1
|
Pujahari SR, Engla S, Soni R, Patra S, Hanawal MK, Kumar A. Structural Similarity of Biological Drugs Using Statistical Signal Processing and Nuclear Magnetic Resonance Spectral Pattern Analysis. Mol Pharm 2025; 22:2684-2693. [PMID: 40198793 DOI: 10.1021/acs.molpharmaceut.5c00108] [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] [Indexed: 04/10/2025]
Abstract
Biosimilar drugs are highly similar to the available marketed drugs and have no clinically meaningful differences in terms of safety, purity, and potency. As per stringent drug regulatory requirements, biosimilar drugs must match closely to all attributes of the listed marketed drug, including establishing high similarity of higher-order structures. Here, we have developed a combined approach using high-resolution two-dimensional nuclear magnetic resonance (NMR) spectra and image-based statistical signal processing algorithms to establish robust comparability of critical quality attributes of biological drugs. We have integrated a computational approach to 2D NMR data analysis, which could replace the traditional methods of manually extracting chemical shift values and intensities for each peak and performing a range of statistical analyses, which are laborious and prone to ambiguity. Our algorithm simplifies and streamlines this process, making it more accurate, less time-consuming, and avoiding personal biases. We have employed our methods with a diverse range of biotherapeutics and complex NMR data and shown a degree of similarity between reference and test drugs with our differentially assigned similarity scores.
Collapse
Affiliation(s)
- Soumya Ranjan Pujahari
- Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Swpnil Engla
- Industrial Engineering and Operations Research, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Rohit Soni
- Industrial Engineering and Operations Research, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Subrata Patra
- Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Manjesh Kumar Hanawal
- Industrial Engineering and Operations Research, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Ashutosh Kumar
- Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| |
Collapse
|
2
|
Roamcharern N, Matthew SAL, Brady DJ, Parkinson JA, Rattray Z, Seib FP. Biomimetic Silk Nanoparticle Manufacture: Calcium Ion-Mediated Assembly. ACS Biomater Sci Eng 2025; 11:1847-1856. [PMID: 39883858 PMCID: PMC11897946 DOI: 10.1021/acsbiomaterials.4c02175] [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] [Received: 11/18/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Silk has emerged as an interesting candidate among protein-based nanocarriers due to its favorable properties, including biocompatibility and a broad spectrum of processing options to tune particle critical quality attributes. The silk protein conformation during storage in the middle silk gland of the silkworm is modulated by various factors, including the most abundant metallic ion, calcium ion (Ca2+). Here, we report spiking of liquid silk with calcium ions to modulate the silk nanoparticle size. Conformational and structural analyses of silk demonstrated Ca2+-induced silk assemblies that resulted in a liquid crystalline-like state, with the subsequent generation of β-sheet-enriched silk nanoparticles. Thioflavin T studies demonstrated that Ca2+ effectively induces self-assembly and conformation changes that also increased model drug loading. Ca2+ incorporation in the biopolymer feed significantly increased the nanoparticle production yield from 16 to 89%, while simultaneously enabling Ca2+ concentration-dependent particle-size tuning with a narrow polydispersity index and altered zeta potential. The resulting silk nanoparticles displayed high biocompatibility in macrophages with baseline levels of cytotoxicity and cellular inflammation. Our strategy for manufacturing biomimetic silk nanoparticles enabled overall tuning of particle size and improved yields─features that are critical for particle-based nanomedicines.
Collapse
Affiliation(s)
- Napaporn Roamcharern
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St., Glasgow G4 0RE,Scotland,U.K.
| | - Saphia A. L. Matthew
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St., Glasgow G4 0RE,Scotland,U.K.
| | - Daniel J. Brady
- Branch Bioresources, Fraunhofer Institute for Molecular Biology and Applied
Ecology, Ohlebergsweg
12, Giessen 35392, Germany
| | - John A. Parkinson
- Department
of Pure and Applied Chemistry, University
of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, Scotland,U.K.
| | - Zahra Rattray
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St., Glasgow G4 0RE,Scotland,U.K.
| | - F. Philipp Seib
- Strathclyde
Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral St., Glasgow G4 0RE,Scotland,U.K.
