1
|
Prass TM, Lindorff-Larsen K, Garidel P, Blech M, Schäfer LV. Optimized Protein-Excipient Interactions in the Martini 3 Force Field. J Chem Inf Model 2025; 65:3581-3592. [PMID: 40129029 DOI: 10.1021/acs.jcim.4c02338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
The high doses of drugs required for biotherapeutics, such as monoclonal antibodies (mAbs), and the small volumes that can be administered to patients by subcutaneous injections pose challenges due to high-concentration formulations. The addition of excipients, such as arginine and glutamate, to high-concentration protein formulations can increase solubility and reduce the tendency of protein particle formation. Molecular dynamics (MD) simulations can provide microscopic insights into the mode of action of excipients in mAb formulations but require large system sizes and long time scales that are currently beyond reach at the fully atomistic level. Computationally efficient coarse-grained models such as the Martini 3 force field can tackle this challenge but require careful parametrization, testing, and validation. This study extends the popular Martini 3 force field toward realistic protein-excipient interactions of arginine and glutamate excipients, using the Fab domains of the therapeutic mAbs trastuzumab and omalizumab as model systems. A novel all-atom to coarse-grained mapping of the amino acid excipients is introduced, which explicitly captures the zwitterionic character of the backbone. The Fab-excipient interactions of arginine and glutamate are characterized concerning molecular contacts with the Fabs at the single-residue level. The Martini 3 simulations are compared with results from all-atom simulations as a reference. Our findings reveal an overestimation of Fab-excipient contacts with the default interaction parameters of Martini 3, suggesting a too strong attraction between protein residues and excipients. Therefore, we reparametrized the protein-excipient interaction parameters in Martini 3 against all-atom simulations. The excipient interactions obtained with the new Martini 3 mapping and Lennard-Jones (LJ) interaction parameters, coined Martini 3-exc, agree closely with the all-atom reference data. This work presents an improved parameter set for mAb-arginine and mAb-glutamate interactions in the Martini 3 coarse-grained force field, a key step toward large-scale coarse-grained MD simulations of high-concentration mAb formulations and the stabilizing effects of excipients.
Collapse
Affiliation(s)
- Tobias M Prass
- Center for Theoretical Chemistry, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Patrick Garidel
- Innovation Unit, Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, D-88397 Biberach an der Riss, Germany
| | - Michaela Blech
- Innovation Unit, Analytical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, D-88397 Biberach an der Riss, Germany
| | - Lars V Schäfer
- Center for Theoretical Chemistry, Ruhr University Bochum, D-44780 Bochum, Germany
| |
Collapse
|
2
|
Xin L, Prorok M, Zhang Z, Barboza G, More R, Bonfiglio M, Cheng L, Robbie K, Ren S, Li Y. Rapid Development of High Concentration Protein Formulation Driven by High-Throughput Technologies. Pharm Res 2025; 42:151-171. [PMID: 39824982 DOI: 10.1007/s11095-024-03801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/01/2024] [Indexed: 01/20/2025]
Abstract
BACKGROUND High concentration protein formulation (HCPF) development needs to balance protein stability attributes such as conformational/colloidal stability, chemical stability, and solution properties such as viscosity and osmolality. METHODOLOGY A three-phase design is established in this work. In Phase 1, conformational and colloidal stability are measured by 384-well-based high-throughput (HT) biophysical screening while viscosity reduction screening is performed with HT viscosity screening. Collectively, the biophysical and viscosity screening data are leveraged to design the phase 2 of short-term stability study, executed using 96-well plates under thermal and freeze/thaw stresses. In phase 2, samples are analyzed by stability-indicating assays and processed with pair-wise Student's t-test analyses to choose the final formulations. In phase 3, the final formulations are then confirmed through a one-month accelerated stability in glass vials. RESULTS Using a model antibody A (mAb-A), the initial HT screening successfully established the 384-well based platform. A lead formulation was chosen from the second round based on statistical analyses and subsequently tested against the commercial formulation of mAb-A as a control. Compared to the control, the lead formulation reduced the viscosity of mAb-A by 30% and decreased subvisible particles after thermal stress by 80%. CONCLUSIONS HT biophysical screening in 384-well plates was demonstrated to effectively guide the rational design of a high-throughput stability screening study using 96-well plates. This platform enables the identification of a high concentration formulation within seven weeks within the first two phases of study that strategically balance stability with solution properties, thus achieving a rapid development of HCPF.
Collapse
Affiliation(s)
- Lun Xin
- BioDev Department WuXi Biologics USA, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | - Monika Prorok
- BioDev Department WuXi Biologics USA, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | - Zhe Zhang
- BioDev Department WuXi Biologics USA, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | - Guilherme Barboza
- BioDev Department WuXi Biologics USA, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | - Rahul More
- BioDev Department WuXi Biologics USA, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | - Michael Bonfiglio
- BioDev Department WuXi Biologics USA, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | - Lv Cheng
- BioDev Department WuXi Biologics USA, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | - Kevin Robbie
- BioDev Department WuXi Biologics USA, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA
| | - Steven Ren
- CMC Management, WuXi Biologics, Cranbury, NJ, USA
| | - Yunsong Li
- BioDev Department WuXi Biologics USA, 1 Cedarbrook Dr, Cranbury, NJ, 08512, USA.
| |
Collapse
|
3
|
Sreenivasan S, Schöneich C, Rathore AS. Aggregation of therapeutic monoclonal antibodies due to thermal and air/liquid interfacial agitation stress: Occurrence, stability assessment strategies, aggregation mechanism, influencing factors, and ways to enhance stability. Int J Pharm 2024; 666:124735. [PMID: 39326478 DOI: 10.1016/j.ijpharm.2024.124735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
Therapeutic proteins, such as monoclonal antibodies (mAbs) are known to undergo stability related issues during various stages of product life cycle resulting in the formation of aggregates and fragments. Aggregates of mAb might result in reduced therapeutic activity and could cause various adverse immunogenic responses. Sample containing mAb undergo aggregation due to various types of stress factors, and there is always a continuous interest among researchers and manufacturers to determine the effect of different factors on the stability of mAb. Thermal stress and air/liquid interfacial agitation stress are among two of the common stress factors to which samples containing mAb are exposed to during various stages. Initial part of this review articles aims to provide a generalized understanding of aggregation of mAb such as size ranges of aggregates, aggregate types, stress factors, analytical techniques, permissible aggregate limits, and stability assessment methods. This article further aims to explain different aspects associated with aggregation of mAb in liquid samples due to thermal and air/liquid interfacial agitation stress. Under each stress category, the occurrence of stress during product life cycle, type of aggregates formed, mechanism of aggregation, strategies used by various researchers to expose mAb containing samples to stress, different factors affecting aggregation, fate of aggregates in human body fluids, and strategies used to enhance mAb stability has been explained in detail. The authors hope that this article provides a detailed understanding about stability of mAb due to thermal and air/liquid interfacial stress with relevance to product life cycle from manufacturing to administration into patients.
