1
|
Döring S, Weller MG, Reinders Y, Konthur Z, Jaeger C. Challenges and Insights in Absolute Quantification of Recombinant Therapeutic Antibodies by Mass Spectrometry: An Introductory Review. Antibodies (Basel) 2025; 14:3. [PMID: 39846611 PMCID: PMC11755444 DOI: 10.3390/antib14010003] [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: 11/15/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 01/24/2025] Open
Abstract
This review describes mass spectrometry (MS)-based approaches for the absolute quantification of therapeutic monoclonal antibodies (mAbs), focusing on technical challenges in sample treatment and calibration. Therapeutic mAbs are crucial for treating cancer and inflammatory, infectious, and autoimmune diseases. We trace their development from hybridoma technology and the first murine mAbs in 1975 to today's chimeric and fully human mAbs. With increasing commercial relevance, the absolute quantification of mAbs, traceable to an international standard system of units (SI units), has attracted attention from science, industry, and national metrology institutes (NMIs). Quantification of proteotypic peptides after enzymatic digestion using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) has emerged as the most viable strategy, though methods targeting intact mAbs are still being explored. We review peptide-based quantification, focusing on critical experimental steps like denaturation, reduction, alkylation, choice of digestion enzyme, and selection of signature peptides. Challenges in amino acid analysis (AAA) for quantifying pure mAbs and peptide calibrators, along with software tools for targeted MS data analysis, are also discussed. Short explanations within each chapter provide newcomers with an overview of the field's challenges. We conclude that, despite recent progress, further efforts are needed to overcome the many technical hurdles along the quantification workflow and discuss the prospects of developing standardized protocols and certified reference materials (CRMs) for this goal. We also suggest future applications of newer technologies for absolute mAb quantification.
Collapse
Affiliation(s)
- Sarah Döring
- Federal Institute of Material Testing and Research (BAM), 12489 Berlin, Germany; (S.D.); (M.G.W.); (Z.K.)
| | - Michael G. Weller
- Federal Institute of Material Testing and Research (BAM), 12489 Berlin, Germany; (S.D.); (M.G.W.); (Z.K.)
| | - Yvonne Reinders
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V., 44139 Dortmund, Germany;
| | - Zoltán Konthur
- Federal Institute of Material Testing and Research (BAM), 12489 Berlin, Germany; (S.D.); (M.G.W.); (Z.K.)
| | - Carsten Jaeger
- Federal Institute of Material Testing and Research (BAM), 12489 Berlin, Germany; (S.D.); (M.G.W.); (Z.K.)
| |
Collapse
|
2
|
Lakis R, Sauvage FL, Pinault E, Marquet P, Saint-Marcoux F, El Balkhi S. Absolute Quantification of Human Serum Albumin Isoforms by Internal Calibration Based on a Top-Down LC-MS Approach. Anal Chem 2024; 96:746-755. [PMID: 38166371 DOI: 10.1021/acs.analchem.3c03933] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Well-characterized biomarkers using reliable quantitative methods are essential for the management of various pathologies such as diabetes, kidney, and liver diseases. Human serum albumin (HSA) isoforms are gaining interest as biomarkers of advanced liver pathologies. In view of the structural alterations observed for HSA, insights into its isoforms are required to establish them as reliable biomarkers. Therefore, a robust absolute quantification method seems necessary. In this study, we developed and validated a far more advanced top-down liquid chromatography-mass spectrometry (LC-MS) method for the absolute quantification of HSA isoforms, using myoglobin (Mb) as an internal standard for quantification and for mass recalibration. Two different quantification approaches were investigated based on peak integration from the deconvoluted spectrum and extracted ion chromatogram (XIC). The protein mixture human serum albumin/myoglobin eluted in well-shaped separated peaks. Mb allowed a systematic mass recalibration for every sample, resulting in extremely low mass deviations compared to conventional deconvolution-based methods. In total, eight HSA isoforms of interest were quantified. Specific-isoform calibration curves showing good linearity were obtained by using the deconvoluted peaks. Noticeably, the HSA ionization behavior appeared to be isoform-dependent, suggesting that the use of an enriched isoform solution as a calibration standard for absolute quantification studies of HSA isoforms is necessary. Good repeatability, reproducibility, and accuracy were observed, with better sensitivity for samples with low albumin concentrations compared to routine biochemical assays. With a relatively simple workflow, the application of this method for absolute quantification shows great potential, especially for HSA isoform studies in a clinical context, where a high-throughput method and sensitivity are needed.