- Branch Bioresources, Fraunhofer Institute for Molecular Biology and Applied
Ecology, Ohlebergsweg
12, Giessen 35392, Germany
- Institute
of Pharmacy, Department of Pharmaceutics and Biopharmaceutics, Friedrich Schiller University Jena, Lessingstr. 8, Jena 07743, Germany
| |
Collapse
|
3
|
Xu X, Poggetto GD, McCoy M, Reibarkh M, Trigo-Mourino P. Rapid Characterization of Structural and Behavioral Changes of Therapeutic Proteins by Relaxation and Diffusion 1H-SOFAST NMR Experiments. Anal Chem 2024; 96:16322-16329. [PMID: 39356572 DOI: 10.1021/acs.analchem.4c03479] [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: 10/04/2024]
Abstract
Biologic drugs have emerged as a rapidly expanding and important modality, offering promising therapeutic solutions by interacting with previously "undruggable" targets, thus significantly expanding the range of modern pharmaceutical applications. However, the inherent complexity of these drugs also introduces liabilities and poses challenges in their development, necessitating efficient screening methods to evaluate the structural stability and behavior. Although nuclear magnetic resonance (NMR) spectroscopy is well-suited for detecting weak interactions, changes in dynamics, high-order structure, and association states of macromolecules in fully formulated samples, the inherent low sensitivity limits its utility as a fast screening and characterization tool. In this study, we present two fast pulsing NMR experiments, namely the band-Selective Optimized Flip-Angle Internally encoded Relaxation (SOFAIR) and the band-Selective Optimized Flip-angle Internally encoded Translational diffusion (SOFIT)), which enable rapid and reliable measurements of transverse relaxation rates and diffusion coefficients with more than 10-fold higher sensitivity compared to commonly used methods, like Carr-Purcell-Meiboom-Gill and diffusion-ordered spectroscopy, allowing the rapid assessment of biologics even at low concentrations. We demonstrated the effectiveness and versatility of these experiments by evaluating several examples, including thermally stressed proteins, proteins at different concentrations, and a therapeutic protein in various formulations. We anticipate that these novel approaches will greatly facilitate the analysis and characterization of biologics during drug discovery.
Collapse
Affiliation(s)
- Xingjian Xu
- Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Guilherme Dal Poggetto
- Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Mark McCoy
- Quantitative Biosciences, MRL, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Mikhail Reibarkh
- Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Pablo Trigo-Mourino
- Analytical Research & Development, MRL, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| |
Collapse
|
4
|
Bramham JE, Wang Y, Moore SA, Golovanov AP. Assessing Photostability of mAb Formulations In Situ Using Light-Coupled NMR Spectroscopy. Anal Chem 2024; 96:9935-9943. [PMID: 38847283 PMCID: PMC11190875 DOI: 10.1021/acs.analchem.4c01164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/29/2024] [Accepted: 05/29/2024] [Indexed: 06/19/2024]
Abstract
Biopharmaceuticals, such as monoclonal antibodies (mAbs), need to maintain their chemical and physical stability in formulations throughout their lifecycle. It is known that exposure of mAbs to light, particularly UV, triggers chemical and physical degradation, which can be exacerbated by trace amounts of photosensitizers in the formulation. Although routine assessments of degradation following defined UV dosages are performed, there is a fundamental lack of understanding regarding the intermediates, transient reactive species, and radicals formed during illumination, as well as their lifetimes and immediate impact post-illumination. In this study, we used light-coupled NMR spectroscopy to monitor in situ live spectral changes in sealed samples during and after UV-A illumination of different formulations of four mAbs without added photosensitizers. We observed a complex evolution of spectra, reflecting the appearance within minutes of transient radicals during illumination and persisting for minutes to tens of minutes after the light was switched off. Both mAb and excipient signals were strongly affected by illumination, with some exhibiting fast irreversible photodegradation and others exhibiting partial recovery in the dark. These effects varied depending on the mAb and the presence of excipients, such as polysorbate 80 (PS80) and methionine. Complementary ex situ high-performance size-exclusion chromatography analysis of the same formulations post-UV exposure in the chamber revealed significant loss of purity, confirming formulation-dependent degradation. Both approaches suggested the presence of degradation processes initiated by light but continuing in the dark. Further studies on photoreaction intermediates and transient reactive species may help mitigate the impact of light on biopharmaceutical degradation.
