1
|
Malani H, Kumar S, Rathore AS. Elucidation of Mg 2+ induced size and charge heterogeneity in monoclonal antibody therapeutics. Int J Biol Macromol 2024; 283:137736. [PMID: 39551289 DOI: 10.1016/j.ijbiomac.2024.137736] [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: 06/14/2024] [Revised: 11/01/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Changes in charge variant profile are known to affect mAb stability and vice versa. This report elucidates the effects of magnesium metal (0.5 mM Mg2+) on trastuzumab (IgG1 antibody). Mg2+ is often used as an excipient (50-100 mM) and lubricant (5-10 % w/w) in biopharmaceutical formulations. Analytical size-exclusion chromatography (SEC) and cation-exchange chromatography (CEX) coupled with mass spectrometry (MS) were used to evaluate the size and charge heterogeneity in the thermal and metal stressed samples and compared to the control sample (room temperature). The present study unveils that presence of Mg2+ significantly increases the rate of aggregation with 9 % aggregation observed in Mg2+ stressed samples as compared to that from thermal stress (~2 %) or control sample (<1 %). Similarly, a 2-fold elevation in acidic variants was observed both in presence of Mg2+ and thermal stress, when contrasted with the control sample. Application of stress also led to the formation of 17 additional chemical modifications (7 due to thermal stress and 10 due to Mg2+ stress) which were not identified in control, predominantly involving deamidation, isomerization of aspartic acid, oxidation, and succinimide modifications. The results indicate the need for a detailed analysis of the impact of presence of metals in biotherapeutic formulations.
Collapse
Affiliation(s)
- Himanshu Malani
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Sunil Kumar
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.
| |
Collapse
|
2
|
Zuo C, Zhou J, Bian S, Zhang Q, Lei Y, Shen Y, Chen Z, Ye P, Shi L, Mu M, Qu JH, Jiang Z, Wang Q. Comparative study of trastuzumab modification analysis using mono/multi-epitope affinity technology with LC-QTOF-MS. J Pharm Anal 2024; 14:101015. [PMID: 39698314 PMCID: PMC11652880 DOI: 10.1016/j.jpha.2024.101015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/19/2024] [Accepted: 05/30/2024] [Indexed: 12/20/2024] Open
Abstract
Dynamic tracking analysis of monoclonal antibodies (mAbs) biotransformation in vivo is crucial, as certain modifications could inactivate the protein and reduce drug efficacy. However, a particular challenge (i.e. immune recognition deficiencies) in biotransformation studies may arise when modifications occur at the paratope recognized by the antigen. To address this limitation, a multi-epitope affinity technology utilizing the metal organic framework (MOF)@Au@peptide@aptamer composite material was proposed and developed by simultaneously immobilizing complementarity determining region (CDR) mimotope peptide (HH24) and non-CDR mimotope aptamer (CH1S-6T) onto the surface of MOF@Au nanocomposite. Comparative studies demonstrated that MOF@Au@peptide@aptamer exhibited significantly enhanced enrichment capabilities for trastuzumab variants in comparison to mono-epitope affinity technology. Moreover, the higher deamidation ratio for LC-Asn-30 and isomerization ratio for HC-Asn-55 can only be monitored by the novel bioanalytical platform based on MOF@Au@peptide@aptamer and liquid chromatography-quadrupole time of flight-mass spectrometry (LC-QTOF-MS). Therefore, multi-epitope affinity technology could effectively overcome the biases of traditional affinity materials for key sites modification analysis of mAb. Particularly, the novel bioanalytical platform can be successfully used for the tracking analysis of trastuzumab modifications in different biological fluids. Compared to the spiked phosphate buffer (PB) model, faster modification trends were monitored in the spiked serum and patients' sera due to the catalytic effect of plasma proteins and relevant proteases. Differences in peptide modification levels of trastuzumab in patients' sera were also monitored. In summary, the novel bioanalytical platform based on the multi-epitope affinity technology holds great potentials for in vivo biotransformation analysis of mAb, contributing to improved understanding and paving the way for future research and clinical applications.