Collapse
Affiliation(s)
- Shravan Sreenivasan
- Department of Chemical Engineering, Indian Institute of Technology Delhi, India
| | | | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, India.
| |
Collapse
|
4
|
Xin L, Lan L, Mellal M, McChesney N, Vaughan R, Berdugo C, Li Y, Zhang J. Leveraging high-throughput analytics and automation to rapidly develop high-concentration mAb formulations: integrated excipient compatibility and viscosity screening. Antib Ther 2024; 7:335-350. [PMID: 39678259 PMCID: PMC11646310 DOI: 10.1093/abt/tbae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/03/2024] [Accepted: 10/10/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Formulation screening is essential to experimentally balance stability and viscosity in high-concentration mAb formulations. We developed a high-throughput approach with automated sample preparation and analytical workflows to enable the integrated assessment of excipient compatibility and viscosity of mAb formulations. METHODS Ninety-six formulations of a trastuzumab biosimilar were screened by combining 8 types of excipient modifiers with 4 types of buffers across a pH range of 4.5 to 7.5. Key stability risks, including high molecular weight (HMW) aggregation and fragmentation, were thoroughly assessed along with viscosity at high concentrations. Additionally, several biophysical parameters were evaluated for their ability to predict stability or viscosity outcomes. Multiple linear regression was applied to fit the data and identify key factors. RESULTS The optimal pH range for the trastuzumab biosimilar was found to be 5.0 to 6.5, based on opposing pH dependencies for stability and viscosity. Buffer type had a minor effect on viscosity and fragmentation but played a significant role in influencing HMW aggregates, with Na-acetate and histidine-HCl being the best candidates. The impact of excipient modifiers on viscosity, HMW, and fragmentation depended on both pH and buffer type, showing strong interactions among factors. Arginine-HCl and lysine-HCl effectively lowered viscosity of the trastuzumab biosimilar at pH levels above 6.0, while glycine formulations were more effective at reducing viscosity below pH 6.0. Histidine-HCl, arginine-HCl, and lysine-HCl lowered the risk of HMW aggregation, whereas formulations containing Na-phosphate or NaCl showed higher HMW aggregation. Formulations with arginine-HCl, lysine-HCl, and NaCl demonstrated a rapid increase in fragmentation at pH levels below 5.0, while Na-aspartate formulations showed increased fragmentation at pH levels above 6.5. CONCLUSION Hence, it is important to optimize the levels of each chosen excipient in the formulation study to balance their benefits against potential incompatibilities. This study serves as a foundation for identifying high-concentration antibody formulations using a high-throughput approach, where minimal materials are required, and optimized formulation design spaces can be quickly identified.
Collapse
Affiliation(s)
- Lun Xin
- Product Development, Catalent Pharma Solutions, 3770 W. Jonathan Dr., Bloomington, IN 47404, United States
| | - Lan Lan
- Product Development, Catalent Pharma Solutions, 3770 W. Jonathan Dr., Bloomington, IN 47404, United States
| | - Mourad Mellal
- Product Development, Catalent Pharma Solutions, 14 School House Rd, Somerset, NJ 08873, United States
| | - Nathan McChesney
- Product Development, Catalent Pharma Solutions, 3770 W. Jonathan Dr., Bloomington, IN 47404, United States
| | - Robert Vaughan
- Product Development, Catalent Pharma Solutions, 3770 W. Jonathan Dr., Bloomington, IN 47404, United States
| | - Claudia Berdugo
- Product Development, Catalent Pharma Solutions, 3770 W. Jonathan Dr., Bloomington, IN 47404, United States
| | - Yunsong Li
- Product Development, Catalent Pharma Solutions, 3770 W. Jonathan Dr., Bloomington, IN 47404, United States
| | - Jingtao Zhang
- Product Development, Catalent Pharma Solutions, 14 School House Rd, Somerset, NJ 08873, United States
| |
Collapse
|
5
|
Salehian M, Emamzadeh R, Nazari M. Exploring the Potential of Arginine to Increase Coelenterazine-Renilla Luciferase Affinity and Enzyme Stability: Kinetic and Molecular Dynamics Studies. Protein J 2024; 43:739-750. [PMID: 38824468 DOI: 10.1007/s10930-024-10208-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2024] [Indexed: 06/03/2024]
Abstract
Renilla luciferase catalyzes the oxidation of coelenterazine to coelenteramide and results in the emission of a photon of light. Although Renilla luciferase has various applications in biotechnology, its low thermal stability limits the development of its applications. Arginine is a well-known stabilizing amino acid that plays a key role in protein stabilization against inactivation. However, its impact on enzyme properties is unpredictable. This study investigates the impact of arginine on the kinetics and thermal stability of Renilla luciferase. The enzyme's performance was significantly enhanced in the presence of arginine, with catalytic efficiency increasing by 3.31-fold and 3.08-fold when exposed to 0.2 M and 0.3 M arginine, respectively. Additionally, arginine improved the thermal stability of Renilla luciferase. Molecular dynamics simulation showed that the addition of 0.2 M arginine reduced the binding of coelenteramide, the reaction product and an enzyme inhibitor, to the active site of the Renilla luciferase. Therefore, the release of the product was accelerated, and the affinity of Renilla luciferase for coelenterazine increased. Furthermore, Molecular dynamics studies indicated an increased network of water molecules surrounding Renilla luciferase in the presence of 0.2 M arginine. This network potentially enhances the hydrophobic effect on the protein structure, ultimately improving enzyme stability. The findings of this study hold promise for the development of commercial kits incorporating Renilla luciferase.
Collapse
Affiliation(s)
- Maryam Salehian
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Rahman Emamzadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Mahboobeh Nazari
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Manning MC, Holcomb RE, Payne RW, Stillahn JM, Connolly BD, Katayama DS, Liu H, Matsuura JE, Murphy BM, Henry CS, Crommelin DJA. Stability of Protein Pharmaceuticals: Recent Advances. Pharm Res 2024; 41:1301-1367. [PMID: 38937372 DOI: 10.1007/s11095-024-03726-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/03/2024] [Indexed: 06/29/2024]
Abstract
There have been significant advances in the formulation and stabilization of proteins in the liquid state over the past years since our previous review. Our mechanistic understanding of protein-excipient interactions has increased, allowing one to develop formulations in a more rational fashion. The field has moved towards more complex and challenging formulations, such as high concentration formulations to allow for subcutaneous administration and co-formulation. While much of the published work has focused on mAbs, the principles appear to apply to any therapeutic protein, although mAbs clearly have some distinctive features. In this review, we first discuss chemical degradation reactions. This is followed by a section on physical instability issues. Then, more specific topics are addressed: instability induced by interactions with interfaces, predictive methods for physical stability and interplay between chemical and physical instability. The final parts are devoted to discussions how all the above impacts (co-)formulation strategies, in particular for high protein concentration solutions.'
Collapse
Affiliation(s)
- Mark Cornell Manning
- Legacy BioDesign LLC, Johnstown, CO, USA.