Collapse
Affiliation(s)
- Jack E. Bramham
- Department
of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M1 7DN, U.K.
| | - Yujing Wang
- Dosage
Form Design & Development, BioPharmaceutical
Development, R&D, AstraZeneca, Cambridge CB2 0AA, U.K.
| | - Stephanie A. Moore
- Dosage
Form Design & Development, BioPharmaceutical
Development, R&D, AstraZeneca, Cambridge CB2 0AA, U.K.
| | - Alexander P. Golovanov
- Department
of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester M1 7DN, U.K.
| |
Collapse
|
5
|
Li M, Beaumont VA, Akbar S, Duncan H, Creasy A, Wang W, Sackett K, Marzilli L, Rouse JC, Kim HY. Comprehensive characterization of higher order structure changes in methionine oxidized monoclonal antibodies via NMR chemometric analysis and biophysical approaches. MAbs 2024; 16:2292688. [PMID: 38117548 PMCID: PMC10761137 DOI: 10.1080/19420862.2023.2292688] [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: 10/19/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023] Open
Abstract
The higher order structure (HOS) of monoclonal antibodies (mAbs) is an important quality attribute with strong contribution to clinically relevant biological functions and drug safety. Due to the multi-faceted nature of HOS, the synergy of multiple complementary analytical approaches can substantially improve the understanding, accuracy, and resolution of HOS characterization. In this study, we applied one- and two-dimensional (1D and 2D) nuclear magnetic resonance (NMR) spectroscopy coupled with chemometric analysis, as well as circular dichroism (CD), differential scanning calorimetry (DSC), and fluorescence spectroscopy as orthogonal methods, to characterize the impact of methionine (Met) oxidation on the HOS of an IgG1 mAb. We used a forced degradation method involving concentration-dependent oxidation by peracetic acid, in which Met oxidation is site-specifically quantified by liquid chromatography-mass spectrometry. Conventional biophysical techniques report nuanced results, in which CD detects no change to the secondary structure and little change in the tertiary structure. Yet, DSC measurements show the destabilization of Fab and Fc domains due to Met oxidation. More importantly, our study demonstrates that 1D and 2D NMR and chemometric analysis can provide semi-quantitative analysis of chemical modifications and resolve localized conformational changes with high sensitivity. Furthermore, we leveraged a novel 15N-Met labeling technique of the antibody to directly observe structural perturbations at the oxidation sites. The NMR methods described here to probe HOS changes are highly reliable and practical in biopharmaceutical characterization.
Collapse
Affiliation(s)
- Mingyue Li
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Victor A. Beaumont
- Pfizer, Inc. Pharmaceutical Sciences Small Molecules, Analytical Research and Development, Sandwich, United Kingdom
| | - Shahajahan Akbar
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Hannah Duncan
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Arch Creasy
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Bioprocess Research and Development, Andover, MA, USA
| | - Wenge Wang
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Bioprocess Research and Development, Andover, MA, USA
| | - Kelly Sackett
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Lisa Marzilli
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Jason C. Rouse
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| | - Hai-Young Kim
- Pfizer, Inc. BioTherapeutics Pharmaceutical Sciences, Analytical Research and Development, Andover, MA, USA
| |
Collapse
|
6
|
Pujahari SR, Mali PS, Purusottam RN, Kumar A. Combined Liquid-State and Solid-State Nuclear Magnetic Resonance at Natural Abundance for Comparative Higher Order Structure Assessment in the Formulated-State of Biphasic Biopharmaceutics. Anal Chem 2023. [PMID: 37154614 DOI: 10.1021/acs.analchem.2c05485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
A higher-order structure (HOS) is critical to a biopharmaceutical drug as the three-dimensional structure governs its function. Even the partial perturbation in the HOS of the drug can alter the biological efficiency and efficacy. Due to current limitations in analytical technologies, it is imperative to develop a protocol to characterize the HOS of biopharmaceuticals in the native formulated state. This becomes even more challenging for the suspension formulations where solution and solid phases co-exist. Here, we have used a combinatorial approach using liquid (1D 1H) and solid-state (13C CP MAS) NMR methodology to demonstrate the HOS in the biphasic microcrystalline suspension drug in its formulated state. The data were further assessed by principal component analysis and Mahalanobis distance (DM) calculation for quantitative assessment. This approach is sufficient to provide information regarding the protein HOS and the local dynamics of the molecule when combined with orthogonal techniques such as X-ray scattering. Our method can be an elegant tool to investigate batch-to-batch variation in the process of manufacture and storage as well as a biosimilarity comparison study for biphasic/microcrystalline suspension.
