1
|
Willis LF, Trayton I, Saunders JC, Brùque MG, Davis Birch W, Westhead DR, Day K, Bond NJ, Devine PWA, Lloyd C, Kapur N, Radford SE, Darton NJ, Brockwell DJ. Rationalizing mAb Candidate Screening Using a Single Holistic Developability Parameter. Mol Pharm 2025; 22:181-195. [PMID: 39681988 DOI: 10.1021/acs.molpharmaceut.4c00829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
A framework for the rational selection of a minimal suite of nondegenerate developability assays (DAs) that maximize insight into candidate developability or storage stability is lacking. To address this, we subjected nine formulation:mAbs to 12 mechanistically distinct DAs together with measurement of their accelerated and long-term storage stability. We show that it is possible to identify a reduced set of key variables from this suite of DAs by using orthogonal statistical methods. We exemplify our approach by predicting the rank formulation:mAb degradation rate at 25 °C (determined over 6 months) using just five DAs that can be measured in less than 1 day, spanning a range of physicochemical features. Implementing such approaches focuses on resources, thus increasing sustainability and decreasing development costs.
Collapse
Affiliation(s)
- Leon F Willis
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, U.K
| | | | | | - Maria G Brùque
- The Discovery Centre─AstraZeneca PLC, Cambridge CB2 0AA, U.K
| | - William Davis Birch
- School of Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - David R Westhead
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Katie Day
- The Discovery Centre─AstraZeneca PLC, Cambridge CB2 0AA, U.K
| | - Nicholas J Bond
- The Discovery Centre─AstraZeneca PLC, Cambridge CB2 0AA, U.K
| | - Paul W A Devine
- The Discovery Centre─AstraZeneca PLC, Cambridge CB2 0AA, U.K
| | | | - Nikil Kapur
- School of Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds LS2 9JT, U.K
| | - Sheena E Radford
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, U.K
| | | | - David J Brockwell
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, U.K
| |
Collapse
|
2
|
Willis LF, Kapur N, Radford SE, Brockwell DJ. Biophysical Analysis of Therapeutic Antibodies in the Early Development Pipeline. Biologics 2024; 18:413-432. [PMID: 39723199 PMCID: PMC11669289 DOI: 10.2147/btt.s486345] [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] [Received: 10/12/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
The successful progression of therapeutic antibodies and other biologics from the laboratory to the clinic depends on their possession of "drug-like" biophysical properties. The techniques and the resultant biophysical and biochemical parameters used to characterize their ease of manufacture can be broadly defined as developability. Focusing on antibodies, this review firstly discusses established and emerging biophysical techniques used to probe the early-stage developability of biologics, aimed towards those new to the field. Secondly, we describe the inter-relationships and redundancies amongst developability assays and how in silico methods aid the efficient deployment of developability to bring a new generation of cost-effective therapeutic proteins from bench to bedside more quickly and sustainably.
Collapse
Affiliation(s)
- Leon F Willis
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Nikil Kapur
- School of Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Sheena E Radford
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - David J Brockwell
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
3
|
Zürcher D, Wuchner K, Arosio P. Mitigation Strategies against Antibody Aggregation Induced by Oleic Acid in Liquid Formulations. Mol Pharm 2024; 21:5761-5771. [PMID: 39444106 PMCID: PMC11539069 DOI: 10.1021/acs.molpharmaceut.4c00754] [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: 07/10/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Polysorbates 20 and 80 (PS20 and PS80) are commonly used in the formulations of biologics to protect against interfacial stresses. However, these surfactants can degrade over time, releasing free fatty acids, which assemble into solid particles or liquid droplets. Here, we apply a droplet microfluidic platform to analyze the interactions between antibodies and oleic acid, the primary free fatty acid resulting from the hydrolysis of PS80. We show that antibodies adsorb within seconds to the polar oleic acid-water interface, forming a viscoelastic protein layer that leads to particle formation upon mechanical rupture. By testing two different monoclonal antibodies of pharmaceutical origin, we show that the propensity to form a rigid viscoelastic layer is protein-specific. We further demonstrate that intact PS80 is effective in preventing antibody adsorption at the oleic acid-water interface only at low antibody concentrations and low pH, where oleic acid is fully protonated. Importantly, introduction of the amino acid l-arginine prevents the formation of the interfacial layer and protein particles even at high antibody concentrations (180 mg mL-1). Overall, our findings indicate that oleic acid droplets in antibody formulations can lead to the formation of protein particles via an interface-mediated mechanism. Depending on the conditions, intact PS80 alone might not be sufficient to protect against antibody aggregation. Additional mitigation strategies include the optimization of protein physicochemical properties, pH, and the addition of arginine.
Collapse
Affiliation(s)
- Dominik Zürcher
- Department
of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zürich, 8093 Zürich, Switzerland
| | - Klaus Wuchner
- Cilag
GmbH International, a Division of Johnson & Johnson TDS-Biologics,
Analytical Development, 8200 Schaffhausen, Switzerland
| | - Paolo Arosio
- Department
of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
4
|
Li W, Lin H, Huang Z, Xie S, Zhou Y, Gong R, Jiang Q, Xiang C, Huang J. DOTAD: A Database of Therapeutic Antibody Developability. Interdiscip Sci 2024; 16:623-634. [PMID: 38530613 DOI: 10.1007/s12539-024-00613-2] [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: 08/13/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 03/28/2024]
Abstract
The development of therapeutic antibodies is an important aspect of new drug discovery pipelines. The assessment of an antibody's developability-its suitability for large-scale production and therapeutic use-is a particularly important step in this process. Given that experimental assays to assess antibody developability in large scale are expensive and time-consuming, computational methods have been a more efficient alternative. However, the antibody research community faces significant challenges due to the scarcity of readily accessible data on antibody developability, which is essential for training and validating computational models. To address this gap, DOTAD (Database Of Therapeutic Antibody Developability) has been built as the first database dedicated exclusively to the curation of therapeutic antibody developability information. DOTAD aggregates all available therapeutic antibody sequence data along with various developability metrics from the scientific literature, offering researchers a robust platform for data storage, retrieval, exploration, and downloading. In addition to serving as a comprehensive repository, DOTAD enhances its utility by integrating a web-based interface that features state-of-the-art tools for the assessment of antibody developability. This ensures that users not only have access to critical data but also have the convenience of analyzing and interpreting this information. The DOTAD database represents a valuable resource for the scientific community, facilitating the advancement of therapeutic antibody research. It is freely accessible at http://i.uestc.edu.cn/DOTAD/ , providing an open data platform that supports the continuous growth and evolution of computational methods in the field of antibody development.