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA.
| | - Ryan E Holcomb
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Robert W Payne
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | - Joshua M Stillahn
- Legacy BioDesign LLC, Johnstown, CO, USA
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | | | | | | | | | | - Charles S Henry
- Department of Chemistry, Colorado State University, Fort Collins, CO, USA
| | | |
Collapse
|
7
|
Ahmad A, Joshi P, Mishra R. Amino acids and glycine derivatives differently affect refolding of mesophilic and thermophilic like α-amylases: implications in protein refolding and aggregation. J Biomol Struct Dyn 2024:1-14. [PMID: 38486406 DOI: 10.1080/07391102.2024.2327540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 03/02/2024] [Indexed: 03/29/2025]
Abstract
α-amylases are industrially important enzymes which are used in different starch-based industries. They are adapted to different environmental conditions like extremes of temperature, pH and salinity. Herein, α-amylases from Bacillus amyloliquifaciens (BAA) and Bacillus licheniformis (BLA), representing mesophilic and thermophilic-like proteins, respectively, have been used to investigate the effect of naturally occurring osmolytes like arginine, proline, glycine and its methyl derivatives, sarcosine and betaine on their refolding. In this study, we have shown that among amino acids and glycine derivatives, betaine is the most promising osmolyte, while arginine and glycine exhibit moderately positive effect at their lower concentrations on the refolding of BAA only. Except betaine, all other osmolytes above 0.25 M showed inhibitory effect on the native enzyme activity of BLA and BAA. However, aggregation kinetics monitored by static light scattering indicates suppression of aggregation by all of these osmolytes. Further investigation by tryptophan and ANS fluorescence spectroscopy indicates the formation of compact hydrophobic core in the presence of the osmolytes. The morphology of protein aggregates having different sizes was visualized by atomic force microscopy ,and it was observed that amorphous aggregates of variable heights were formed. Our study highlights the importance of differential effects of arginine, proline, glycine, sarcosine and betaine on the native state as well as on refolding of BLA and BAA which may be helpful in devising strategies for developing effective protein formulation and prevention of aggregation of industrially and therapeutically important proteins.
Collapse
Affiliation(s)
- Aziz Ahmad
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Prachi Joshi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Rajesh Mishra
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
8
|
Prass TM, Garidel P, Schäfer LV, Blech M. Residue-resolved insights into the stabilization of therapeutic proteins by excipients: A case study of two monoclonal antibodies with arginine and glutamate. MAbs 2024; 16:2427771. [PMID: 39540607 PMCID: PMC11572152 DOI: 10.1080/19420862.2024.2427771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Protein formulation development relies on the selection of excipients that inhibit protein-protein interactions preventing aggregation. Empirical strategies involve screening many excipient and buffer combinations by physicochemical characterization using forced degradation or temperature-induced stress, mostly under accelerated conditions. Such methods do not readily provide information on the inter- and intramolecular interactions responsible for the effects of excipients. Here, we describe a combined experimental and computational approach for investigating the effect of protein-excipient interactions on formulation stability, which allows the identification of preferential interaction sites and thus can aid in the selection of excipients to be experimentally screened. Model systems composed of two marketed therapeutic IgG1 monoclonal antibodies with identical Fc domain sequences, trastuzumab and omalizumab, were investigated with commonly used excipients arginine, glutamate, and equimolar arginine/glutamate mixtures. Protein-excipient interactions were studied using all-atom molecular dynamics (MD) simulations, which show accumulation of the excipients at specific antibody regions. Preferential excipient-interaction sites were particularly found for charged and aromatic residues and in the complementary-determining regions, with more pronounced arginine contacts for omalizumab than trastuzumab. These computational findings are in line with the more pronounced stabilizing effects of arginine observed in the long-term storage stability study. Furthermore, the aggregation and solubility propensity predicted by commonly used in silico tools do not align with the preferential excipient-interaction sites identified by the MD simulations, suggesting that different physicochemical mechanisms are at play.
Collapse
Affiliation(s)
- Tobias M. Prass
- Center for Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany
| | - Patrick Garidel
- Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, Biberach and der Riss, Germany
| | - Lars V. Schäfer
- Center for Theoretical Chemistry, Ruhr University Bochum, Bochum, Germany
| | - Michaela Blech
- Pharmaceutical Development Biologicals, Boehringer Ingelheim Pharma GmbH & Co. KG, Innovation Unit, Biberach and der Riss, Germany
| |
Collapse
|
9
|
Wu J, Lv J, Zhao L, Zhao R, Gao T, Xu Q, Liu D, Yu Q, Ma F. Exploring the role of microbial proteins in controlling environmental pollutants based on molecular simulation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167028. [PMID: 37704131 DOI: 10.1016/j.scitotenv.2023.167028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/03/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
Molecular simulation has been widely used to study microbial proteins' structural composition and dynamic properties, such as volatility, flexibility, and stability at the microscopic scale. Herein, this review describes the key elements of molecular docking and molecular dynamics (MD) simulations in molecular simulation; reviews the techniques combined with molecular simulation, such as crystallography, spectroscopy, molecular biology, and machine learning, to validate simulation results and bridge information gaps in the structure, microenvironmental changes, expression mechanisms, and intensity quantification; illustrates the application of molecular simulation, in characterizing the molecular mechanisms of interaction of microbial proteins with four different types of contaminants, namely heavy metals (HMs), pesticides, dyes and emerging contaminants (ECs). Finally, the review outlines the important role of molecular simulations in the study of microbial proteins for controlling environmental contamination and provides ideas for the application of molecular simulation in screening microbial proteins and incorporating targeted mutagenesis to obtain more effective contaminant control proteins.
Collapse
Affiliation(s)
- Jieting Wu
- School of Environmental Science, Liaoning University, Shenyang 110036, China
| | - Jin Lv
- School of Environmental Science, Liaoning University, Shenyang 110036, China
| | - Lei Zhao
- State Key Laboratory of Urban Water Resources & Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Ruofan Zhao
- School of Environment, Beijing Normal University, Beijing 100875, China
| | - Tian Gao
- Key Laboratory of Integrated Regulation and Resource Development of Shallow Lakes, Ministry of Education, College of Environment, Hohai University, Xikang Road #1, Nanjing 210098, China
| | - Qi Xu
- PetroChina Fushun Petrochemical Company, Fushun 113000, China
| | - Dongbo Liu
- School of Environmental Science, Liaoning University, Shenyang 110036, China
| | - Qiqi Yu
- School of Environmental Science, Liaoning University, Shenyang 110036, China
| | - Fang Ma
- State Key Laboratory of Urban Water Resources & Environment, Harbin Institute of Technology, Harbin 150090, China.