Collapse
Affiliation(s)
| | - Pramod S Mali
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Rudra N Purusottam
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ashutosh Kumar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
7
|
Alhazmi HA, Albratty M. Analytical Techniques for the Characterization and Quantification of Monoclonal Antibodies. Pharmaceuticals (Basel) 2023; 16:291. [PMID: 37259434 PMCID: PMC9967501 DOI: 10.3390/ph16020291] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 08/12/2023] Open
Abstract
Monoclonal antibodies (mAbs) are a fast-growing class of biopharmaceuticals. They are widely used in the identification and detection of cell makers, serum analytes, and pathogenic agents, and are remarkably used for the cure of autoimmune diseases, infectious diseases, or malignancies. The successful application of therapeutic mAbs is based on their ability to precisely interact with their appropriate target sites. The precision of mAbs rely on the isolation techniques delivering pure, consistent, stable, and safe lots that can be used for analytical, diagnostic, or therapeutic applications. During the creation of a biologic, the key quality features of a particular mAb, such as structure, post-translational modifications, and activities at the biomolecular and cellular levels, must be characterized and profiled in great detail. This implies the requirement of powerful state of the art analytical techniques for quality control and characterization of mAbs. Until now, various analytical techniques have been developed to characterize and quantify the mAbs according to the regulatory guidelines. The present review summarizes the major techniques used for the analyses of mAbs which include chromatographic, electrophoretic, spectroscopic, and electrochemical methods in addition to the modifications in these methods for improving the quality of mAbs. This compilation of major analytical techniques will help students and researchers to have an overview of the methodologies employed by the biopharmaceutical industry for structural characterization of mAbs for eventual release of therapeutics in the drug market.
Collapse
Affiliation(s)
- Hassan A. Alhazmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
8
|
Ó'Fágáin C. Protein Stability: Enhancement and Measurement. Methods Mol Biol 2023; 2699:369-419. [PMID: 37647007 DOI: 10.1007/978-1-0716-3362-5_18] [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] [Indexed: 09/01/2023]
Abstract
This chapter defines protein stability, emphasizes its importance, and surveys the field of protein stabilization, with summary reference to a selection of 2014-2021 publications. One can enhance stability, particularly by protein engineering strategies but also by chemical modification and by other means. General protocols are set out on how to measure a given protein's (i) kinetic thermal stability and (ii) oxidative stability and (iii) how to undertake chemical modification of a protein in solution.
Collapse
Affiliation(s)
- Ciarán Ó'Fágáin
- School of Biotechnology, Dublin City University, Dublin, Ireland.
| |
Collapse
|
9
|
Ó'Fágáin C, Colliton K. Storage and Lyophilization of Pure Proteins. Methods Mol Biol 2023; 2699:421-475. [PMID: 37647008 DOI: 10.1007/978-1-0716-3362-5_19] [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] [Indexed: 09/01/2023]
Abstract
This chapter outlines empirical procedures for the storage of pure proteins with preservation of high levels of biological activity. It describes simple and workable means of preventing microbial contamination and proteolytic degradation and the use of various types of stabilizing additives. It sets out the principles of lyophilization (a complex process comprising freezing, primary drying, and secondary drying stages, otherwise known as freeze-drying). There follows a general procedure for the use of lyophilizer apparatus with emphasis on best practice and on pitfalls to avoid. The use of modulated differential scanning calorimetry to measure the glass transition temperature, a key parameter in the design and successful operation of lyophilization processes, is described. This chapter concludes with brief summaries of interesting recent work in the field.
Collapse
Affiliation(s)
- Ciarán Ó'Fágáin
- School of Biotechnology, Dublin City University, Dublin, Ireland.
| | - Keith Colliton
- Pfizer Ireland Pharmaceuticals, Grange Castle Business Park, Dublin, Ireland
| |
Collapse
|
10
|
Hickey AJ, Stewart IE. Inhaled antibodies: Quality and performance considerations. Hum Vaccin Immunother 2022; 18:1940650. [PMID: 34191682 PMCID: PMC9116391 DOI: 10.1080/21645515.2021.1940650] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/11/2021] [Accepted: 06/05/2021] [Indexed: 12/22/2022] Open
Abstract
The use of antibodies in the treatment of lung diseases is of increasing interest especially as the search for COVID-19 therapies has unfolded. Historically, the use of antibody therapy was based on multiple targets including receptors involved in local hyper-reactivity in asthma, viruses and micro-organisms involved in a variety of pulmonary infectious disease. Generally, protein therapeutics pose challenges with respect to formulation and delivery to retain activity and assure therapy. The specificity of antibodies amplifies the need for attention to molecular integrity not only in formulation but also during aerosol delivery for pulmonary administration. Drug product development can be viewed from considerations of route of administration, dosage form, quality, and performance measures. Nebulizers and dry powder inhalers have been used to deliver protein therapeutics and each has its advantages that should be matched to the needs of the drug and the disease. This review offers insight into quality and performance barriers and the opportunities that arise from meeting them effectively.