Collapse
Affiliation(s)
- Roy Lakis
- Pharmacology & Transplantation (P&T), Université de Limoges, INSERM U1248, Limoges 87000, France
| | - François-Ludovic Sauvage
- Pharmacology & Transplantation (P&T), Université de Limoges, INSERM U1248, Limoges 87000, France
| | - Emilie Pinault
- Pharmacology & Transplantation (P&T), Université de Limoges, INSERM U1248, Limoges 87000, France
| | - Pierre Marquet
- Pharmacology & Transplantation (P&T), Université de Limoges, INSERM U1248, Limoges 87000, France
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges 87000, France
| | - Franck Saint-Marcoux
- Pharmacology & Transplantation (P&T), Université de Limoges, INSERM U1248, Limoges 87000, France
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges 87000, France
| | - Souleiman El Balkhi
- Pharmacology & Transplantation (P&T), Université de Limoges, INSERM U1248, Limoges 87000, France
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges 87000, France
| |
Collapse
|
3
|
Massonnet P, Grifnée E, Farré-Segura J, Demeuse J, Huyghebaert L, Dubrowski T, Dufour P, Schoumacher M, Peeters S, Le Goff C, Cavalier E. Concise review on the combined use of immunocapture, mass spectrometry and liquid chromatography for clinical applications. Clin Chem Lab Med 2023; 61:1700-1707. [PMID: 37128992 DOI: 10.1515/cclm-2023-0253] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
Immunocapture is now a well-established method for sample preparation prior to quantitation of peptides and proteins in complex matrices. This short review will give an overview of some clinical applications of immunocapture methods, as well as protocols with and without enzymatic digestion in a clinical context. The advantages and limitations of both approaches are discussed in detail. Challenges related to the choice of mass spectrometer are also discussed. Top-down, middle-down, and bottom-up approaches are discussed. Even though immunocapture has its limitations, its main advantage is that it provides an additional dimension of separation and/or isolation when working with peptides and proteins. Overall, this short review demonstrates the potential of such techniques in the field of proteomics-based clinical medicine and paves the way for better personalized medicine.
Collapse
Affiliation(s)
- Philippe Massonnet
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Elodie Grifnée
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Jordi Farré-Segura
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Justine Demeuse
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Loreen Huyghebaert
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Thomas Dubrowski
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Patrice Dufour
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | | | - Stéphanie Peeters
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
| | - Caroline Le Goff
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| |
Collapse
|
4
|
Wang S, Wang F, Wang L, Liu Z, Liu M, Li S, Wang Y, Sun X, Jiang J. Detection of antibody-conjugate payload in cynomolgus monkey serum by a high throughput capture LC-MS/MS bioanalysis method. J Pharm Biomed Anal 2023; 227:115069. [PMID: 36854219 DOI: 10.1016/j.jpba.2022.115069] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 08/23/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022]
Abstract
Antibody-drug conjugate (ADC) plays a vital role in oncology indications. The efficacy and toxicity of ADC generally depend on the concentration of the drugs in the body system, and physiologically-based pharmacokinetic (P.K.) is a quantitative tool to understand the drug concentration in the body. To characterize the whole drug carefully, sophisticated bioanalysis was required. ADC bioanalysis generally needs multiple analysis strategies, which can accurately quantify total antibody (TAb), antibody-drug conjugate (ADC), antibody-conjugate payload (ac-payload), and free-payload. In this work, we mainly described and validated a high throughput capture Liquid Chromatography tandem-Mass Spectrometry (LC-MS/MS) bioanalysis method to detect the concentrations of ac-payload (such as MMAE) in cynomolgus monkey serum. This method was allowed to determinate the Drug to Antibody Ratio (DAR), obtained by n of ac-payload/ n of TAb. In addition, the technique could significantly improve the throughput of the pre-coated antibody on a 96-well plate. Besides, this method had no interference or carryover in endogenous substances and showed linearity (R2 ≥0.99) in the concentration range within 15.6-2000.0 ng/mL. The inter-run accuracy ranged from 75.8 % to 120.0 %, and precision was within ≤ 20.0 %. Meanwhile, selectivity and the benchtop stability of the method were also validated. This optimization method was successfully applied to the change of average DAR in P.K. study.