Collapse
Affiliation(s)
- Wenzhen Li
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Hongyan Lin
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Ziru Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Shiyang Xie
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yuwei Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Rong Gong
- School of Computer Science and Technology, Aba Teachers University, Aba, 623002, China
| | - Qianhu Jiang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - ChangCheng Xiang
- School of Computer Science and Technology, Aba Teachers University, Aba, 623002, China.
| | - Jian Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China.
| |
Collapse
|
5
|
Jungbauer A, Ferreira G, Butler M, D'Costa S, Brower K, Rayat A, Willson R. Status and future developments for downstream processing of biological products: Perspectives from the Recovery XIX yield roundtable discussions. Biotechnol Bioeng 2024; 121:2524-2541. [PMID: 38795025 DOI: 10.1002/bit.28738] [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: 12/30/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/27/2024]
Abstract
Governments and biopharmaceutical organizations aggressively leveraged expeditious communication capabilities, decision models, and global strategies to make a COVID-19 vaccine happen within a period of 12 months. This was an unusual effort and cannot be transferred to normal times. However, this focus on a single vaccine has also led to other treatments and drug developments being sidelined. Society expects the pharmaceutical industry to provide an uninterrupted supply of medicines. However, it is often overlooked how complex the manufacture of these compounds is and what logistics are required, not to mention the time needed to develop new drugs. The overarching theme, therefore, is patient access and how we can help ensure access and extend it to low- and middle-income countries. Despite unceasing efforts to make medications available to all patient populations, this must never be done at the expense of patient safety. A major fraction of the costs in biopharmaceutical manufacturing are for drug discovery, process development, and clinical studies. Infrastructure costs are very difficult to quantify because they often depend on whether a greenfield facility or an existing, depreciated facility is used or adapted for a new product. To accelerate process development concepts of platform process and prior knowledge are increasingly leveraged. While more traditional protein therapeutics continue to dominate the field, we are also experiencing the exciting emergence and evolution of other therapeutic formats (bispecifics, tetravalent mAbs, antibody-drug conjugates, enzymes, peptides, etc.) that offer unique treatment options for patients. Protein modalities are still dominant, but new modalities are being developed that can be learned from including advanced therapeutics-like cell and gene therapies. The industry must develop a model-based strategy for process development and technologies such as continuous integrated biomanufacturing must be adopted. The overall conclusion is that the pandemic pace was unsustainable, focused on vaccine delivery at the expense of other modalities/disease targets, and had implications for professional and personal life (work-life balance). Routinely reducing development time from 10 years to 1 year is nearly impossible to achieve. Environmental aspects of sustainable downstream processing are also described.
Collapse
Affiliation(s)
- Alois Jungbauer
- Department of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Gisela Ferreira
- Process Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Michelle Butler
- Pharmaceutical Technical Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Susan D'Costa
- Technology Development and Manufacturing, Genezen Laboratories, Indianapolis, Indiana, USA
| | - Kevin Brower
- Mammalian Platform, Sanofi, Framingham, Massachusetts, USA
| | - Andrea Rayat
- Department of Biochemical Engineering, University College London, London, UK
| | - Richard Willson
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas, USA
| |
Collapse
|
6
|
Condado-Morales I, Dingfelder F, Waibel I, Turnbull OM, Patel B, Cao Z, Rose Bjelke J, Nedergaard Grell S, Bennet A, Hummer AM, Raybould MIJ, Deane CM, Egebjerg T, Lorenzen N, Arosio P. A comparative study of the developability of full-length antibodies, fragments, and bispecific formats reveals higher stability risks for engineered constructs. MAbs 2024; 16:2403156. [PMID: 39364796 PMCID: PMC11457596 DOI: 10.1080/19420862.2024.2403156] [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: 04/22/2024] [Revised: 08/16/2024] [Accepted: 09/07/2024] [Indexed: 10/05/2024] Open
Abstract
Engineered antibody formats, such as antibody fragments and bispecifics, have the potential to offer improved therapeutic efficacy compared to traditional full-length monoclonal antibodies (mAbs). However, the translation of these non-natural molecules into successful therapeutics can be hampered by developability challenges. Here, we systematically analyzed 64 different antibody constructs targeting Tumor Necrosis Factor (TNF) which cover 8 distinct molecular format families, encompassing full-length antibodies, various types of single chain variable fragments, and bispecifics. We measured 15 biophysical properties related to activity, manufacturing, and stability, scoring variants with a flag-based risk approach and a recent in silico developability profiler. Our comparative assessment revealed that overall developability is higher for the natural full-length antibody format. Bispecific antibodies, antibodies with scFv fragments at the C-terminus of the light chain, and single-chain Fv antibody fragments (scFvs) have intermediate developability properties, while more complicated formats, such as scFv- scFv, bispecific mAbs with one Fab exchanged with a scFv, and diabody formats are collectively more challenging. In particular, our study highlights the propensity for fragmentation and aggregation, both in bulk and at interfaces, for many current engineered formats.
Collapse
Affiliation(s)
- Itzel Condado-Morales
- Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Fabian Dingfelder
- Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Isabel Waibel
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | - Bhargav Patel
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Zheng Cao
- Department of Bioanalysis, Beijing Novo Nordisk Pharmaceutical Science & Technology Co. Ltd (Novo Nordisk R&D China), Beijing, China
| | - Jais Rose Bjelke
- Department of Purification Technologies, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | | | - Anja Bennet
- Department of Kidney Biology, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | | | | | | | - Thomas Egebjerg
- Department of Mammalian Expression, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | - Nikolai Lorenzen
- Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv, Denmark
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, Zurich, Switzerland
| |
Collapse
|
7
|
Kulakova A, Augustijn D, El Bialy I, Gentiluomo L, Greco ML, Hervø-Hansen S, Indrakumar S, Mahapatra S, Martinez Morales M, Pohl C, Polimeni M, Roche A, Svilenov HL, Tosstorff A, Zalar M, Curtis R, Derrick JP, Frieß W, Golovanov AP, Lund M, Nørgaard A, Khan TA, Peters GHJ, Pluen A, Roessner D, Streicher WW, van der Walle CF, Warwicker J, Uddin S, Winter G, Bukrinski JT, Rinnan Å, Harris P. Chemometrics in Protein Formulation: Stability Governed by Repulsion and Protein Unfolding. Mol Pharm 2023. [PMID: 37146162 DOI: 10.1021/acs.molpharmaceut.3c00013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Therapeutic proteins can be challenging to develop due to their complexity and the requirement of an acceptable formulation to ensure patient safety and efficacy. To date, there is no universal formulation development strategy that can identify optimal formulation conditions for all types of proteins in a fast and reliable manner. In this work, high-throughput characterization, employing a toolbox of five techniques, was performed on 14 structurally different proteins formulated in 6 different buffer conditions and in the presence of 4 different excipients. Multivariate data analysis and chemometrics were used to analyze the data in an unbiased way. First, observed changes in stability were primarily determined by the individual protein. Second, pH and ionic strength are the two most important factors determining the physical stability of proteins, where there exists a significant statistical interaction between protein and pH/ionic strength. Additionally, we developed prediction methods by partial least-squares regression. Colloidal stability indicators are important for prediction of real-time stability, while conformational stability indicators are important for prediction of stability under accelerated stress conditions at 40 °C. In order to predict real-time storage stability, protein-protein repulsion and the initial monomer fraction are the most important properties to monitor.