| |
Collapse
|
10
|
Willis LF, Toprani V, Wijetunge S, Sievers A, Lin L, Williams J, Crowley TJ, Radford SE, Kapur N, Brockwell DJ. Exploring a role for flow-induced aggregation assays in platform formulation optimisation for antibody-based proteins. J Pharm Sci 2023; 113:S0022-3549(23)00441-0. [PMID: 39492475 DOI: 10.1016/j.xphs.2023.10.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
The development time of therapeutic monoclonal antibodies (mAbs) has been shortened by formulation platforms and the assessment of 'protein stability' using 'developability' assays. A range of assays are used to measure stability to a variety of stresses, including forces induced by hydrodynamic flow. We have previously developed a low-volume Extensional Flow Device (EFD) which subjects proteins to defined fluid flow fields in the presence of glass interfaces and used it to identify robust candidate sequences. Here, we study the aggregation of mAbs and Fc-fusion proteins using the EFD and orbital shaking under different formulations, investigating the relationship between these assays and evaluating their potential in formulation optimisation. EFD experiments identified the least aggregation-prone molecule using a fraction of the material and time involved in traditional screening. We also show that the EFD can differentiate between different formulations and that protective formulations containing polysorbate 80 stabilised poorly developable Fc-fusion proteins against EFD-induced aggregation up to two-fold. Our work highlights common platform formulation additives that affect the extent of aggregation under EFD-stress, as well as identifying factors that modulate the underlying aggregation mechanism. Together, our data could aid the choice of platform formulations early in development for next-generation therapeutics including fusion proteins.
Collapse
Affiliation(s)
- Leon F Willis
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds UK LS2 9JT
| | - Vishal Toprani
- Pharmaceutical Research and Development, Pfizer Inc. 1 Burtt Road, Andover, Massachusetts, USA, 01810.
| | - Sashini Wijetunge
- Pharmaceutical Research and Development, Pfizer Inc. 1 Burtt Road, Andover, Massachusetts, USA, 01810
| | - Annette Sievers
- BioMedicine Design, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, MA 02139
| | - Laura Lin
- BioMedicine Design, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, MA 02139
| | - Jeanine Williams
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds UK LS2 9JT
| | - Tom J Crowley
- Pharmaceutical Research and Development, Pfizer Inc. 1 Burtt Road, Andover, Massachusetts, USA, 01810
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds UK LS2 9JT
| | - Nikil Kapur
- School of Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds UK LS2 9JT
| | - David J Brockwell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds UK LS2 9JT.
| |
Collapse
|
11
|
Ren S. Effects of arginine in therapeutic protein formulations: a decade review and perspectives. Antib Ther 2023; 6:265-276. [PMID: 38075239 PMCID: PMC10702853 DOI: 10.1093/abt/tbad022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/05/2023] [Accepted: 10/01/2023] [Indexed: 10/14/2024] Open
Abstract
Arginine (Arg) is a natural amino acid with an acceptable safety profile and a unique chemical structure. Arg and its salts are highly effective in enhancing protein refolding and solubilization, suppressing protein-protein interaction and aggregation and reducing viscosity of high concentration protein formulations. Arg and its salts have been used in research and 20 approved protein injectables. This review summarizes the effects of Arg as an excipient in therapeutic protein formulations with the focus on its physicochemical properties, safety, applications in approved protein products, beneficial and detrimental effects in liquid and lyophilized protein formulations when combined with different counterions and mechanism on protein stabilization and destabilization. The decade literature review indicates that the benefits of Arg overweigh its risks when it is used appropriately. It is recommended to add Arg along with glutamate as a counterion to high concentration protein formulations on top of sugars or polyols to counterbalance the negative effects of Arg hydrochloride. The use of Arg as a viscosity reducer and protein stabilizer in high concentration formulations will be the inevitable future trend of the biopharmaceutical industry for subcutaneous administration.
Collapse
Affiliation(s)
- Steven Ren
- CMC Management, WuXi Biologics, 7 Clarke Drive, Cranbury, NJ 08512, USA
| |
Collapse
|
12
|
Garcia-Villen F, Gallego I, Sainz-Ramos M, Ordoyo-Pascual J, Ruiz-Alonso S, Saenz-del-Burgo L, O’Mahony C, Pedraz JL. Stability of Monoclonal Antibodies as Solid Formulation for Auto-Injectors: A Pilot Study. Pharmaceutics 2023; 15:2049. [PMID: 37631263 PMCID: PMC10459033 DOI: 10.3390/pharmaceutics15082049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Drug adherence is a significant medical issue, often responsible for sub-optimal outcomes during the treatment of chronic diseases such as rheumatoid or psoriatic arthritis. Monoclonal antibodies (which are exclusively given parenterally) have been proven to be an effective treatment in these cases. The use of auto-injectors is an effective strategy to improve drug adherence in parenteral treatments since these pen-like devices offer less discomfort and increased user-friendliness over conventional syringe-based delivery. This study aims to investigate the feasibility of including a monoclonal antibody as a solid formulation inside an auto-injector pen. Specifically, the objective was to evaluate the drug stability after a concentration (to reduce the amount of solvent and space needed) and freeze-drying procedure. A preliminary screening of excipients to improve stability was also performed. The nano-DSC results showed that mannitol improved the stability of the concentrated, freeze-dried antibody in comparison to its counterpart without it. However, a small instability of the CH2 domain was still found for mannitol samples, which will warrant further investigation. The present results serve as a stepping stone towards advancing future drug delivery systems that will ultimately improve the patient experience and associated drug adherence.
Collapse
Affiliation(s)
- Fatima Garcia-Villen
- NanoBioCel Group, Laboratory of Pharmaceutics, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria-Gasteiz, Spain (L.S.-d.-B.)
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Resarch Group, 01009 Vitoria-Gasteiz, Spain
| | - Idoia Gallego
- NanoBioCel Group, Laboratory of Pharmaceutics, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria-Gasteiz, Spain (L.S.-d.-B.)
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Resarch Group, 01009 Vitoria-Gasteiz, Spain
| | - Myriam Sainz-Ramos
- NanoBioCel Group, Laboratory of Pharmaceutics, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria-Gasteiz, Spain (L.S.-d.-B.)
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Resarch Group, 01009 Vitoria-Gasteiz, Spain
| | - Jorge Ordoyo-Pascual
- NanoBioCel Group, Laboratory of Pharmaceutics, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria-Gasteiz, Spain (L.S.-d.-B.)
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Resarch Group, 01009 Vitoria-Gasteiz, Spain
| | - Sandra Ruiz-Alonso
- NanoBioCel Group, Laboratory of Pharmaceutics, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria-Gasteiz, Spain (L.S.-d.-B.)
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Resarch Group, 01009 Vitoria-Gasteiz, Spain
| | - Laura Saenz-del-Burgo
- NanoBioCel Group, Laboratory of Pharmaceutics, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria-Gasteiz, Spain (L.S.-d.-B.)
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Resarch Group, 01009 Vitoria-Gasteiz, Spain
| | - Conor O’Mahony
- Tyndall National Institute, University College Cork, T12 R5CP Cork, Ireland;
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria-Gasteiz, Spain (L.S.-d.-B.)