Collapse
|
11
|
Hjalte J, Hossain S, Hugerth A, Sjögren H, Wahlgren M, Larsson P, Lundberg D. Aggregation Behavior of Structurally Similar Therapeutic Peptides Investigated by 1H NMR and All-Atom Molecular Dynamics Simulations. Mol Pharm 2022; 19:904-917. [PMID: 35104408 PMCID: PMC8905580 DOI: 10.1021/acs.molpharmaceut.1c00883] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 11/30/2022]
Abstract
Understanding of peptide aggregation propensity is an important aspect in pharmaceutical development of peptide drugs. In this work, methodologies based on all-atom molecular dynamics (AA-MD) simulations and 1H NMR (in neat H2O) were evaluated as tools for identification and investigation of peptide aggregation. A series of structurally similar, pharmaceutically relevant peptides with known differences in aggregation behavior (D-Phe6-GnRH, ozarelix, cetrorelix, and degarelix) were investigated. The 1H NMR methodology was used to systematically investigate variations in aggregation with peptide concentration and time. Results show that 1H NMR can be used to detect the presence of coexisting classes of aggregates and the inclusion or exclusion of counterions in peptide aggregates. Interestingly, results suggest that the acetate counterions are included in aggregates of ozarelix and cetrorelix but not in aggregates of degarelix. The peptides investigated in AA-MD simulations (D-Phe6-GnRH, ozarelix, and cetrorelix) showed the same rank order of aggregation propensity as in the NMR experiments. The AA-MD simulations also provided molecular-level insights into aggregation dynamics, aggregation pathways, and the influence of different structural elements on peptide aggregation propensity and intermolecular interactions within the aggregates. Taken together, the findings from this study illustrate that 1H NMR and AA-MD simulations can be useful, complementary tools in early evaluation of aggregation propensity and formulation development for peptide drugs.
Collapse
Affiliation(s)
- Johanna Hjalte
- Food
Technology, Engineering and Nutrition, Lund
University, Box 124, 221 00 Lund, Sweden
| | - Shakhawath Hossain
- Department
of Pharmacy, Drug Delivery, Uppsala University, Box 580, 751 23 Uppsala, Sweden
| | - Andreas Hugerth
- Ferring
Pharmaceuticals A/S, Amager Strandvej 405, 2770 Kastrup, Denmark
| | - Helen Sjögren
- Ferring
Pharmaceuticals A/S, Amager Strandvej 405, 2770 Kastrup, Denmark
| | - Marie Wahlgren
- Food
Technology, Engineering and Nutrition, Lund
University, Box 124, 221 00 Lund, Sweden
| | - Per Larsson
- Department
of Pharmacy, Drug Delivery, Uppsala University, Box 580, 751 23 Uppsala, Sweden
| | - Dan Lundberg
- CR
Competence AB, Center for Chemistry and Chemical Engineering, Box 124, 221 00 Lund, Sweden
| |
Collapse
|
12
|
Panchal J, Falk BT, Antochshuk V, McCoy MA. Investigating protein-excipient interactions of a multivalent V HH therapeutic protein using NMR spectroscopy. MAbs 2022; 14:2124902. [PMID: 36166705 PMCID: PMC9519013 DOI: 10.1080/19420862.2022.2124902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Multispecific therapeutic proteins come in a variety of formats, including bi- and tri-specific antibodies, dual-variable domain antibodies, and CrossMabs. These multivalent proteins are engineered to interact with multiple therapeutic target proteins with high specificity. Multi-domain proteins can be created by linking together a variety of high-affinity antibody fragments. The choice of protein domains and linkers not only affects the interactions of these molecules with therapeutic targets but also influences the intrinsic behavior in solution that affects their stability. The complexity of solution interactions may translate into developability and manufacturing challenges. Here, we use nuclear magnetic resonance (NMR) spectroscopy to study the solution behavior of a multivalent VHH molecule composed of three flexibly linked heavy-chain-only domains that show dramatic stabilization against thermal degradation in the presence of sucrose. A collection of NMR fingerprinting and profiling methods were used to simultaneously monitor the protein solution behavior and capture details of protein–excipient interactions. We provide a framework to characterize and begin to understand the role of molecular flexibility in protein stabilization with potential applications in the design of novel therapeutic protein scaffolds that include multivalent proteins, fusion proteins, antibody-drug conjugates, and proteins modified with flexible lipids.