Collapse
Affiliation(s)
- Alina Kulakova
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| | - Dillen Augustijn
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, Frederiksberg 1958, Denmark
| | - Inas El Bialy
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Lorenzo Gentiluomo
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
- Wyatt Technology Europe GmbH, Hochstrasse 18, Dernbach 56307, Germany
| | - Maria Laura Greco
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Stefan Hervø-Hansen
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund 22100, Sweden
| | - Sowmya Indrakumar
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| | | | - Marcello Martinez Morales
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Christin Pohl
- Novozymes A/S, Krogshoejvej 36, Bagsvaerd 2880, Denmark
| | - Marco Polimeni
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund 22100, Sweden
| | - Aisling Roche
- Department of Chemical Engineering, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Hristo L Svilenov
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Andreas Tosstorff
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Matja Zalar
- Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, and Manchester Institute of Biotechnology, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| | - Robin Curtis
- Department of Chemical Engineering, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Jeremy P Derrick
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, U.K
| | - Wolfgang Frieß
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | - Alexander P Golovanov
- Department of Chemistry, School of Natural Sciences, Faculty of Science and Engineering, and Manchester Institute of Biotechnology, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K
| | - Mikael Lund
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, P.O. Box 124, Lund 22100, Sweden
| | | | - Tarik A Khan
- Pharmaceutical Development & Supplies, Pharma Technical Development Biologics Europe, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| | - Alain Pluen
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, U.K
| | - Dierk Roessner
- Wyatt Technology Europe GmbH, Hochstrasse 18, Dernbach 56307, Germany
| | | | - Christopher F van der Walle
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Jim Warwicker
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester M13 9PT, U.K
| | - Shahid Uddin
- Dosage Form Design and Development, AstraZeneca, Sir Aaron Klug Building, Granta Park, Cambridge CB21 6GH, U.K
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, Munich 81377, Germany
| | | | - Åsmund Rinnan
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, Frederiksberg 1958, Denmark
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, Kongens, Lyngby 2800, Denmark
| |
Collapse
|
8
|
Svilenov HL, Arosio P, Menzen T, Tessier P, Sormanni P. Approaches to expand the conventional toolbox for discovery and selection of antibodies with drug-like physicochemical properties. MAbs 2023; 15:2164459. [PMID: 36629855 PMCID: PMC9839375 DOI: 10.1080/19420862.2022.2164459] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/12/2023] Open
Abstract
Antibody drugs should exhibit not only high-binding affinity for their target antigens but also favorable physicochemical drug-like properties. Such drug-like biophysical properties are essential for the successful development of antibody drug products. The traditional approaches used in antibody drug development require significant experimentation to produce, optimize, and characterize many candidates. Therefore, it is attractive to integrate new methods that can optimize the process of selecting antibodies with both desired target-binding and drug-like biophysical properties. Here, we summarize a selection of techniques that can complement the conventional toolbox used to de-risk antibody drug development. These techniques can be integrated at different stages of the antibody development process to reduce the frequency of physicochemical liabilities in antibody libraries during initial discovery and to co-optimize multiple antibody features during early-stage antibody engineering and affinity maturation. Moreover, we highlight biophysical and computational approaches that can be used to predict physical degradation pathways relevant for long-term storage and in-use stability to reduce the need for extensive experimentation.
Collapse
Affiliation(s)
- Hristo L. Svilenov
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Gent, Belgium
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Tim Menzen
- Coriolis Pharma Research GmbH, Martinsried, 82152, Germany
| | - Peter Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Pietro Sormanni
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
9
|
Indrakumar S, Kulakova A, Harris P, Peters GHJ. Dynamics of Human Serum Transferrin in Varying Physicochemical Conditions Explored by Using Molecular Dynamics Simulations. Mol Pharm 2022; 19:2795-2806. [PMID: 35776490 DOI: 10.1021/acs.molpharmaceut.2c00158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Conformational stability of human serum transferrin (Tf) at varying pH values and salt and excipient concentrations were investigated using molecular dynamics (MD) simulations, and the results are compared with previously published small-angle X-ray scattering (SAXS) experiments. SAXS study showed that at pH 5, Tf is predominantly present in a partially open (PO) form, and the factions of PO differ based on the physicochemical condition and drift toward the closed form (HO) as the pH increases. Tf is a bilobal glycoprotein that is composed of homologous halves termed the N- and C-lobes. The current study shows that the protonation of Y188 and K206 at pH 5 is the primary conformational drive into PO, which shifts toward the closed (HO) conformer as the pH increases. Furthermore, at pH 6.5, PO is unfavorable due to negative charge-charge repulsion at the N/C-lobe interface linker region causing increased hinge distance when compared to HO, which has favorable attractive electrostatic interactions in this region. Subsequently, the effect of salt concentration was studied at 70 and 140 mM NaCl. At 70 mM NaCl and pH 5, chloride ions bind strongly in the N-lobe iron-binding site, whereas these interactions are weak at pH 6.5. With increasing salt concentration at pH 5, the regions surrounding the N-lobe iron-binding site are saturated, and as a consequence, sodium and chloride ions accumulate into the bulk. Additionally, protein-excipient interactions were investigated. At pH 5, the excipients interact in similar loop regions, E89-T93, and D416-D420, located in the N- and C-lobes of the HO conformer, respectively. It is anticipated that interactions of additives in these two loop regions cause conformational changes that lead to iron-coordinating residues in the N-lobe to drift away from iron and thus drive HO to PO conversion. Furthermore, at pH 6.5 and 140 mM histidine, these interactions are negligible leading to the stabilization of HO.
Collapse
Affiliation(s)
- Sowmya Indrakumar
- Technical University of Denmark, Department of Chemistry, 2800 Kgs. Lyngby, Denmark
| | - Alina Kulakova
- Technical University of Denmark, Department of Chemistry, 2800 Kgs. Lyngby, Denmark
| | - Pernille Harris
- Technical University of Denmark, Department of Chemistry, 2800 Kgs. Lyngby, Denmark
| | - Günther H J Peters
- Technical University of Denmark, Department of Chemistry, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
10
|
Assessment of Therapeutic Antibody Developability by Combinations of In Vitro and In Silico Methods. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2313:57-113. [PMID: 34478132 DOI: 10.1007/978-1-0716-1450-1_4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although antibodies have become the fastest-growing class of therapeutics on the market, it is still challenging to develop them for therapeutic applications, which often require these molecules to withstand stresses that are not present in vivo. We define developability as the likelihood of an antibody candidate with suitable functionality to be developed into a manufacturable, stable, safe, and effective drug that can be formulated to high concentrations while retaining a long shelf life. The implementation of reliable developability assessments from the early stages of antibody discovery enables flagging and deselection of potentially problematic candidates, while focussing available resources on the development of the most promising ones. Currently, however, thorough developability assessment requires multiple in vitro assays, which makes it labor intensive and time consuming to implement at early stages. Furthermore, accurate in vitro analysis at the early stage is compromised by the high number of potential candidates that are often prepared at low quantities and purity. Recent improvements in the performance of computational predictors of developability potential are beginning to change this scenario. Many computational methods only require the knowledge of the amino acid sequences and can be used to identify possible developability issues or to rank available candidates according to a range of biophysical properties. Here, we describe how the implementation of in silico tools into antibody discovery pipelines is increasingly offering time- and cost-effective alternatives to in vitro experimental screening, thus streamlining the drug development process. We discuss in particular the biophysical and biochemical properties that underpin developability potential and their trade-offs, review various in vitro assays to measure such properties or parameters that are predictive of developability, and give an overview of the growing number of in silico tools available to predict properties important for antibody development, including the CamSol method developed in our laboratory.