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Resarch Group, 01009 Vitoria-Gasteiz, Spain
| |
Collapse
|
13
|
Rembert KB, Zhang J, Lee YJ. Effects of Salts and Surface Charge on the Biophysical Stability of a Low pI Monoclonal Antibody. J Pharm Sci 2023; 112:947-953. [PMID: 36395898 DOI: 10.1016/j.xphs.2022.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022]
Abstract
The impact of five representative Hofmeister salts (NaCl, KCl, MgCl2, Na2SO4, and NaSCN) on the thermal stability and aggregation kinetics of a slightly acidic monoclonal antibody (mAb) were investigated under different pH conditions. The thermal stability of the mAb was assessed by measuring the lowest unfolding transition temperature, Tm, with differential scanning fluorimetry. MgCl2 and NaSCN significantly decreased Tm at all three charged states of the mAb, but to the greatest extent when the mAb surface charge was net positive. Non-native aggregation kinetics was monitored by measuring Rayleigh light scattering. When the mAb surface charge was net positive or net neutral, the nucleation rate increased non-monotonically with MgCl2 and NaSCN but decreased monotonically with NaCl, KCl, and Na2SO4. By contrast, when the mAb surface was negatively charged, there were only minor changes in the nucleation rate with all salts tested. Furthermore, there was less structural perturbation and slower aggregation rates when the mAb was net negatively charged than when it was net neutrally or positively charged. The observed salt effects on thermal unfolding are consistent with ion-specific mechanisms dominated by short-range amide backbone binding. On the other hand, the salt effects on nucleation rates appear to be influenced by both amide backbone binding and long-range electrostatic binding of ions to charged amino acid side chains.
Collapse
Affiliation(s)
- Kelvin B Rembert
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, United States
| | - Jifeng Zhang
- Department of Drug Delivery and Device Development, Medimmune-AstraZeneca, Gaithersburg, MD 20878, United States.
| | - Young Jong Lee
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD 20899, United States.
| |
Collapse
|
14
|
Two peak elution behavior of a monoclonal antibody in cation exchange chromatography as a screening tool for excipients. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1214:123563. [PMID: 36525885 DOI: 10.1016/j.jchromb.2022.123563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Aggregation of proteins is a critical quality attribute and a major concern during the purification of therapeutic proteins, like monoclonal antibodies. In-solution experiments applying different stress scenarios, e.g., mechanical, or physical stresses, can determine the overall conformational stability of the protein to enhance drug product shelf-life. Several groups have reported surface-induced unfolding and aggregation of monoclonal antibodies and their derivatives during cation exchange chromatography, which results in a two-peak elution behavior of the protein and its species. We have investigated universal influencing factors, like temperature and hold time, on this phenomenon. The formation of the second peak is a kinetic process, which is strongly influenced by temperature during the hold time. However, our main focus was the application of excipients and their influence on the two-peak elution behavior. We compared the on-column screening results with results obtained through a "traditional" in-solution screening using nanoDSF. Mostly, stabilizing excipients, like Sucrose, show their stabilizing abilities in both systems, but some discrepancies, e.g., using Arginine, between the two orthogonal techniques show the potential of the on-column screening system to lead to unexpected results, which would not necessarily be visible in in-solution experiments.
Collapse
|
15
|
Ling MX, Nguyen M, McFaul CA, Lee RC. Peroxidation of tetronic 1107 reduces protein chaperone effect. Phys Biol 2022; 19. [PMID: 35545073 DOI: 10.1088/1478-3975/ac6eaf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 05/11/2022] [Indexed: 11/12/2022]
Abstract
To enhance the stability of protein therapeutics, pharmaceutical companies have long used various copolymer surfactants as excipients. They act to stabilize proteins by adhering to the hydrophobic surface of the protein preventing denaturation and aggregation. However, some commonly used excipients possess polyoxyalkylene chains that are susceptible to oxidative degradation while in aqueous solution. We postulate that oxidation reactions involving the hydrophobic domains reduce the surfactant's ability to stabilize the native protein structure. We investigated the effect of UV (λ=254 nm) radiated Poloxamine T1107 (T1107) on its ability to disaggregate DTT denatured hen egg-white lysozyme (HEWL). Peroxidation of UV irradiated T1107 was analyzed using FTIR spectroscopy, the Fe+2 to Fe+3 ion reduction assay method, and 1H NMR. Our results indicate that increased UV irradiation led to structural changes in T1107, specifically the addition of a carbonyl on the formate group. The structural change decreased T1107's ability to disaggregate HEWL. These results indicate that peroxide content is an important parameter to control in polyoxyalkylene-based excipients.
Collapse
Affiliation(s)
- Michelle X Ling
- Plastic and Reconstructive Surgery, University of Chicago Biological Sciences Division, 5841 S. Maryland Ave., Chicago, Illinois, 60637-5416, UNITED STATES
| | - Michelle Nguyen
- Plastic and Reconstructive Surgery, University of Chicago Biological Sciences Division, 5841 S. Maryland Ave., Chicago, Illinois, 60637-5416, UNITED STATES
| | - Colin A McFaul
- Plastic and Reconstructive Surgery, University of Chicago Biological Sciences Division, 5841 S. Maryland Ave., Chicago, Illinois, 60637-5416, UNITED STATES
| | - Raphael C Lee
- Plastics and Reconstructive Surgery, University of Chicago Biological Sciences Division, 5841 S. Maryland Ave., Chicago, Illinois, 60637-5416, UNITED STATES
| |
Collapse
|
16
|
Munjal B, Patel SM, Suryanarayanan R. Role of Arginine Salts in Preventing Freezing-induced Increase in Subvisible Particles in Protein Formulations. Int J Pharm 2022; 619:121694. [PMID: 35331829 DOI: 10.1016/j.ijpharm.2022.121694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022]
Abstract
While arginine hydrochloride (ArgHCl) has emerged as a potential stabilizer of protein drugs in liquid formulations the purpose of this manuscript was to evaluate its stabilization potential in frozen solutions. The phase behavior of frozen AgHCl solutions was investigated by differential scanning calorimetry and low temperature powder X-ray diffractometry. The aggregation of β-galactosidase was evaluated following freeze-thaw cycling in ArgHCl solutions with and without mannitol. ArgHCl (5% w/v) was retained amorphous in frozen aqueous solutions and effectively inhibited protein aggregation even after 5 freeze-thaw cycles. Annealing frozen arginine solution (5% w/v) containing mannitol (10% w/v) induced mannitol crystallization which in turn facilitated crystallization of ArgHCl. The stabilizing effect of ArgHCl was completely lost in the presence of mannitol. Use of alternate arginine salts (aspartate, glutamate, and acetate) allowed selective crystallization of mannitol while the arginine was retained amorphous and stabilized the protein.