Collapse
Affiliation(s)
- Chengyi Zuo
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research of China, Jinan University, Guangzhou, 510632, China
| | - Jingwei Zhou
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research of China, Jinan University, Guangzhou, 510632, China
| | - Sumin Bian
- School of Engineering, Westlake University, Hangzhou, 310024, China
| | - Qing Zhang
- The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Yutian Lei
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, Guang dong, 518107, China
| | - Yuan Shen
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research of China, Jinan University, Guangzhou, 510632, China
| | - Zhiwei Chen
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research of China, Jinan University, Guangzhou, 510632, China
| | - Peijun Ye
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research of China, Jinan University, Guangzhou, 510632, China
| | - Leying Shi
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research of China, Jinan University, Guangzhou, 510632, China
| | - Mao Mu
- Guangdong Institute for Drug Control, Guangzhou, 510663, China
| | - Jia-Huan Qu
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research of China, Jinan University, Guangzhou, 510632, China
| | - Zhengjin Jiang
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research of China, Jinan University, Guangzhou, 510632, China
| | - Qiqin Wang
- Institute of Pharmaceutical Analysis, College of Pharmacy/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research of China, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
3
|
Harris CT, Cohen S. Reducing Immunogenicity by Design: Approaches to Minimize Immunogenicity of Monoclonal Antibodies. BioDrugs 2024; 38:205-226. [PMID: 38261155 PMCID: PMC10912315 DOI: 10.1007/s40259-023-00641-2] [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] [Accepted: 12/13/2023] [Indexed: 01/24/2024]
Abstract
Monoclonal antibodies (mAbs) have transformed therapeutic strategies for various diseases. Their high specificity to target antigens makes them ideal therapeutic agents for certain diseases. However, a challenge to their application in clinical practice is their potential risk to induce unwanted immune response, termed immunogenicity. This challenge drives the continued efforts to deimmunize these protein therapeutics while maintaining their pharmacokinetic properties and therapeutic efficacy. Because mAbs hold a central position in therapeutic strategies against an array of diseases, the importance of conducting comprehensive immunogenicity risk assessment during the drug development process cannot be overstated. Such assessment necessitates the employment of in silico, in vitro, and in vivo strategies to evaluate the immunogenicity risk of mAbs. Understanding the intricacies of the mechanisms that drive mAb immunogenicity is crucial to improving their therapeutic efficacy and safety and developing the most effective strategies to determine and mitigate their immunogenic risk. This review highlights recent advances in immunogenicity prediction strategies, with a focus on protein engineering strategies used throughout development to reduce immunogenicity.
Collapse
Affiliation(s)
- Chantal T Harris
- Department of BioAnalytical Sciences, Genentech Inc., South San Francisco, CA, 94080-4990, USA
| | - Sivan Cohen
- Department of BioAnalytical Sciences, Genentech Inc., South San Francisco, CA, 94080-4990, USA.
| |
Collapse
|
4
|
Lteif M, Pallardy M, Turbica I. Antibodies internalization mechanisms by dendritic cells and their role in therapeutic antibody immunogenicity. Eur J Immunol 2024; 54:e2250340. [PMID: 37985174 DOI: 10.1002/eji.202250340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Internalization and processing by antigen-presenting cells such as dendritic cells (DCs) are critical steps for initiating a T-cell response to therapeutic antibodies. Consequences are the production of neutralizing antidrug antibodies altering the clinical response, the presence of immune complexes, and, in some rare cases, hypersensitivity reactions. In recent years, significant progress has been made in the knowledge of cellular uptake mechanisms of antibodies in DCs. The uptake of antibodies could be directly related to their immunogenicity by regulating the quantity of materials entering the DCs in relation to antibody structure. Here, we summarize the latest insights into cellular uptake mechanisms and pathways in DCs. We highlight the approaches to study endocytosis, the impact of endocytosis routes on T-cell response, and discuss the link between how DCs internalize therapeutic antibodies and the potential mechanisms that could give rise to immunogenicity. Understanding these processes could help in developing assays to evaluate the immunogenicity potential of biotherapeutics.
Collapse
Affiliation(s)
- Maria Lteif
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Marc Pallardy
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Isabelle Turbica
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| |
Collapse
|
5
|
Pérez-Robles R, Salmerón-García A, Hermosilla J, Torrente-López A, Clemente-Bautista S, Jiménez-Lozano I, Cabañas-Poy MJ, Cabeza J, Navas N. Comprehensive physicochemical characterization of a peptide-based medicine: Teduglutide (Revestive®) structural description and stress testing. Eur J Pharm Biopharm 2023; 184:103-115. [PMID: 36669672 DOI: 10.1016/j.ejpb.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023]
Abstract
Teduglutide (Revestive®) is a glucagon-like peptide-2 analogue used for the treatment of short bowel syndrome, a rare life-threatening condition in which the amount of functional gut is too short to enable proper absorption of nutrients and fluids. During handling prior to administration to the patient in hospital, it is possible that peptide-based medicines may be exposed to environmental stress conditions that could affect their quality. It is therefore essential to carry out stress testing studies to evaluate how such medicines respond to these stresses. For this reason, in this paper we present a strategy for a comprehensive analytical characterization of a peptide and a stress testing study in which it was subjected to various stress conditions: heating at 40 °C and 60 °C, light exposure and shaking. Several complementary analytical techniques were used throughout this study: Far UV circular dichroism, intrinsic protein fluorescence spectroscopy, dynamic light scattering, size-exclusion chromatography and intact and peptide mapping reverse-phase chromatography coupled to mass spectrometry. To the best of our knowledge, this is the first study to offer an in-depth description of the chemical structure of teduglutide peptide and its physicochemical characteristics after stress stimuli were applied to the reconstituted medicine Revestive®.