Collapse
Affiliation(s)
- Jainik Panchal
- Sterile and Specialty Products, Merck & Co Inc, Kenilworth, NJ (United States)
| | - Bradley T Falk
- Mass Spectrometry and Biophysics, Merck & Co Inc, Kenilworth, NJ United States
| | - Valentyn Antochshuk
- Sterile and Specialty Products, Merck & Co Inc, Kenilworth, NJ (United States)
| | - Mark A McCoy
- Mass Spectrometry and Biophysics, Merck & Co Inc, Kenilworth, NJ United States
| |
Collapse
|
13
|
Elliott KW, Delaglio F, Wikström M, Marino JP, Arbogast LW. Principal Component Analysis of 1D 1H Diffusion Edited NMR Spectra of Protein Therapeutics. J Pharm Sci 2021; 110:3385-3394. [PMID: 34166704 PMCID: PMC10408412 DOI: 10.1016/j.xphs.2021.06.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 11/25/2022]
Abstract
The one-dimensional (1D) diffusion edited proton NMR method, Protein Fingerprint by Lineshape Enhancement (PROFILE) has been demonstrated to be suitable for higher order structure (HOS) characterization of protein therapeutics including monoclonal antibodies. Recent reports in the literature have demonstrated its advantages for HOS characterization over traditional methods such as circular dichroism and Fourier-transform infrared spectroscopy. Previously, we have demonstrated that the PROFILE method is complementary with high resolution 2D methyl correlated NMR methods and how both may be deployed as a multi-modal platform to further the utility of NMR for HOS characterization. A major limitation of the PROFILE method remains its need for high signal to noise data due to its reliance on convolution difference processing and linear correlation metrics to assess spectral similarity. Here we present an alternative method for analyzing 1D diffusion edited spectra, which overcomes this limitation by using nonlinear iterative partial least squares (NIPALS) principal component analysis, and which we dub PROtein Fingerprint Observed Using NIPALS Decomposition (PROFOUND). We demonstrate that results from the PROFOUND method are robust with respect to instrument, operator and in the presence of high experimental noise and how it may be employed to provide quantitative assessment of spectral similarity.
Collapse
Affiliation(s)
- Korth W Elliott
- Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology, 9600 Gudelsky Dr. Rockville, MD 20850 USA
| | - Frank Delaglio
- Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology, 9600 Gudelsky Dr. Rockville, MD 20850 USA
| | - Mats Wikström
- Higher Order Structure, Attribute Sciences, Amgen, Inc. One Amgen Center Dr., Thousand Oaks, CA 91320 USA
| | - John P Marino
- Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology, 9600 Gudelsky Dr. Rockville, MD 20850 USA
| | - Luke W Arbogast
- Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology, 9600 Gudelsky Dr. Rockville, MD 20850 USA.
| |
Collapse
|
14
|
NMR Spectroscopy for Protein Higher Order Structure Similarity Assessment in Formulated Drug Products. Molecules 2021; 26:molecules26144251. [PMID: 34299526 PMCID: PMC8307401 DOI: 10.3390/molecules26144251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 11/21/2022] Open
Abstract
Peptide and protein drug molecules fold into higher order structures (HOS) in formulation and these folded structures are often critical for drug efficacy and safety. Generic or biosimilar drug products (DPs) need to show similar HOS to the reference product. The solution NMR spectroscopy is a non-invasive, chemically and structurally specific analytical method that is ideal for characterizing protein therapeutics in formulation. However, only limited NMR studies have been performed directly on marketed DPs and questions remain on how to quantitively define similarity. Here, NMR spectra were collected on marketed peptide and protein DPs, including calcitonin-salmon, liraglutide, teriparatide, exenatide, insulin glargine and rituximab. The 1D 1H spectral pattern readily revealed protein HOS heterogeneity, exchange and oligomerization in the different formulations. Principal component analysis (PCA) applied to two rituximab DPs showed consistent results with the previously demonstrated similarity metrics of Mahalanobis distance (DM) of 3.3. The 2D 1H-13C HSQC spectral comparison of insulin glargine DPs provided similarity metrics for chemical shift difference (Δδ) and methyl peak profile, i.e., 4 ppb for 1H, 15 ppb for 13C and 98% peaks with equivalent peak height. Finally, 2D 1H-15N sofast HMQC was demonstrated as a sensitive method for comparison of small protein HOS. The application of NMR procedures and chemometric analysis on therapeutic proteins offer quantitative similarity assessments of DPs with practically achievable similarity metrics.