Collapse
Affiliation(s)
- Shujuan Wang
- RemeGen, Ltd, Yantai 264000, Shandong, China; Rongchang Industry College, Yantai 264003, Shandong, China
| | - Fengzhu Wang
- RemeGen, Ltd, Yantai 264000, Shandong, China; School of Biological Sciences, University of California, Irvine, CA 92697, United States
| | - Ling Wang
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Zhihao Liu
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Meiling Liu
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Shenjun Li
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Ying Wang
- Pharmaron (Beijing) Inc., Beijing 100176, China
| | | | - Jing Jiang
- Rongchang Industry College, Yantai 264003, Shandong, China; Department of Pharmacology, Binzhou Medical University, Yantai 264003, Shandong, China.
| |
Collapse
|
5
|
Moran AB, Domínguez-Vega E, Wuhrer M, Lageveen-Kammeijer GSM. Software-Assisted Data Processing Workflow for Intact Glycoprotein Mass Spectrometry. J Proteome Res 2023; 22:1367-1376. [PMID: 36857466 PMCID: PMC10088042 DOI: 10.1021/acs.jproteome.2c00762] [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: 03/03/2023]
Abstract
Intact protein analysis by mass spectrometry is important for several applications such as assessing post-translational modifications and biotransformation. In particular, intact protein analysis allows the detection of proteoforms that are commonly missed by other approaches such as proteolytic digestion followed by bottom-up analysis. Two quantification methods are mainly used for intact protein data quantification, namely the extracted ion and deconvolution approaches. However, a consensus with regard to a single best practice for intact protein data processing is lacking. Furthermore, many data processing tools are not fit-for-purpose and, as a result, the analysis of intact proteins is laborious and lacks the throughput required to be implemented for the analysis of clinical cohorts. Therefore, in this study, we investigated the application of a software-assisted data analysis and processing workflow in order to streamline intact protein integration, annotation, and quantification via deconvolution. In addition, the assessment of orthogonal data sets generated via middle-up and bottom-up analysis enabled the cross-validation of cleavage proteoform assignments present in seminal prostate-specific antigen (PSA). Furthermore, deconvolution quantification of PSA from patients' urine revealed results that were comparable with manually performed quantification based on extracted ion electropherograms. Overall, the presented workflow allows fast and efficient processing of intact protein data. The raw data is available on MassIVE using the identifier MSV000086699.
Collapse
Affiliation(s)
- Alan B Moran
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Elena Domínguez-Vega
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Guinevere S M Lageveen-Kammeijer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands
| |
Collapse
|
6
|
Kellie JF, Schneck NA, Causon JC, Baba T, Mehl JT, Pohl KI. Top-Down Characterization and Intact Mass Quantitation of a Monoclonal Antibody Drug from Serum by Use of a Quadrupole TOF MS System Equipped with Electron-Activated Dissociation. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:17-26. [PMID: 36459688 DOI: 10.1021/jasms.2c00206] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Time-of-flight MS systems for biopharmaceutical and protein characterization applications may play an even more pivotal role in the future as biotherapeutics increase in drug pipelines and as top-down MS approaches increase in use. Here, a recently developed TOF MS system is examined for monoclonal antibody (mAb) characterization from serum samples. After immunocapture, purified drug material spiked into monkey serum or dosed for an in-life study is analyzed by top-down MS. While characterization aspects are a distinct advantage of the MS platform, MS system and software capabilities are also shown regarding intact protein quantitation. Such applications are demonstrated to help enable comprehensive protein molecule quantitation and characterization by use of TOF MS instrumentation.
Collapse
Affiliation(s)
- John F Kellie
- GSK, Collegeville, Pennsylvania 19426, United States
| | | | | | | | - John T Mehl
- GSK, Collegeville, Pennsylvania 19426, United States
| | | |
Collapse
|
7
|
Determination of drug-to-antibody ratio of antibody-drug conjugate in biological samples using microflow-liquid chromatography/high-resolution mass spectrometry. Bioanalysis 2022; 14:1533-1545. [PMID: 36825963 DOI: 10.4155/bio-2022-0219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Background: Antibody-drug conjugates (ADCs) are a promising modality for cancer treatment; however, considering their complicated nature, analytical complexity in understanding their pharmacokinetics and pharmacodynamics in the body presents a significant challenge. Results: Vorsetuzumab maleimidocaproyl valine-citrulline p-aminobenzyloxycarbonyl monomethyl auristatin E was used to develop pretreatment and analytical workflows suitable for ADCs. Monomethyl auristatin E release and drug-to-antibody ratio retention were consistent in mouse plasma but inconsistent in monkey and human plasma. Further, metabolites were species-specific. Microflow-liquid chromatography/high-resolution mass spectrometry (LC-HRMS) resulted in a 4-7-fold improvement in detection sensitivity compared with conventional flow LC-HRMS. Conclusion: Microflow-LC-HRMS can be a useful tool in understanding the complex properties of ADCs in the body from a drug metabolism and pharmacokinetics point of view.