Collapse
|
11
|
Heads JT, Kelm S, Tyson K, Lawson ADG. A computational method for predicting the aggregation propensity of IgG1 and IgG4(P) mAbs in common storage buffers. MAbs 2022; 14:2138092. [PMID: 36418193 PMCID: PMC9704409 DOI: 10.1080/19420862.2022.2138092] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The propensity for some monoclonal antibodies (mAbs) to aggregate at physiological and manufacturing pH values can prevent their use as therapeutic molecules or delay time to market. Consequently, developability assessments are essential to select optimum candidates, or inform on mitigation strategies to avoid potential late-stage failures. These studies are typically performed in a range of buffer solutions because factors such as pH can dramatically alter the aggregation propensity of the test mAbs (up to 100-fold in extreme cases). A computational method capable of robustly predicting the aggregation propensity at the pH values of common storage buffers would have substantial value. Here, we describe a mAb aggregation prediction tool (MAPT) that builds on our previously published isotype-dependent, charge-based model of aggregation. We show that the addition of a homology model-derived hydrophobicity descriptor to our electrostatic aggregation model enabled the generation of a robust mAb developability indicator. To contextualize our aggregation scoring system, we analyzed 97 clinical-stage therapeutic mAbs. To further validate our approach, we focused on six mAbs (infliximab, tocilizumab, rituximab, CNTO607, MEDI1912 and MEDI1912_STT) which have been reported to cover a large range of aggregation propensities. The different aggregation propensities of the case study molecules at neutral and slightly acidic pH were correctly predicted, verifying the utility of our computational method.
Collapse
Affiliation(s)
- James T. Heads
- UCB Pharma, 208 Bath Road, SloughSL1 3WE, UK,CONTACT James T. Heads UCB Pharma, 208 Bath Road, SloughSL1 3WE, UK
| | | | - Kerry Tyson
- UCB Pharma, 208 Bath Road, SloughSL1 3WE, UK
| | | |
Collapse
|
12
|
Mahapatra S, Polimeni M, Gentiluomo L, Roessner D, Frieß W, Peters GHJ, Streicher WW, Lund M, Harris P. Self-Interactions of Two Monoclonal Antibodies: Small-Angle X-ray Scattering, Light Scattering, and Coarse-Grained Modeling. Mol Pharm 2021; 19:508-519. [PMID: 34939811 DOI: 10.1021/acs.molpharmaceut.1c00627] [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] [Indexed: 11/29/2022]
Abstract
Using light scattering (LS), small-angle X-ray scattering (SAXS), and coarse-grained Monte Carlo (MC) simulations, we studied the self-interactions of two monoclonal antibodies (mAbs), PPI03 and PPI13. With LS measurements, we obtained the osmotic second virial coefficient, B22, and the molecular weight, Mw, of the two mAbs, while with SAXS measurements, we studied the mAbs' self-interaction behavior in the high protein concentration regime up to 125 g/L. Through SAXS-derived coarse-grained representations of the mAbs, we performed MC simulations with either a one-protein or a two-protein model to predict B22. By comparing simulation and experimental results, we validated our models and obtained insights into the mAbs' self-interaction properties, highlighting the role of both ion binding and charged patches on the mAb surfaces. Our models provide useful information about mAbs' self-interaction properties and can assist the screening of conditions driving to colloidal stability.
Collapse
Affiliation(s)
- Sujata Mahapatra
- Novozymes A/S, Biologiens Vej 2, 2800 Kgs. Lyngby, Denmark.,Department of Chemistry, Technical University of Denmark, Kemitorvet Building 207, 2800 Kgs. Lyngby, Denmark
| | - Marco Polimeni
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, Naturvetarvägen 14, 223 62 Lund, Sweden
| | - Lorenzo Gentiluomo
- Wyatt Technology Europe GmbH, Hochstrasse 12a, 56307 Dernbach, Germany.,Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig Maximilians-Universität München, Butenandtstrasse 5, 81377 Munich, Germany
| | - Dierk Roessner
- Wyatt Technology Europe GmbH, Hochstrasse 12a, 56307 Dernbach, Germany
| | - Wolfgang Frieß
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig Maximilians-Universität München, Butenandtstrasse 5, 81377 Munich, Germany
| | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, Kemitorvet Building 207, 2800 Kgs. Lyngby, Denmark
| | | | - Mikael Lund
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, Naturvetarvägen 14, 223 62 Lund, Sweden.,Advanced X-ray and Neutron Science (LINXS), Lund University, Scheelevägen 19, 22370 Lund, Sweden
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark, Kemitorvet Building 207, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
13
|
Pohl C, Polimeni M, Indrakumar S, Streicher W, Peters GHJ, Nørgaard A, Lund M, Harris P. Electrostatics Drive Oligomerization and Aggregation of Human Interferon Alpha-2a. J Phys Chem B 2021; 125:13657-13669. [PMID: 34898211 PMCID: PMC8713289 DOI: 10.1021/acs.jpcb.1c07090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Aggregation is a
common phenomenon in the field of protein therapeutics
and can lead to function loss or immunogenic patient responses. Two
strategies are currently used to reduce aggregation: (1) finding a
suitable formulation, which is labor-intensive and requires large
protein quantities, or (2) engineering the protein, which requires
extensive knowledge about the protein aggregation pathway. We present
a biophysical characterization of the oligomerization and aggregation
processes by Interferon alpha-2a (IFNα-2a), a protein drug with
antiviral and immunomodulatory properties. This study combines experimental
high throughput screening with detailed investigations by small-angle
X-ray scattering and analytical ultracentrifugation. Metropolis Monte
Carlo simulations are used to gain insight into the underlying intermolecular
interactions. IFNα-2a forms soluble oligomers that are controlled
by a fast pH and concentration-dependent equilibrium. Close to the
isoelectric point of 6, IFNα-2a forms insoluble aggregates which
can be prevented by adding salt. We show that monomer attraction is driven mainly by molecular anisotropic dipole–dipole interactions
that increase with increasing pH. Repulsion is due
to monopole–monopole interactions and depends on the charge
of IFNα-2a. The study highlights how combining multiple methods
helps to systematically dissect the molecular mechanisms driving oligomer
formation and to design ultimately efficient strategies for preventing
detrimental protein aggregation.
