novel use of UV spectrometry to monitor the particle aggregation quantitatively, in situ, in a nondestructive and time-resolved manner.
Collapse
Affiliation(s)
- Daniel J. Belton
- Department of Chemical and Biological Sciences, University of Huddersfield, Queensgate, Huddersfield HD1 3DH, UK
| | - Aline F. Miller
- School of Chemical Engineering and Analytical Science and Manchester Interdisciplinary Biocentre, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| |
Collapse
|
30
|
The role of trehalose for metastable state and functional form of recombinant interferon beta-1b. J Biotechnol 2013. [DOI: 10.1016/j.jbiotec.2012.11.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
31
|
Kim N, Remmele RL, Liu D, Razinkov VI, Fernandez EJ, Roberts CJ. Aggregation of anti-streptavidin immunoglobulin gamma‐1 involves Fab unfolding and competing growth pathways mediated by pH and salt concentration. Biophys Chem 2013; 172:26-36. [DOI: 10.1016/j.bpc.2012.12.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 11/29/2012] [Accepted: 12/18/2012] [Indexed: 12/16/2022]
|
32
|
Iram A, Amani S, Furkan M, Naeem A. Equilibrium studies of cellulase aggregates in presence of ascorbic and boric acid. Int J Biol Macromol 2013; 52:286-95. [DOI: 10.1016/j.ijbiomac.2012.10.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 09/08/2012] [Accepted: 10/19/2012] [Indexed: 11/17/2022]
|
33
|
Mellor BL, Wood SJ, Mazzeo BA. Quantitation of pH-induced aggregation in binary protein mixtures by dielectric spectroscopy. Protein J 2012; 31:703-9. [PMID: 23001617 DOI: 10.1007/s10930-012-9450-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This paper presents a quantitative approach for measuring pH-controlled protein aggregation using dielectric spectroscopy. The technique is demonstrated through two aggregation experiments, the first between β-lactoglobulin (β-Lg) and hen lysozyme (HENL) and the second between bovine serum albumin (BSA) and HENL. In both experiments, the formation of aggregates is strongly dependent on the solution pH and is clearly indicated by a decrease in the measured permittivity when the second protein is added. A quantifiable lower-bound on the ratio of proteins involved in the aggregation process is obtained from the permittivity spectra. Lower-bound aggregation ratios of 83 % for β-Lg/HENL at pH 6.0 and 48 % for BSA/HENL at pH 9.2 were consistent with turbidity measurements made on the same solutions.
Collapse
Affiliation(s)
- Brett L Mellor
- Department of Electrical and Computer Engineering, Brigham Young University, Provo, UT 84602, USA
| | | | | |
Collapse
|
34
|
Kinetic mechanism of p53 oncogenic mutant aggregation and its inhibition. Proc Natl Acad Sci U S A 2012; 109:13584-9. [PMID: 22869713 DOI: 10.1073/pnas.1211550109] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aggregation of destabilized mutants of the tumor suppressor p53 is a major route for its loss of activity. In order to assay drugs that inhibit aggregation of p53, we established the basic kinetics of aggregation of its core domain, using the mutant Y220C that has a mutation-induced, druggable cavity. Aggregation monitored by light scattering followed lag kinetics. Electron microscopy revealed the formation of small aggregates that subsequently grew to larger amorphous aggregates. The kinetics of aggregation produced surprising results: progress curves followed either by the binding of Thioflavin T or the fluorescence of the protein at 340 nm fitted well to simple two-step sequential first-order lag kinetics with rate constants k(1) and k(2) that were independent of protein concentration, and not to classical nucleation-growth. We suggest a mechanism of first-order formation of an aggregation competent state as being rate determining followed by rapid polymerization with the higher order kinetics. By measuring the inhibition kinetics of k(1) and k(2), we resolved that the process with the higher rate constant followed that of the lower. Further, there was only partial inhibition of k(1) and k(2), which showed two parallel pathways of aggregation, one via a state that requires unfolding of the protein and the other of partial unfolding with the ligand still bound. Inhibition kinetics of ligands provides a useful tool for probing an aggregation mechanism.