Collapse
|
15
|
Bramham JE, Davies SA, Podmore A, Golovanov AP. Stability of a high-concentration monoclonal antibody solution produced by liquid-liquid phase separation. MAbs 2021; 13:1940666. [PMID: 34225583 PMCID: PMC8265796 DOI: 10.1080/19420862.2021.1940666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Subcutaneous injection of a low volume (<2 mL) high concentration (>100 mg/mL) formulation is an attractive administration strategy for monoclonal antibodies (mAbs) and other biopharmaceutical proteins. Using concentrated solutions may also be beneficial at various stages of bioprocessing. However, concentrating proteins by conventional techniques, such as ultrafiltration, can be time consuming and challenging. Isolation of the dense fraction produced by macroscopic liquid–liquid phase separation (LLPS) has been suggested as a means to produce high-concentration solutions, but practicality of this method, and the stability of the resulting protein solution have not previously been demonstrated. In this proof-of-concept study, we demonstrate that LLPS can be used to concentrate a mAb solution to >170 mg/mL. We show that the structure of the mAb is not altered by LLPS, and unperturbed mAb is recoverable following dilution of the dense fraction, as judged by 1H nuclear magnetic resonance spectroscopy. Finally, we show that the physical properties and stability of a model high concentration protein formulation obtained from the dense fraction can be improved, for example through the addition of the excipient arginine·glutamate. This results in a stable high-concentration protein formulation with reduced viscosity and no further macroscopic LLPS. Concentrating mAb solutions by LLPS represents a simple and effective technique to progress toward producing high-concentration protein formulations for bioprocessing or administration. Abbreviations Arginine·glutamate (Arg·Glu), Carr-Purcell-Meiboom-Gill (CPMG), critical temperature (TC), high-performance size-exclusion chromatography (HPSEC), liquid–liquid phase separation (LLPS), monoclonal antibody (mAb), nuclear magnetic resonance (NMR), transverse relaxation rate (R2)
Collapse
Affiliation(s)
- Jack E Bramham
- Manchester Institute of Biotechnology and Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Stephanie A Davies
- Dosage Form Design & Development, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Adrian Podmore
- Dosage Form Design & Development, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Alexander P Golovanov
- Manchester Institute of Biotechnology and Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| |
Collapse
|
16
|
Phyo P, Zhao X, Templeton AC, Xu W, Cheung JK, Su Y. Understanding molecular mechanisms of biologics drug delivery and stability from NMR spectroscopy. Adv Drug Deliv Rev 2021; 174:1-29. [PMID: 33609600 DOI: 10.1016/j.addr.2021.02.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/20/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023]
Abstract
Protein therapeutics carry inherent limitations of membrane impermeability and structural instability, despite their predominant role in the modern pharmaceutical market. Effective formulations are needed to overcome physiological and physicochemical barriers, respectively, for improving bioavailability and stability. Knowledge of membrane affinity, cellular internalization, encapsulation, and release of drug-loaded carrier vehicles uncover the structural basis for designing and optimizing biopharmaceuticals with enhanced delivery efficiency and therapeutic efficacy. Understanding stabilizing and destabilizing interactions between protein drugs and formulation excipients provide fundamental mechanisms for ensuring the stability and quality of biological products. This article reviews the molecular studies of biologics using solution and solid-state NMR spectroscopy on structural attributes pivotal to drug delivery and stability. In-depth investigation of the structure-function relationship of drug delivery systems based on cell-penetrating peptides, lipid nanoparticles and polymeric colloidal, and biophysical and biochemical stability of peptide, protein, monoclonal antibody, and vaccine, as the integrative efforts on drug product design, will be elaborated.
Collapse
Affiliation(s)
- Pyae Phyo
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Xi Zhao
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Allen C Templeton
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Wei Xu
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Jason K Cheung
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Yongchao Su
- Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ 07033, United States.
| |
Collapse
|