Collapse
Affiliation(s)
- Christin Pohl
- Novozymes A/S, Bagsvaerd, 2880, Denmark.,Department of Chemistry, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Marco Polimeni
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, 221 00, Lund, Sweden
| | - Sowmya Indrakumar
- Department of Chemistry, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | | | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | | | - Mikael Lund
- Division of Theoretical Chemistry, Department of Chemistry, Lund University, 221 00, Lund, Sweden
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| |
Collapse
|
14
|
Pohl C, Mahapatra S, Kulakova A, Streicher W, Peters GHJ, Nørgaard A, Harris P. Combination of high throughput and structural screening to assess protein stability - a screening perspective. Eur J Pharm Biopharm 2021; 171:1-10. [PMID: 34826593 DOI: 10.1016/j.ejpb.2021.08.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 11/04/2022]
Abstract
High throughput screening to measure the stability of industrially relevant proteins and their variants is necessary for quality assessment in the development process. Advances in automation, measurement time and sample consumption for many techniques allow rapid measurements with minimal amount of protein. However, many methods include automated data analysis, potentially neglecting important aspects of the proteińs behavior in certain conditions. In this study we implement small angle X-ray scattering (SAXS), typically not used to assess protein behavior in industrial screening, in a high throughput screening workflow to address problems of contradicting results and reproducibility among different high throughput methods. As a case study we use the lipases of Thermomyces lanuginosus and Rhizomucor miehei, widely used industrial biocatalysts. We show that even the initial analysis of the SAXS data without performing any time-consuming modelling provide valuable information on interparticle interactions. We conclude that recent advances in automation and data processing, have enabled SAXS to be used more widely as a tool to gain in-depth knowledge highly useful for protein formulation development. This is especially relevant in light of increasing accessibility to SAXS due to the commercial availability of benchtop instruments.
Collapse
Affiliation(s)
- Christin Pohl
- Novozymes A/S, Krogshoejvej 36, 2880, Bagsvaerd, Denmark; Technical University of Denmark, Department of Chemistry, Kemitorvet 207, 2800 Kongens Lyngby, Denmark.
| | - Sujata Mahapatra
- Novozymes A/S, Krogshoejvej 36, 2880, Bagsvaerd, Denmark; Technical University of Denmark, Department of Chemistry, Kemitorvet 207, 2800 Kongens Lyngby, Denmark
| | - Alina Kulakova
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen
| | | | - Günther H J Peters
- Technical University of Denmark, Department of Chemistry, Kemitorvet 207, 2800 Kongens Lyngby, Denmark
| | - Allan Nørgaard
- Novozymes A/S, Krogshoejvej 36, 2880, Bagsvaerd, Denmark
| | - Pernille Harris
- Technical University of Denmark, Department of Chemistry, Kemitorvet 207, 2800 Kongens Lyngby, Denmark.
| |
Collapse
|
15
|
Austerberry J, Edwards J, Eyes T, Derrick JP. Use of Peptide Microarrays for Fast and Informative Profiling of Therapeutic Antibody Formulation Conditions. Mol Pharm 2021; 18:4131-4139. [PMID: 34658237 DOI: 10.1021/acs.molpharmaceut.1c00543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Methods to optimize the solution behavior of therapeutic proteins are frequently time-consuming, provide limited information, and often use milligram quantities of material. Here, we present a simple, versatile method that provides valuable information to guide the identification and comparison of formulation conditions for, in principle, any biopharmaceutical drug. The subject protein is incubated with a designed synthetic peptide microarray; the extent of binding to each peptide is dependent on the solution conditions. The array is washed, and the adhesion of the subject protein is detected using a secondary antibody. We exemplify the method using a well-characterized human single-chain Fv and a selection of human monoclonal antibodies. Correlations of peptide adhesion profiles can be used to establish quantitative relationships between different solution conditions, allowing subgrouping into dendrograms. Multidimensional reduction methods, such as t-distributed stochastic neighbor embedding, can be applied to compare how different monoclonals vary in their adhesion properties under different solution conditions. Finally, we screened peptide binding profiles using a selection of monoclonal antibodies for which a range of biophysical measurements were available under specified buffer conditions. We used a neural network method to train the data against aggregation temperature, kD, percentage recovery after incubation at 25 °C, and melting temperature. The results demonstrate that peptide binding profiles can indeed be effectively trained on these indicators of protein stability and self-association in solution. The method opens up multiple possibilities for the application of machine learning methods in therapeutic protein formulation.
Collapse
Affiliation(s)
- James Austerberry
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - John Edwards
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Tim Eyes
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Jeremy P Derrick
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
16
|
Gomes DC, Teixeira SCM, Leão JB, Razinkov VI, Qi W, Rodrigues MA, Roberts CJ. In Situ Monitoring of Protein Unfolding/Structural States under Cold High-Pressure Stress. Mol Pharm 2021; 18:4415-4427. [PMID: 34699230 DOI: 10.1021/acs.molpharmaceut.1c00604] [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] [Indexed: 12/24/2022]
Abstract
Biopharmaceutical formulations may be compromised by freezing, which has been attributed to protein conformational changes at a low temperature, and adsorption to ice-liquid interfaces. However, direct measurements of unfolding/conformational changes in sub-0 °C environments are limited because at ambient pressure, freezing of water can occur, which limits the applicability of otherwise commonly used analytical techniques without specifically tailored instrumentation. In this report, small-angle neutron scattering (SANS) and intrinsic fluorescence (FL) were used to provide in situ analysis of protein tertiary structure/folding at temperatures as low as -15 °C utilizing a high-pressure (HP) environment (up to 3 kbar) that prevents water from freezing. The results show that the α-chymotrypsinogen A (aCgn) structure is reasonably maintained under acidic pH (and corresponding pD) for all conditions of pressure and temperature tested. On the other hand, reversible structural changes and formation of oligomeric species were detected near -10 °C via HP-SANS for ovalbumin under neutral pD conditions. This was found to be related to the proximity of the temperature of cold denaturation of ovalbumin (TCD ∼ -17 °C; calculated via isothermal chemical denaturation and Gibbs-Helmholtz extrapolation) rather than a pressure effect. Significant structural changes were also observed for a monoclonal antibody, anti-streptavidin IgG1 (AS-IgG1), under acidic conditions near -5 °C and a pressure of ∼2 kbar. The conformational perturbation detected for AS-IgG1 is proposed to be consistent with the formation of unfolding intermediates such as molten globule states. Overall, the in situ approaches described here offer a means to characterize the conformational stability of biopharmaceuticals and proteins more generally under cold-temperature stress by the assessment of structural alteration, self-association, and reversibility of each process. This offers an alternative to current ex situ methods that are based on higher temperatures and subsequent extrapolation of the data and interpretations to the cold-temperature regime.