Collapse
Affiliation(s)
- Raquel Pérez-Robles
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Department of Analytical Chemistry, Science Faculty, University of Granada, Granada, Spain; Fundación para la Investigación Biosanitaria de Andalucía Oriental-Alejandro Otero, Granada, Spain
| | - Antonio Salmerón-García
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Department of Clinical Pharmacy, San Cecilio University Hospital, Granada, Spain
| | - Jesus Hermosilla
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Department of Analytical Chemistry, Science Faculty, University of Granada, Granada, Spain
| | - Anabel Torrente-López
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Department of Analytical Chemistry, Science Faculty, University of Granada, Granada, Spain
| | | | - Inés Jiménez-Lozano
- Maternal and Child Pharmacy Service, Vall d'Hebron Hospital, Pharmacy, Barcelona, Spain
| | | | - Jose Cabeza
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Department of Clinical Pharmacy, San Cecilio University Hospital, Granada, Spain
| | - Natalia Navas
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain; Department of Analytical Chemistry, Science Faculty, University of Granada, Granada, Spain.
| |
Collapse
|
6
|
Mieczkowski C, Zhang X, Lee D, Nguyen K, Lv W, Wang Y, Zhang Y, Way J, Gries JM. Blueprint for antibody biologics developability. MAbs 2023; 15:2185924. [PMID: 36880643 PMCID: PMC10012935 DOI: 10.1080/19420862.2023.2185924] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/24/2023] [Indexed: 03/08/2023] Open
Abstract
Large-molecule antibody biologics have revolutionized medicine owing to their superior target specificity, pharmacokinetic and pharmacodynamic properties, safety and toxicity profiles, and amenability to versatile engineering. In this review, we focus on preclinical antibody developability, including its definition, scope, and key activities from hit to lead optimization and selection. This includes generation, computational and in silico approaches, molecular engineering, production, analytical and biophysical characterization, stability and forced degradation studies, and process and formulation assessments. More recently, it is apparent these activities not only affect lead selection and manufacturability, but ultimately correlate with clinical progression and success. Emerging developability workflows and strategies are explored as part of a blueprint for developability success that includes an overview of the four major molecular properties that affect all developability outcomes: 1) conformational, 2) chemical, 3) colloidal, and 4) other interactions. We also examine risk assessment and mitigation strategies that increase the likelihood of success for moving the right candidate into the clinic.
Collapse
Affiliation(s)
- Carl Mieczkowski
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Xuejin Zhang
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Dana Lee
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Khanh Nguyen
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Wei Lv
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Yanling Wang
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Yue Zhang
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Jackie Way
- Department of Protein Sciences, Hengenix Biotech, Inc, Milpitas, CA, USA
| | - Jean-Michel Gries
- President, Discovery Research, Hengenix Biotech, Inc, Milpitas, CA, USA
| |
Collapse
|
7
|
Akbarian M, Chen SH. Instability Challenges and Stabilization Strategies of Pharmaceutical Proteins. Pharmaceutics 2022; 14:2533. [PMID: 36432723 PMCID: PMC9699111 DOI: 10.3390/pharmaceutics14112533] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Maintaining the structure of protein and peptide drugs has become one of the most important goals of scientists in recent decades. Cold and thermal denaturation conditions, lyophilization and freeze drying, different pH conditions, concentrations, ionic strength, environmental agitation, the interaction between the surface of liquid and air as well as liquid and solid, and even the architectural structure of storage containers are among the factors that affect the stability of these therapeutic biomacromolecules. The use of genetic engineering, side-directed mutagenesis, fusion strategies, solvent engineering, the addition of various preservatives, surfactants, and additives are some of the solutions to overcome these problems. This article will discuss the types of stress that lead to instabilities of different proteins used in pharmaceutics including regulatory proteins, antibodies, and antibody-drug conjugates, and then all the methods for fighting these stresses will be reviewed. New and existing analytical methods that are used to detect the instabilities, mainly changes in their primary and higher order structures, are briefly summarized.
Collapse
Affiliation(s)
| | - Shu-Hui Chen
- Department of Chemistry, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|