Collapse
Affiliation(s)
- Bhushan Munjal
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Sajal M Patel
- Dosage Form Design & Development, Biopharmaceutical Development, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Raj Suryanarayanan
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
17
|
Ye Y, Huo X, Yin Z. Protein-protein interactions at high concentrations: Effects of ArgHCl and NaCl on the stability, viscosity and aggregation mechanisms of protein solution. Int J Pharm 2021; 601:120535. [PMID: 33811966 DOI: 10.1016/j.ijpharm.2021.120535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/10/2021] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
The aim of this work was to use the diffusion coefficient ration (Dm/Dline) as a parameter to characterize the stability of protein at high concentration, to compare the effects of ArgHCl and NaCl on the interaction of highly concentrated proteins under different pH conditions, and to explore the correlation with protein stability. For this purpose, a high-concentration bovine serum albumin solution (BSA) was selected as the model system, and the diffusion coefficient, aggregation degree, conformational stability, and solution viscosity of the protein were studied by dynamic light scattering (DLS) and spectral detection techniques. The result showed that there was a significant correlation between the Dm/Dline and the protein aggregation. The Dm/Dline of the protein was minimum at pH 7.4, which corresponded to the maximum degree of aggregation and the highest solution viscosity. At pH 7.4, the hydrophobic interactions and the increased conformational stability of ArgHCl maximized the stability of the protein and reduced the viscosity of the solution by 69.3%. At pH 3.0, the strong charge shielding effect of ArgHCl and NaCl and the decreased conformational stability induced protein aggregation and the gel formation. These findings provided valuable insights into the mechanism of protein aggregation and the diffusion coefficient ration (Dm/Dline) could be a potential tool for the pre-formulation studies.
Collapse
Affiliation(s)
- Yalin Ye
- Key Laboratory of Drug Targeting and Novel Drug Delivery System Ministry of Education, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Xingli Huo
- Key Laboratory of Drug Targeting and Novel Drug Delivery System Ministry of Education, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China
| | - Zongning Yin
- Key Laboratory of Drug Targeting and Novel Drug Delivery System Ministry of Education, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
18
|
Ohshita N, Motoyama K, Iohara D, Hirayama F, Taharabaru T, Watabe N, Kawabata Y, Onodera R, Higashi T. Polypseudorotaxane-based supramolecular hydrogels consisting of cyclodextrins and Pluronics as stabilizing agents for antibody drugs. Carbohydr Polym 2021; 256:117419. [PMID: 33483011 DOI: 10.1016/j.carbpol.2020.117419] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/27/2020] [Accepted: 11/16/2020] [Indexed: 11/18/2022]
Abstract
Recently, antibody drugs have been used worldwide, and based on worldwide sales, 7 of the top 10 pharmaceutical products in 2019 were antibody-based drugs. However, antibody drugs often form aggregates upon thermal and shaking stresses with few efficient stabilizing agents against both stresses. Herein, we developed polypseudorotaxane (PpRX) hydrogels consisting of cyclodextrins (CyDs) and polyethylene glycol (PEG)-polypropylene glycol (PPG)-PEG block copolymers (Pluronics F108, F87, F68, and L44), and evaluated their utility as antibody stabilizing agents. α- and γ-CyDs formed PpRX hydrogels with Pluronics, where CyD/F108 gels showed remarkable stabilizing effects for human immunoglobulin G (IgG) against both thermal and shaking stresses beyond CyD/PEG gels or generic gels. The effects were probably due to the interaction between IgG and the free PPG block of Pluronic F108, resulting in the strong IgG retention in the gels. These findings suggest the great potential of CyD/Pluronic gels as pharmaceutical materials for antibody formulations.
Collapse
Affiliation(s)
- Naoko Ohshita
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Daisuke Iohara
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Fumitoshi Hirayama
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 860-0082, Japan
| | - Toru Taharabaru
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Naoki Watabe
- Renishaw plc. 4-29-8 Yotsuya, Shinjuku-ku, Tokyo 160-0004, Japan
| | - Youhei Kawabata
- Renishaw plc. 4-29-8 Yotsuya, Shinjuku-ku, Tokyo 160-0004, Japan; Graduate School of Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| | - Risako Onodera
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Priority Organization for Innovation and Excellence, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| |
Collapse
|
19
|
Powell T, Knight MJ, Wood A, O'Hara J, Burkitt W. Photoinduced cross-linking of formulation buffer amino acids to monoclonal antibodies. Eur J Pharm Biopharm 2021; 160:35-41. [PMID: 33508437 DOI: 10.1016/j.ejpb.2021.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/22/2020] [Accepted: 01/20/2021] [Indexed: 12/27/2022]
Abstract
The correct choice of formulation buffer is a critical aspect of drug development and is chosen primarily to improve the stability of a protein therapeutic and protect against degradation. Amino acids are frequently incorporated into formulation buffers. In this study we have identified and characterized light induced cross-links between the side chain of histidine residues in an IgG4 monoclonal antibody and different amino acids commonly used in formulation buffers. These reactions have the potential to impact the overall product quality of the drug. The structure of each cross-link identified was elucidated using high performance liquid chromatography (HPLC) hyphenated to tandem mass spectrometry (MS/MS) with higher energy collisional dissociation (HCD). Furthermore, we speculate on the role of amino acids in formulation buffers and their influence on mAb stability. We theorize that whilst the adduction of formulation buffer amino acids could have a negative impact on product quality, it may protect against other pathways of photo-degradation.
Collapse
Affiliation(s)
- Thomas Powell
- Biomolecular Formulation and Characterization Sciences, UCB, Slough SL3WE, UK.
| | - Michael J Knight
- Biomolecular Formulation and Characterization Sciences, UCB, Slough SL3WE, UK
| | - Amanda Wood
- Biomolecular Formulation and Characterization Sciences, UCB, Slough SL3WE, UK
| | - John O'Hara
- Biomolecular Formulation and Characterization Sciences, UCB, Slough SL3WE, UK
| | - William Burkitt
- Biomolecular Formulation and Characterization Sciences, UCB, Slough SL3WE, UK
| |
Collapse
|
20
|
Eronina TB, Mikhaylova VV, Chebotareva NA, Shubin VV, Kleymenov SY, Kurganov BI. Effect of arginine on stability and aggregation of muscle glycogen phosphorylase b. Int J Biol Macromol 2020; 165:365-374. [PMID: 32961195 DOI: 10.1016/j.ijbiomac.2020.09.101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 11/26/2022]
Abstract
Arginine (Arg) is frequently used in biotechnology and pharmaceutics to stabilize protein preparations. When using charged ions like Arg, it is necessary to take into account their contribution to the increase in ionic strength, in addition to the effect of Arg on particular processes occurring under the conditions of constancy of ionic strength. Here, we examined contribution of ionic strength (0.15 and 0.5 M) to the effects of Arg on denaturation, thermal inactivation and aggregation of skeletal muscle glycogen phosphorylase b (Phb). Dynamic light scattering, analytical ultracentrifugation, differential scanning calorimetry, circular dichroism and enzymatic activity assay were used to assess the effects of Arg at constant ionic strength compared with the effects of ionic strength alone. We found that high ionic strength did not affect the secondary structure of Phb, but changed conformation of the protein. Such a destabilization of the enzyme causes an increase in the initial rate of aggregation and inactivation of Phb thereby affecting its denaturation. Binding of Arg causes additional changes in the protein conformation, weakening the bonds between monomers in the dimer. This causes the dimer to dissociate into monomers, which rapidly aggregate. Thus, Arg acts on these processes much stronger than just ionic strength.