Collapse
Affiliation(s)
- Diana C Gomes
- Centro de Química Estrutural, Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, Delaware 19713, United States
| | - Susana C M Teixeira
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, Delaware 19713, United States.,NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Juscelino B Leão
- NIST Center for Neutron Research, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, Maryland 20899, United States
| | - Vladimir I Razinkov
- Drug Product Development, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Wei Qi
- Drug Product Development, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Miguel A Rodrigues
- Centro de Química Estrutural, Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Christopher J Roberts
- Department of Chemical and Biomolecular Engineering, University of Delaware, 590 Avenue 1743, Newark, Delaware 19713, United States
| |
Collapse
|
17
|
Long-term stability predictions of therapeutic monoclonal antibodies in solution using Arrhenius-based kinetics. Sci Rep 2021; 11:20534. [PMID: 34654882 PMCID: PMC8519954 DOI: 10.1038/s41598-021-99875-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/01/2021] [Indexed: 11/08/2022] Open
Abstract
Long-term stability of monoclonal antibodies to be used as biologics is a key aspect in their development. Therefore, its possible early prediction from accelerated stability studies is of major interest, despite currently being regarded as not sufficiently robust. In this work, using a combination of accelerated stability studies (up to 6 months) and first order degradation kinetic model, we are able to predict the long-term stability (up to 3 years) of multiple monoclonal antibody formulations. More specifically, we can robustly predict the long-term stability behaviour of a protein at the intended storage condition (5 °C), based on up to six months of data obtained for multiple quality attributes from different temperatures, usually from intended (5 °C), accelerated (25 °C) and stress conditions (40 °C). We have performed stability studies and evaluated the stability data of several mAbs including IgG1, IgG2, and fusion proteins, and validated our model by overlaying the 95% prediction interval and experimental stability data from up to 36 months. We demonstrated improved robustness, speed and accuracy of kinetic long-term stability prediction as compared to classical linear extrapolation used today, which justifies long-term stability prediction and shelf-life extrapolation for some biologics such as monoclonal antibodies. This work aims to contribute towards further development and refinement of the regulatory landscape that could steer toward allowing extrapolation for biologics during the developmental phase, clinical phase, and also in marketing authorisation applications, as already established today for small molecules.
Collapse
|
18
|
Ban D, Rice CT, McCoy MA. Quantification of natural abundance NMR data differentiates the solution behavior of monoclonal antibodies and their fragments. MAbs 2021; 13:1978132. [PMID: 34612804 PMCID: PMC8496538 DOI: 10.1080/19420862.2021.1978132] [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: 11/07/2022] Open
Abstract
Biotherapeutics are an important class of molecules for the treatment of a wide range of diseases. They include low molecular weight peptides, highly engineered protein scaffolds and monoclonal antibodies. During their discovery and development, assessments of the biophysical attributes is critical to understanding the solution behavior of therapeutic proteins and for de-risking liabilities. Thus, methods that can quantify, characterize, and provide a basis to inform risks and drive the selection of more optimal antibody and alternative scaffolds are needed. Nuclear Magnetic Resonance (NMR) spectroscopy is a technique that provides a means to probe antibody and antibody-like molecules in solution, at atomic resolution, under any formulated conditions. Here, all samples were profiled at natural abundance requiring no isotope enrichment. We present a numerical approach that quantitates two-dimensional methyl spectra. The approach was tested with a reference dataset that contained different types of antibody and antibody-like molecules. This dataset was processed through a procedure we call a Random Sampling of NMR Peaks for Covariance Analysis. This analysis revealed that the first two components were well correlated with the hydrodynamic radius of the molecules included in the reference set. Higher-order principal components were also linked to dynamic features between different tethered antibody-like molecules and contributed to decisions around candidate selection. The reference set provides a basis to characterize molecules with unknown solution behavior and is sensitive to the behavior of a molecule formulated under different conditions. The approach is independent of protein design, scaffold, formulation and provides a facile method to quantify solution behavior.
Collapse
Affiliation(s)
- David Ban
- Department of Computational and Structural Chemistry, Merck & Co., Inc, Kenilworth, NJ, USA
| | - Cory T Rice
- Department of Computational and Structural Chemistry, Merck & Co., Inc, Kenilworth, NJ, USA
| | - Mark A McCoy
- Department of Computational and Structural Chemistry, Merck & Co., Inc, Kenilworth, NJ, USA
| |
Collapse
|
19
|
Narayanan H, Dingfelder F, Condado Morales I, Patel B, Heding KE, Bjelke JR, Egebjerg T, Butté A, Sokolov M, Lorenzen N, Arosio P. Design of Biopharmaceutical Formulations Accelerated by Machine Learning. Mol Pharm 2021; 18:3843-3853. [PMID: 34519511 DOI: 10.1021/acs.molpharmaceut.1c00469] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In addition to activity, successful biological drugs must exhibit a series of suitable developability properties, which depend on both protein sequence and buffer composition. In the context of this high-dimensional optimization problem, advanced algorithms from the domain of machine learning are highly beneficial in complementing analytical screening and rational design. Here, we propose a Bayesian optimization algorithm to accelerate the design of biopharmaceutical formulations. We demonstrate the power of this approach by identifying the formulation that optimizes the thermal stability of three tandem single-chain Fv variants within 25 experiments, a number which is less than one-third of the experiments that would be required by a classical DoE method and several orders of magnitude smaller compared to detailed experimental analysis of full combinatorial space. We further show the advantage of this method over conventional approaches to efficiently transfer historical information as prior knowledge for the development of new biologics or when new buffer agents are available. Moreover, we highlight the benefit of our technique in engineering multiple biophysical properties by simultaneously optimizing both thermal and interface stabilities. This optimization minimizes the amount of surfactant in the formulation, which is important to decrease the risks associated with corresponding degradation processes. Overall, this method can provide high speed of converging to optimal conditions, the ability to transfer prior knowledge, and the identification of new nonlinear combinations of excipients. We envision that these features can lead to a considerable acceleration in formulation design and to parallelization of operations during drug development.