Collapse
|
35
|
Chou DK, Krishnamurthy R, Manning MC, Randolph TW, Carpenter JF. Physical Stability of Albinterferon-α2b in Aqueous Solution: Effects of Conformational Stability and Colloidal Stability on Aggregation. J Pharm Sci 2012; 101:2702-19. [DOI: 10.1002/jps.23215] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 03/28/2012] [Accepted: 05/08/2012] [Indexed: 11/12/2022]
|
36
|
Buck PM, Kumar S, Wang X, Agrawal NJ, Trout BL, Singh SK. Computational methods to predict therapeutic protein aggregation. Methods Mol Biol 2012; 899:425-451. [PMID: 22735968 DOI: 10.1007/978-1-61779-921-1_26] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Protein based biotherapeutics have emerged as a successful class of pharmaceuticals. However, these macromolecules endure a variety of physicochemical degradations during manufacturing, shipping, and storage, which may adversely impact the drug product quality. Of these degradations, the irreversible self-association of therapeutic proteins to form aggregates is a major challenge in the formulation of these molecules. Tools to predict and mitigate protein aggregation are, therefore, of great interest to biopharmaceutical research and development. In this chapter, a number of such computational tools developed to understand and predict the various steps involved in protein aggregation are described. These tools can be grouped into three general classes: unfolding kinetics and native state thermal stability, colloidal stability, and sequence/structure based aggregation liabilities. Chapter sections introduce each class by discussing how these predictive tools provide insight into the molecular events leading to protein aggregation. The computational methods are then explained in detail along with their advantages and limitations.
Collapse
Affiliation(s)
- Patrick M Buck
- Biotherapeutics Pharmaceutical Research and Development, Pfizer, Inc, St. Louis, MO, USA
| | | | | | | | | | | |
Collapse
|
37
|
Kumar V, Dixit N, Zhou L(L, Fraunhofer W. Impact of short range hydrophobic interactions and long range electrostatic forces on the aggregation kinetics of a monoclonal antibody and a dual-variable domain immunoglobulin at low and high concentrations. Int J Pharm 2011; 421:82-93. [DOI: 10.1016/j.ijpharm.2011.09.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 08/17/2011] [Accepted: 09/19/2011] [Indexed: 10/17/2022]
|
38
|
Viscosity of concentrated therapeutic protein compositions. Adv Drug Deliv Rev 2011; 63:1107-17. [PMID: 22014592 DOI: 10.1016/j.addr.2011.09.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 09/02/2011] [Indexed: 11/22/2022]
Abstract
The use of monoclonal antibodies as therapeutic agents has been increasing steadily over the last decade for the treatment of various conditions. There is often a need to deliver a large dose of the protein, so there is a trend toward developing commercially viable liquid formulations of highly concentrated antibodies. Such concentrated solutions are associated with a number of challenges, including optimization of production processes, plus chemical and physical stability of the final product where solution viscosity becomes a critical quality attribute. Assessment of the rheological characteristics of concentrated compositions is essential as are development strategies to reduce the viscosity. This review covers the state-of-the-art rheology measurement techniques, focusing particularly on concentrated protein solutions. Current understanding of the mechanisms leading to high viscosity and control by formulation parameters is discussed.
Collapse
|
39
|
Kamerzell TJ, Esfandiary R, Joshi SB, Middaugh CR, Volkin DB. Protein-excipient interactions: mechanisms and biophysical characterization applied to protein formulation development. Adv Drug Deliv Rev 2011; 63:1118-59. [PMID: 21855584 DOI: 10.1016/j.addr.2011.07.006] [Citation(s) in RCA: 350] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 07/19/2011] [Accepted: 07/26/2011] [Indexed: 12/18/2022]
Abstract
The purpose of this review is to demonstrate the critical importance of understanding protein-excipient interactions as a key step in the rational design of formulations to stabilize and deliver protein-based therapeutic drugs and vaccines. Biophysical methods used to examine various molecular interactions between solutes and protein molecules are discussed with an emphasis on applications to pharmaceutical excipients in terms of their effects on protein stability. Key mechanisms of protein-excipient interactions such as electrostatic and cation-pi interactions, preferential hydration, dispersive forces, and hydrogen bonding are presented in the context of different physical states of the formulation such as frozen liquids, solutions, gels, freeze-dried solids and interfacial phenomenon. An overview of the different classes of pharmaceutical excipients used to formulate and stabilize protein therapeutic drugs is also presented along with the rationale for use in different dosage forms including practical pharmaceutical considerations. The utility of high throughput analytical methodologies to examine protein-excipient interactions is presented in terms of expanding formulation design space and accelerating experimental timelines.