Collapse
Affiliation(s)
- Tatiana B Eronina
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Sciences, Leninsky pr. 33, Moscow 119071, Russia
| | - Valeriya V Mikhaylova
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Sciences, Leninsky pr. 33, Moscow 119071, Russia
| | - Natalia A Chebotareva
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Sciences, Leninsky pr. 33, Moscow 119071, Russia
| | - Vladimir V Shubin
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Sciences, Leninsky pr. 33, Moscow 119071, Russia
| | - Sergey Y Kleymenov
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Sciences, Leninsky pr. 33, Moscow 119071, Russia; Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova 26, Moscow 119991, Russia
| | - Boris I Kurganov
- Bach Institute of Biochemistry, Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Sciences, Leninsky pr. 33, Moscow 119071, Russia.
| |
Collapse
|
21
|
Reslan M, Sifniotis V, Cruz E, Sumer-Bayraktar Z, Cordwell S, Kayser V. Enhancing the stability of adalimumab by engineering additional glycosylation motifs. Int J Biol Macromol 2020; 158:189-196. [PMID: 32360204 DOI: 10.1016/j.ijbiomac.2020.04.147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/23/2020] [Accepted: 04/18/2020] [Indexed: 12/23/2022]
Abstract
Monoclonal antibodies (mAbs) are of high value in the diagnostic and treatment of many debilitating diseases such as cancers, auto-immune disorders and infections. Unfortunately, protein aggregation is one of the ongoing challenges, limiting the development and application of mAbs as therapeutic products by decreasing half-life, increasing immunogenicity and reducing activity. We engineered an aggregation-prone region of adalimumab, the top selling mAb product worldwide - with additional glycosylation sites to enhance its resistance to aggregation by steric hindrance as a next generation biologic. We found that the addition of N-glycans in the Fab domain significantly enhanced its conformational stability, with some variants increasing the melting temperature of the Fab domain by >6 °C. The mutations tested had minimal impact on antigen binding affinity, or affinity to Fcγ receptors responsible for effector function. Our findings highlight the significant utility of this rational engineering approach for enhancing the conformational stability of therapeutic mAbs and other next-generation antibody formats.
Collapse
Affiliation(s)
- Mouhamad Reslan
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | - Vicki Sifniotis
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | - Esteban Cruz
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| | - Zeynep Sumer-Bayraktar
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
| | - Stuart Cordwell
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
| | - Veysel Kayser
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
22
|
Binding of excipients is a poor predictor for aggregation kinetics of biopharmaceutical proteins. Eur J Pharm Biopharm 2020; 151:127-136. [PMID: 32283214 DOI: 10.1016/j.ejpb.2020.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 12/11/2022]
Abstract
One of the major challenges in formulation development of biopharmaceuticals is improving long-term storage stability, which is often achieved by addition of excipients to the final formulation. Finding the optimal excipient for a given protein is usually done using a trial-and-error approach, due to the lack of general understanding of how excipients work for a particular protein. Previously, preferential interactions (binding or exclusion) of excipients with proteins were postulated as a mechanism explaining diversity in the stabilisation effects. Weak preferential binding is however difficult to quantify experimentally, and the question remains whether the formulation process should seek excipients which preferentially bind with proteins, or not. Here, we apply solution NMR spectroscopy to comprehensively evaluate protein-excipient interactions between therapeutically relevant proteins and commonly used excipients. Additionally, we evaluate the effect of excipients on thermal and colloidal protein stability, on aggregation kinetics and protein storage stability at elevated temperatures. We show that there is a weak negative correlation between the strength of protein-excipient interactions and effect on enhancing protein thermal stability. We found that the overall protein-excipient binding per se can be a poor criterion for choosing excipients enhancing formulation stability. Experiments on a diverse set of excipients and test proteins reveal that while excipients affect all of the different aspects of protein stability, the effects are very much protein specific, and care must be taken to avoid apparent generalisations if a smaller dataset is being used.
Collapse
|
23
|
Le Basle Y, Chennell P, Tokhadze N, Astier A, Sautou V. Physicochemical Stability of Monoclonal Antibodies: A Review. J Pharm Sci 2020; 109:169-190. [DOI: 10.1016/j.xphs.2019.08.009] [Citation(s) in RCA: 258] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 01/10/2023]
|
24
|
Orthogonal Techniques to Study the Effect of pH, Sucrose, and Arginine Salts on Monoclonal Antibody Physical Stability and Aggregation During Long-Term Storage. J Pharm Sci 2020; 109:584-594. [DOI: 10.1016/j.xphs.2019.10.065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/14/2019] [Accepted: 10/31/2019] [Indexed: 11/18/2022]
|
25
|
Qin K, Shi W, Zhao L, Li M, Tang Y, Faridoon, Jiang B, Tang F, Huang W. Thermostability detection and optimization of glycoengineered antibodies and antibody-drug conjugates based on differential scanning flouremitry analysis. Bioorg Chem 2020; 94:103391. [DOI: 10.1016/j.bioorg.2019.103391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 01/02/2023]
|
26
|
Xu AY, Castellanos MM, Mattison K, Krueger S, Curtis JE. Studying Excipient Modulated Physical Stability and Viscosity of Monoclonal Antibody Formulations Using Small-Angle Scattering. Mol Pharm 2019; 16:4319-4338. [DOI: 10.1021/acs.molpharmaceut.9b00687] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Amy Yuanyuan Xu
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, Maryland 20899, United States
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, Maryland 20850, United States
| | - Maria Monica Castellanos
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, Maryland 20899, United States
- Institute for Bioscience and Biotechnology Research, University of Maryland, 9600 Gudelsky Drive, Rockville, Maryland 20850, United States
| | - Kevin Mattison
- Malvern Panalytical, 117 Flanders Road, Westborough, Massachusetts 01581, United States
| | - Susan Krueger
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, Maryland 20899, United States
| | - Joseph E. Curtis
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Mail Stop 6102, Gaithersburg, Maryland 20899, United States
| |
Collapse
|
27
|
Wang W, Ohtake S. Science and art of protein formulation development. Int J Pharm 2019; 568:118505. [PMID: 31306712 DOI: 10.1016/j.ijpharm.2019.118505] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023]
Abstract
Protein pharmaceuticals have become a significant class of marketed drug products and are expected to grow steadily over the next decade. Development of a commercial protein product is, however, a rather complex process. A critical step in this process is formulation development, enabling the final product configuration. A number of challenges still exist in the formulation development process. This review is intended to discuss these challenges, to illustrate the basic formulation development processes, and to compare the options and strategies in practical formulation development.