Collapse
Affiliation(s)
- Harini Narayanan
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, 8093 Zurich, Switzerland
| | - Fabian Dingfelder
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, 8093 Zurich, Switzerland.,Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv 2760, Denmark
| | - Itzel Condado Morales
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, 8093 Zurich, Switzerland.,Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv 2760, Denmark
| | - Bhargav Patel
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, 8093 Zurich, Switzerland
| | - Kristine Enemærke Heding
- Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv 2760, Denmark
| | - Jais Rose Bjelke
- Department of Purification Technologies, Global Research Technologies, Novo Nordisk A/S, Måløv 2760, Denmark
| | - Thomas Egebjerg
- Department of Mammalian Expression, Global Research Technologies, Novo Nordisk A/S, Måløv 2760, Denmark
| | | | | | - Nikolai Lorenzen
- Department of Biophysics and Injectable Formulation, Global Research Technologies, Novo Nordisk A/S, Måløv 2760, Denmark
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology, 8093 Zurich, Switzerland
| |
Collapse
|
20
|
Kopp MRG, Wolf Pérez AM, Zucca MV, Capasso Palmiero U, Friedrichsen B, Lorenzen N, Arosio P. An accelerated surface-mediated stress assay of antibody instability for developability studies. MAbs 2021; 12:1815995. [PMID: 32954930 PMCID: PMC7577746 DOI: 10.1080/19420862.2020.1815995] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
High physical stability is required for the development of monoclonal antibodies (mAbs) into successful therapeutic products. Developability assays are used to predict physical stability issues such as high viscosity and poor conformational stability, but protein aggregation remains a challenging property to predict. Among different types of stresses, air–water and solid–liquid interfaces are well known to potentially trigger protein instability and induce aggregation. Yet, in contrast to the increasing number of developability assays to evaluate bulk properties, there is still a lack of experimental methods to evaluate antibody stability against interfaces. Here, we investigate the potential of a hydrophobic nanoparticle surface-mediated stress assay to assess the stability of mAbs during the early stages of development. We evaluate this surface-mediated accelerated stability assay on a rationally designed library of 14 variants of a humanized IgG4, featuring a broad span of solubility values and other developability properties. The assay could identify variants characterized by high instability against agitation in the presence of air–water interfaces. Remarkably, for the set of investigated molecules, we observe strong correlations between the extent of aggregation induced by the surface-mediated stress assay and other developability properties of the molecules, such as aggregation upon storage at 45°C, self-association (evaluated by affinity-capture self-interaction nanoparticle spectroscopy) and nonspecific interactions (estimated by cross-interaction chromatography, stand-up monolayer chromatography (SMAC), SMAC*). This highly controlled surface-mediated stress assay has the potential to complement and increase the ability of the current set of screening techniques to assess protein aggregation and developability potential of mAbs during the early stages of drug development. Abbreviations:AC-SINS: Affinity-Capture Self-Interaction Nanoparticle Spectroscopy; AMS: Ammonium sulfate precipitation; ANS: 1-anilinonaphtalene-8-sulfonate; CIC: Cross-interaction chromatography; DLS: Dynamic light scattering; HIC: Hydrophobic interaction chromatography; HNSSA: Hydrophobic nanoparticles surface-stress assay; mAb: Monoclonal antibody; NP: Nanoparticle; SEC: Size exclusion chromatography; SMAC: Stand-up monolayer chromatography; WT: Wild type
Collapse
Affiliation(s)
- Marie R G Kopp
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology , Zurich, Switzerland
| | - Adriana-Michelle Wolf Pérez
- Department of Biophysics, Biophysics and Injectable Formulation, Novo Nordisk , Måløv, Denmark.,Aarhus University, iNANO , Aarhus C, Denmark
| | - Marta Virginia Zucca
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology , Zurich, Switzerland
| | - Umberto Capasso Palmiero
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology , Zurich, Switzerland
| | | | - Nikolai Lorenzen
- Department of Biophysics, Biophysics and Injectable Formulation, Novo Nordisk , Måløv, Denmark
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, Swiss Federal Institute of Technology , Zurich, Switzerland
| |
Collapse
|
21
|
Berner C, Menzen T, Winter G, Svilenov HL. Combining Unfolding Reversibility Studies and Molecular Dynamics Simulations to Select Aggregation-Resistant Antibodies. Mol Pharm 2021; 18:2242-2253. [PMID: 33928776 DOI: 10.1021/acs.molpharmaceut.1c00017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The efficient development of new therapeutic antibodies relies on developability assessment with biophysical and computational methods to find molecules with drug-like properties such as resistance to aggregation. Despite the many novel approaches to select well-behaved proteins, antibody aggregation during storage is still challenging to predict. For this reason, there is a high demand for methods that can identify aggregation-resistant antibodies. Here, we show that three straightforward techniques can select the aggregation-resistant antibodies from a dataset with 13 molecules. The ReFOLD assay provided information about the ability of the antibodies to refold to monomers after unfolding with chemical denaturants. Modulated scanning fluorimetry (MSF) yielded the temperatures that start causing irreversible unfolding of the proteins. Aggregation was the main reason for poor unfolding reversibility in both ReFOLD and MSF experiments. We therefore performed temperature ramps in molecular dynamics (MD) simulations to obtain partially unfolded antibody domains in silico and used CamSol to assess their aggregation potential. We compared the information from ReFOLD, MSF, and MD to size-exclusion chromatography (SEC) data that shows whether the antibodies aggregated during storage at 4, 25, and 40 °C. Contrary to the aggregation-prone molecules, the antibodies that were resistant to aggregation during storage at 40 °C shared three common features: (i) higher tendency to refold to monomers after unfolding with chemical denaturants, (ii) higher onset temperature of nonreversible unfolding, and (iii) unfolding of regions containing aggregation-prone sequences at higher temperatures in MD simulations.
Collapse
Affiliation(s)
- Carolin Berner
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstr. 5, 81377 Munich, Germany
| | - Tim Menzen
- Coriolis Pharma Research GmbH, Fraunhoferstr. 18 b, 82152 Martinsried, Germany
| | - Gerhard Winter
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstr. 5, 81377 Munich, Germany
| | - Hristo L Svilenov
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstr. 5, 81377 Munich, Germany
| |
Collapse
|
22
|
Shahfar H, Forder JK, Roberts CJ. Toward a Suite of Coarse-Grained Models for Molecular Simulation of Monoclonal Antibodies and Therapeutic Proteins. J Phys Chem B 2021; 125:3574-3588. [PMID: 33821645 DOI: 10.1021/acs.jpcb.1c01903] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A series of coarse-grained models for molecular simulation of proteins are considered, with emphasis on the application of predicting protein-protein self-interactions for monoclonal antibodies (MAbs). As an illustrative example and for quantitative comparison, the models are used to predict osmotic virial coefficients over a broad range of attractive and repulsive self-interactions and solution conditions for a series of MAbs where the second osmotic virial coefficient has been experimentally determined in prior work. The models are compared based on how well they can predict experimental behavior, their computational burdens, and scalability. An intermediate-resolution model is also introduced that can capture specific electrostatic interactions with improved efficiency and similar or improved accuracy when compared to the previously published models. Guidance is included for the selection of coarse-grained models more generally for capturing a balance of electrostatic, steric, and short-ranged nonelectrostatic interactions for proteins from low to high concentrations.
Collapse
Affiliation(s)
- Hassan Shahfar
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States.,Department of Physics and Astronomy, University of Delaware, Newark, Delaware 19716, United States
| | - James K Forder
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Christopher J Roberts
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
23
|
Krieg D, Berner C, Winter G, Svilenov HL. Biophysical Characterization of Binary Therapeutic Monoclonal Antibody Mixtures. Mol Pharm 2020; 17:2971-2986. [PMID: 32687367 DOI: 10.1021/acs.molpharmaceut.0c00370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Coformulations containing two therapeutic monoclonal antibodies (mAbs) could offer various benefits like enhanced therapeutic efficacy and better patient compliance. However, there are very few published studies on coformulations and binary mixtures of mAbs. It remains unclear to what extent mAbs with different physicochemical properties can be combined in solution without detrimental effects on protein stability. Here, we present a study including six model mAbs of the IgG1 subclass that are commercially available. In silico and biophysical characterization shows that the proteins have different physicochemical properties. Thus, their combinations represent various scenarios for coformulation development. We prepared all possible binary mixtures of the six mAbs and determined several biophysical parameters that are assessed during early-stage protein drug product development. The measured biophysical parameters are indicative of the conformational protein stability (inflection points of the thermal protein unfolding transitions) and the colloidal protein stability (aggregation onset temperatures and interaction parameter kD from dynamic light scattering). Remarkably, all 15 binary mAb mixtures do not exhibit biophysical parameters that indicate inferior conformational or colloidal stability compared to the least stable mAb in the mixture. Our findings suggest that the coformulation of some therapeutic monoclonal antibodies of the IgG1 subclass could be possible in a straightforward way as severe detrimental effects on the stability of these proteins in binary mixtures were not observed.