Collapse
Affiliation(s)
- Tim J Kamerzell
- Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | | | | | | | |
Collapse
|
40
|
He F, Woods CE, Becker GW, Narhi LO, Razinkov VI. High-throughput assessment of thermal and colloidal stability parameters for monoclonal antibody formulations. J Pharm Sci 2011; 100:5126-41. [PMID: 21789772 DOI: 10.1002/jps.22712] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 06/04/2011] [Accepted: 06/30/2011] [Indexed: 11/07/2022]
Abstract
Design of experiment and statistical analyses were applied to evaluate the effects of several formulation components on the thermal and colloidal stability for a series of monoclonal antibody (mAb) formulations. The high-throughput assessment of the protein stability was performed by measuring the temperature of hydrophobic exposure (T(h) , thermal stability) and the diffusion interaction parameter (k(D) , colloidal stability). To correlate the measured parameters with protein stability, the propensity to aggregate was tested by exposing the mAb samples to two types of stress: mechanical stress caused by shaking agitation and thermal stress. Mechanical stress led to increased formation of large particles, whereas temperature stress resulted in an increase in oligomers. The data obtained from the stress studies were used to determine the critical values for the stability parameters. The optimal formulation compositions were determined based on the statistical models and the predication tests. This approach of high-throughput thermal and colloidal stability screening can be applied to the characterization and prediction of protein formulation properties.
Collapse
Affiliation(s)
- Feng He
- Process & Product Development, Amgen Inc., Seattle, Washington 98119, USA
| | | | | | | | | |
Collapse
|
41
|
Brummitt RK, Nesta DP, Chang L, Kroetsch AM, Roberts CJ. Nonnative Aggregation of an IgG1 Antibody in Acidic Conditions, Part 2: Nucleation and Growth Kinetics with Competing Growth Mechanisms. J Pharm Sci 2011; 100:2104-19. [DOI: 10.1002/jps.22447] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2010] [Revised: 10/09/2010] [Accepted: 11/17/2010] [Indexed: 01/22/2023]
|
42
|
Vázquez-Rey M, Lang DA. Aggregates in monoclonal antibody manufacturing processes. Biotechnol Bioeng 2011; 108:1494-508. [DOI: 10.1002/bit.23155] [Citation(s) in RCA: 334] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Revised: 03/06/2011] [Accepted: 03/30/2011] [Indexed: 12/11/2022]
|
43
|
Roberts CJ, Das TK, Sahin E. Predicting solution aggregation rates for therapeutic proteins: approaches and challenges. Int J Pharm 2011; 418:318-33. [PMID: 21497188 DOI: 10.1016/j.ijpharm.2011.03.064] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 03/17/2011] [Accepted: 03/24/2011] [Indexed: 01/05/2023]
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.
Collapse
Affiliation(s)
- Christopher J Roberts
- Department of Chemical Engineering and Center for Molecular and Engineering Thermodynamics, University of Delaware, Newark, DE 19716, United States.
| | | | | |
Collapse
|
44
|
Goldberg DS, Bishop SM, Shah AU, Sathish HA. Formulation Development of Therapeutic Monoclonal Antibodies Using High-Throughput Fluorescence and Static Light Scattering Techniques: Role of Conformational and Colloidal Stability. J Pharm Sci 2011; 100:1306-15. [DOI: 10.1002/jps.22371] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 09/13/2010] [Accepted: 09/16/2010] [Indexed: 11/11/2022]
|
45
|
Bai S, Manning MC, Randolph TW, Carpenter JF. BIOTECHNOLOGY: Aggregation of Recombinant Human Botulinum Protein Antigen Serotype C in Varying Solution Conditions: Implications of Conformational Stability for Aggregation Kinetics. J Pharm Sci 2011; 100:836-48. [DOI: 10.1002/jps.22345] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 07/19/2010] [Accepted: 08/13/2010] [Indexed: 11/10/2022]
|
46
|
Webster GT, Dusting J, Balabani S, Blanch EW. Detecting the early onset of shear-induced fibril formation of insulin in situ. J Phys Chem B 2011; 115:2617-26. [PMID: 21348502 DOI: 10.1021/jp110367t] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A new approach is presented for detecting the early onset of amyloid fibril formation of insulin in a fluidic environment. The fibrillogenesis of insulin in a well-characterized Taylor-Couette flow cell was analyzed in situ using Raman spectroscopy in combination with principal components analysis (PCA). Raman spectra recorded using a 532.5 nm excitation laser revealed a more rapid fibrillogenesis process during the first 90 min of shearing than previously reported for samples exposed to flow. Bands corresponding to intermolecular H-bonded β-sheet structure of insulin at 1678, 1630, and 1625 cm(-1) observed in the Raman difference spectra between unsheared insulin and sheared insulin show an increase in intensity as a function of shear exposure time, which is characteristic of fibril formation, with the first changes detected after 10 min. Additional analysis of samples removed from the flow cell after specific time periods provided conformation of the flow-enhanced fibrillogenesis process, including the detection of early fibril formation after only 1 min of shearing. FT-IR spectra of the insulin solutions showed evolution of bands at 1673 and 1633 cm(-1) from an increase in H-bonded β-turn and β-sheet structures, respectively, while fluorescence emission spectra detected the presence of a new emission band at 482 nm. TEM images confirmed the early onset of fibril formation at 1 min shear exposure, before a maturation and concentration increase of fibrils with further shearing. This study highlights the ability of fluid flows to accelerate insulin fibril formation, which has important implications for biotechnology applications such as the purification process of insulin therapeutic drugs in the pharmaceutical industry, as well as the use of optical-based methods for detecting fibrillogenesis.