Collapse
Affiliation(s)
- Wei Wang
- Biological Development, Bayer USA, LLC, 800 Dwight Way, Berkeley, CA 94710, United States.
| | - Satoshi Ohtake
- Pharmaceutical Research and Development, Pfizer Biotherapeutics Pharmaceutical Sciences, Chesterfield, MO 63017, United States
| |
Collapse
|
28
|
Wang W, Roberts CJ. Protein aggregation – Mechanisms, detection, and control. Int J Pharm 2018; 550:251-268. [DOI: 10.1016/j.ijpharm.2018.08.043] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/18/2018] [Accepted: 08/20/2018] [Indexed: 12/19/2022]
|
29
|
Hong T, Iwashita K, Shiraki K. Viscosity Control of Protein Solution by Small Solutes: A Review. Curr Protein Pept Sci 2018; 19:746-758. [PMID: 29237380 PMCID: PMC6182935 DOI: 10.2174/1389203719666171213114919] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 12/22/2022]
Abstract
Viscosity of protein solution is one of the most troublesome issues for the high-concentration formulation of protein drugs. In this review, we summarize the practical methods that suppress the viscosity of protein solution using small molecular additives. The small amount of salts decreases the viscosity that results from electrostatic repulsion and attraction. The chaotrope suppresses the hydrophobic attraction and cluster formation, which can lower the solution viscosity. Arginine hydrochloride (ArgHCl) also suppresses the solution viscosity due to the hydrophobic and aromatic interactions between protein molecules. The small molecular additives are the simplest resolution of the high viscosity of protein solution as well as understanding of the primary cause in complex phenomena of protein interactions.
Collapse
Affiliation(s)
- Taehun Hong
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki305-8573, Japan
| | - Kazuki Iwashita
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki305-8573, Japan
| | - Kentaro Shiraki
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki305-8573, Japan
| |
Collapse
|
30
|
Shtykova EV, Bogacheva EN, Dadinova LA, Jeffries CM, Fedorova NV, Golovko AO, Baratova LA, Batishchev OV. Small-angle X-Ray analysis of macromolecular structure: the structure of protein NS2 (NEP) in solution. CRYSTALLOGR REP+ 2017. [DOI: 10.1134/s1063774517060220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Wang S, Zhang X, Wu G, Tian Z, Qian F. Optimization of high-concentration endostatin formulation: Harmonization of excipients' contributions on colloidal and conformational stabilities. Int J Pharm 2017; 530:173-186. [PMID: 28755991 DOI: 10.1016/j.ijpharm.2017.07.057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/05/2017] [Accepted: 07/19/2017] [Indexed: 12/24/2022]
Abstract
Recently, increasing research efforts have been devoted into developing high-concentration protein drugs for subcutaneous injection, especially for those with short half-lives and high-dose requirement. Proteins at high concentrations normally present increased colloidal and structural instability, such as aggregation, fibrillation and gelation, which significantly challenges the high-concentration formulation development of protein drugs. Here we used endostatin, a 20kD recombinant protein, as a model drug for high-concentration formulation optimization. The colloidal and conformational stability of endostatin at high concentration of 30mg/mL were investigated in formulations containing various excipients, including saccharides (mannitol, sorbitol and sucrose), salts (ArgHCl and NaCl), and surfactants (tween 20 and 80). Protein fibrillation was characterized and semi-quantified by optical polarized light microscopy and transmission electron microscopy, and the amount of fiber formation at elevated temperature of 40°C was determined. The soluble protein aggregates were characterized by dynamic and static light scattering before and after dilution. The conformational stability were characterized by polyacrylamide gel electrophoresis, fluorescence, circular dichroism, and differential scanning calorimetry. We observed that the soluble aggregation, fibrillation and gelation, induced by conformational and colloidal instabilities of the protein solution, could be substantially optimized by using suitable stabilizers such as combinations of saccharides and surfactants; while formation of gel and soluble aggregates at high protein concentration (e.g., 30mg/mL) and elevated temperature (40°C) could be prevented by avoiding the usage of salts. It's worth emphasizing that some stabilizers, such as salts and surfactants, could show opposite contributions in conformational and colloidal stabilities of endostatin. Therefore, cautions are needed when one attempts to correlate the colloidal stability of high-concentration proteins with their conformational stability, and the colloidal and conformational protein stabilities must be harmonized by a balanced selection of various types of excipients.
Collapse
Affiliation(s)
- Shujing Wang
- School of Pharmaceutical Sciences & Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Xinyi Zhang
- School of Pharmaceutical Sciences & Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Guoliang Wu
- School of Pharmaceutical Sciences & Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Zhou Tian
- School of Pharmaceutical Sciences & Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China
| | - Feng Qian
- School of Pharmaceutical Sciences & Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
32
|
Dipicolinic acid as a novel spore-inspired excipient for antibody formulation. Int J Pharm 2017; 526:332-338. [PMID: 28495581 DOI: 10.1016/j.ijpharm.2017.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/20/2017] [Accepted: 05/05/2017] [Indexed: 01/02/2023]
Abstract
Ionic excipients are commonly used in aqueous therapeutic monoclonal antibody (mAb) formulations. Novel excipients are of industrial interest, with a recent focus on Arg salt forms and their application as viscosity reducing and stabilizing additives. Here, we report that the calcium salt of dipicolinic acid (DPA, pyridine-2,6-dicarboxylic acid), uniquely present in nature in the core of certain bacterial spores, reduces the viscosity of a mAb formulated at 150mg/mL, below that achieved by Arg hydrochloride at the same concentration (10mM). DPA also reduced the reversible phase separation of the same formulation, which characteristically occurs for this mAb upon cooling to 4°C. Differential scanning calorimetry and differential scanning fluorimetry did not reveal a conformation destabilisation of the mAb in the presence of 10mM DPA, or by the related quinolinic acid (QA, pyridine-2,3-dicarboxylic acid). However, fluorescence spectrophotometry did reveal localised (aromatic) conformational changes to the mAb attributed to DPA, dependent on the salt form. While precise mechanisms of action remain to be identified, our preliminary data suggest that these DPA salts are worthy of further investigation as novel ionic excipient for biologics formulation.
Collapse
|
33
|
Kheddo P, Bramham JE, Dearman RJ, Uddin S, van der Walle CF, Golovanov AP. Investigating Liquid–Liquid Phase Separation of a Monoclonal Antibody Using Solution-State NMR Spectroscopy: Effect of Arg·Glu and Arg·HCl. Mol Pharm 2017; 14:2852-2860. [DOI: 10.1021/acs.molpharmaceut.7b00418] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Priscilla Kheddo
- Manchester
Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN, U.K
| | - Jack E. Bramham
- Manchester
Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN, U.K
| | - Rebecca J. Dearman
- School
of Biological Sciences, The University of Manchester, Manchester, M13 9PL, U.K
| | - Shahid Uddin
- Formulation
Sciences, MedImmune Ltd., Aaron Klug Building, Granta Park, Cambridge, CB21 6GH, U.K
| | | | - Alexander P. Golovanov
- Manchester
Institute of Biotechnology and School of Chemistry, The University of Manchester, Manchester, M1 7DN, U.K
| |
Collapse
|