Collapse
Affiliation(s)
- Dennis Krieg
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5-13, Munich D-81377, Germany
| | - Carolin Berner
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5-13, Munich D-81377, Germany
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5-13, Munich D-81377, Germany
| | - Hristo L Svilenov
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5-13, Munich D-81377, Germany.,Department of Chemistry, Technische Universitaet Muenchen, Garching 85747, Germany
| |
Collapse
|
24
|
Kulakova A, Indrakumar S, Sønderby Tuelung P, Mahapatra S, Streicher WW, Peters GHJ, Harris P. Albumin-neprilysin fusion protein: understanding stability using small angle X-ray scattering and molecular dynamic simulations. Sci Rep 2020; 10:10089. [PMID: 32572086 PMCID: PMC7308280 DOI: 10.1038/s41598-020-67002-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/27/2020] [Indexed: 12/16/2022] Open
Abstract
Fusion technology is widely used in protein-drug development to increase activity, stability, and bioavailability of protein therapeutics. Fusion proteins, like any other type of biopharmaceuticals, need to remain stable during production and storage. Due to the high complexity and additional intramolecular interactions, it is not possible to predict the behavior of fusion proteins based on the behavior the individual proteins. Therefore, understanding the stability of fusion proteins on the molecular level is crucial for the development of biopharmaceuticals. The current study on the albumin-neprilysin (HSA-NEP) fusion protein uses a combination of thermal and chemical unfolding with small angle X-ray scattering and molecular dynamics simulations to show a correlation between decreasing stability and increasing repulsive interactions, which is unusual for most biopharmaceuticals. It is also seen that HSA-NEP is not fully flexible: it is present in both compact and extended conformations. Additionally, the volume fraction of each conformation changes with pH. Finally, the presence of NaCl and arginine increases stability at pH 6.5, but decreases stability at pH 5.0.
Collapse
Affiliation(s)
- Alina Kulakova
- Department of Chemistry, Technical University of Denmark, Kemitorvet building 207, Kgs. Lyngby, 2800, Denmark
| | - Sowmya Indrakumar
- Department of Chemistry, Technical University of Denmark, Kemitorvet building 207, Kgs. Lyngby, 2800, Denmark
| | - Pernille Sønderby Tuelung
- Department of Chemistry, Technical University of Denmark, Kemitorvet building 207, Kgs. Lyngby, 2800, Denmark
| | | | | | - Günther H J Peters
- Department of Chemistry, Technical University of Denmark, Kemitorvet building 207, Kgs. Lyngby, 2800, Denmark
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark, Kemitorvet building 207, Kgs. Lyngby, 2800, Denmark.
| |
Collapse
|
25
|
Augustijn D, Kulakova A, Mahapatra S, Harris P, Rinnan Å. Isothermal Chemical Denaturation: Data Analysis, Error Detection, and Correction by PARAFAC2. Anal Chem 2020; 92:6958-6967. [PMID: 32323977 DOI: 10.1021/acs.analchem.9b05748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Characterization of a protein's conformational stability is a key step in the development of biotherapeutics, where protein unfolding leads to adverse properties, such as aggregation and loss of efficacy. Isothermal chemical denaturation (ICD) can be applied to determine chemical stability, aiming to identify the optimal solvent conditions, in terms of pH, salt concentration, and added excipients. For seven monoclonal antibodies, this study investigates the observed intrinsic protein fluorescence emission spectra as a function of denaturant concentration. Protein formulations are screened in two experimental series. We show how the peak shapes of folded and unfolded proteins are preserved under added salt (0-140 mM NaCl) and added excipients concentrations, as typically found in biotherapeutic formulations and that only minor effects in tryptophan fluorescence peak tailing are observed over a large pH range (5.5-9.0). The data of seven mAbs, where GuHCl was a suitable denaturant, are modeled using PARAFAC2. PARAFAC2, a linear decomposition method, is well suited for the data and yields robust, valid, and automated models that allow for the detection of erroneous measurements. Analysis of the errors show correlation with the well-based experimental setup, and differences in observed errors between the two experimental series. We additionally show a correction method for these outliers based on PARAFAC2 model scores, such that full transition curves can be retrieved, increasing the accuracy of any subsequent analysis.
Collapse
Affiliation(s)
- Dillen Augustijn
- Department of Food Science, Faculty of Life Sciences, University of Copenhagen, Rolighedsvej 30, DK-1958 Frederiksberg C, Denmark
| | - Alina Kulakova
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Sujata Mahapatra
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark.,Novozymes A/S, Biologiens Vej 2, 2800 Kongens Lyngby, Denmark
| | - Pernille Harris
- Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Åsmund Rinnan
- Department of Food Science, Faculty of Life Sciences, University of Copenhagen, Rolighedsvej 30, DK-1958 Frederiksberg C, Denmark
| |
Collapse
|
26
|
Gentiluomo L, Roessner D, Frieß W. Application of machine learning to predict monomer retention of therapeutic proteins after long term storage. Int J Pharm 2020; 577:119039. [PMID: 31953088 DOI: 10.1016/j.ijpharm.2020.119039] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/06/2020] [Accepted: 01/11/2020] [Indexed: 12/11/2022]
Abstract
An important aspect of initial developability assessments as well formulation development and selection of therapeutic proteins is the evaluation of data obtained under accelerated stress condition, i.e. at elevated temperatures. We propose the application of artificial neural networks (ANNs) to predict long term stability in real storage condition from accelerated stability studies and other high-throughput biophysical properties e.g. the first apparent temperature of unfolding (Tm). Our models have been trained on therapeutic relevant proteins, including monoclonal antibodies, in various pharmaceutically relevant formulations. Further, we developed network architectures with good prediction power using the least amount of input features, i.e. experimental effort to train the network. This provides an empiric means to highlight the most important parameters in the prediction of real-time protein stability. Further, several models were developed by a different validation means (i.e. leave-one-protein-out cross-validation) to test the robustness and the limitations of our approach. Finally, we apply surrogate machine learning algorithms (e.g. linear regression) to build trust in the ANNs decision making procedure and to highlight the connection between the leading inputs and the outputs.
Collapse
Affiliation(s)
- Lorenzo Gentiluomo
- Wyatt Technology Europe GmbH, Hochstrasse 18, 56307 Dernbach, Germany; Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, 81377 Munich, Germany.
| | - Dierk Roessner
- Wyatt Technology Europe GmbH, Hochstrasse 18, 56307 Dernbach, Germany
| | - Wolfgang Frieß
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilians-Universitaet Muenchen, Butenandtstrasse 5, 81377 Munich, Germany
| |
Collapse
|