Collapse
Affiliation(s)
- Grant T Webster
- Manchester Interdisciplinary Biocentre and Faculty of Life Sciences, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK.
| | | | | | | |
Collapse
|
47
|
Protasevich I, Yang Z, Wang C, Atwell S, Zhao X, Emtage S, Wetmore D, Hunt JF, Brouillette CG. Thermal unfolding studies show the disease causing F508del mutation in CFTR thermodynamically destabilizes nucleotide-binding domain 1. Protein Sci 2011; 19:1917-31. [PMID: 20687133 DOI: 10.1002/pro.479] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Misfolding and degradation of CFTR is the cause of disease in patients with the most prevalent CFTR mutation, an in-frame deletion of phenylalanine (F508del), located in the first nucleotide-binding domain of human CFTR (hNBD1). Studies of (F508del)CFTR cellular folding suggest that both intra- and inter-domain folding is impaired. (F508del)CFTR is a temperature-sensitive mutant, that is, lowering growth temperature, improves both export, and plasma membrane residence times. Yet, paradoxically, F508del does not alter the fold of isolated hNBD1 nor did it seem to perturb its unfolding transition in previous isothermal chemical denaturation studies. We therefore studied the in vitro thermal unfolding of matched hNBD1 constructs ±F508del to shed light on the defective folding mechanism and the basis for the thermal instability of (F508del)CFTR. Using primarily differential scanning calorimetry (DSC) and circular dichroism, we show for all hNBD1 pairs studied, that F508del lowers the unfolding transition temperature (T(m)) by 6-7°C and that unfolding occurs via a kinetically-controlled, irreversible transition in isolated monomers. A thermal unfolding mechanism is derived from nonlinear least squares fitting of comprehensive DSC data sets. All data are consistent with a simple three-state thermal unfolding mechanism for hNBD1 ± F508del: N(±MgATP) <==> I(T)(±MgATP) → A(T) → (A(T))(n). The equilibrium unfolding to intermediate, I(T), is followed by the rate-determining, irreversible formation of a partially folded, aggregation-prone, monomeric state, A(T), for which aggregation to (A(T))(n) and further unfolding occur with no detectable heat change. Fitted parameters indicate that F508del thermodynamically destabilizes the native state, N, and accelerates the formation of A(T).
Collapse
Affiliation(s)
- Irina Protasevich
- Center for Biophysical Sciences and Engineering, University of Alabama at Birmingham, Birmingham, Alabama 35294-4400, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Pavišić R, Dodig I, Horvatić A, Mijić L, Sedić M, Linarić MR, Sovulj IG, Preočanin T, Krajačić MB, Cindrić M. Differences between reversible (self-association) and irreversible aggregation of rHuG-CSF in carbohydrate and polyol formulations. Eur J Pharm Biopharm 2010; 76:357-65. [DOI: 10.1016/j.ejpb.2010.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 09/06/2010] [Accepted: 09/14/2010] [Indexed: 10/19/2022]
|
49
|
Wang L, Schubert D, Sawaya MR, Eisenberg D, Riek R. Multidimensional structure-activity relationship of a protein in its aggregated states. Angew Chem Int Ed Engl 2010; 49:3904-8. [PMID: 20397175 PMCID: PMC3004770 DOI: 10.1002/anie.201000068] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Lei Wang
- Laboratorium für Physikalische Chemie, ETH Zürich, 8093 Zürich, Switzerland
| | | | | | | | | |
Collapse
|
50
|
Ding Y, Chuan YP, He L, Middelberg AP. Modeling the competition between aggregation and self-assembly during virus-like particle processing. Biotechnol Bioeng 2010; 107:550-60. [DOI: 10.1002/bit.22821] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|