Collapse
|
8
|
DelGuidice CE, Ismaiel OA, Mylott WR, Yuan M, Halquist MS. Intact quantitative bioanalytical method development and fit-for-purpose validation of a monoclonal antibody and its related fab fragment in human vitreous and aqueous humor using LC-HRMS. Anal Bioanal Chem 2022; 414:4189-4202. [PMID: 35451621 DOI: 10.1007/s00216-022-04071-x] [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: 09/29/2021] [Revised: 03/27/2022] [Accepted: 04/06/2022] [Indexed: 11/29/2022]
Abstract
Ranibizumab is an FDA-approved drug used to treat wet age-related macular degeneration (AMD), diabetic retinopathy, macular edema, and myopic choroidal neovascularization. Bevacizumab is another drug often used off-label to treat wet AMD. In order to reduce unwanted angiogenesis, ranibizumab and bevacizumab target circulating VEGF-A in the eye. Concentration levels in human vitreous and aqueous humor can be used to provide valuable efficacy information. However, vitreous and aqueous humor's aqueous environment, and vitreous humor's viscosity, as well as the stickiness of the analytes can provide bioanalytical challenges. In this manuscript, we describe the development, optimization, and fit-for-purpose validation of an LC-HRMS method designed for intact quantitative bioanalysis of ranibizumab and bevacizumab in human vitreous and aqueous humor following intravitreal administration. In order to fully develop this method, evaluations were conducted to optimize the conditions, including the data processing model (extracted ion chromatograms (XICs) vs deconvolution), carryover mitigation, sample preparation scheme optimization for surrogate and primary matrices, use of internal standard/immunocapture/deglycosylation, and optimization of the extraction and dilution procedure, as well as optimization of the liquid chromatography and mass spectrometry conditions. Once the method was fully optimized, a fit-for-purpose validation was conducted, including matrix parallelism, with a linear calibration range of 10 to 200 µg/mL. The development of this intact quantitative method using LC-HRMS provides a proof-of-concept template for challenging, but valuable new and exciting bioanalytical techniques.
Collapse
Affiliation(s)
- Catherine E DelGuidice
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA. .,PPD Laboratories, Richmond, VA, USA.
| | - Omnia A Ismaiel
- Department of Analytical Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | | | | | - Matthew S Halquist
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
9
|
Mu R, Yuan J, Huang Y, Meissen JK, Mou S, Liang M, Rosenbaum AI. Bioanalytical Methods and Strategic Perspectives Addressing the Rising Complexity of Novel Bioconjugates and Delivery Routes for Biotherapeutics. BioDrugs 2022; 36:181-196. [PMID: 35362869 PMCID: PMC8972746 DOI: 10.1007/s40259-022-00518-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2022] [Indexed: 12/20/2022]
Abstract
In recent years, an increase in the discovery and development of biotherapeutics employing new modalities, such as bioconjugates or novel routes of delivery, has created bioanalytical challenges. The inherent complexity of conjugated molecular structures means that quantification of the bioconjugate and its multiple components is critical for preclinical/clinical studies to inform drug discovery and development. Moreover, bioconjugates involve additional multifactorial complexity because of the potential for in vivo catabolism and biotransformation, which may require thorough investigations in multiple biological matrices. Furthermore, excipients that enhance absorption are frequently evaluated and employed for the development of oral and inhaled biotherapeutics. Risk-benefit assessments are required for novel or existing excipients that utilize dosages above previously approved levels. Bioanalytical methods that can measure both excipients and potential drug metabolites in biological matrices are highly relevant to these emerging bioanalysis challenges. We discuss the bioanalytical strategies for analyzing bioconjugates such as antibody-drug conjugates and antibody-oligonucleotide conjugates and review recent advances in bioanalytical methods for the quantification and characterization of novel bioconjugates. We also discuss bioanalytical considerations for both biotherapeutics and excipients through novel administration routes and review analyses in various biological matrices, from the extensively studied serum or plasma to tissue biopsy in the context of preclinical and clinical studies from both technical and regulatory perspectives.
Collapse
Affiliation(s)
- Ruipeng Mu
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Jiaqi Yuan
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Yue Huang
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - John K Meissen
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Si Mou
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Meina Liang
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA
| | - Anton I Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA, USA.
| |
Collapse
|
10
|
Kellie JF, Tran JC, Jian W, Jones B, Mehl JT, Ge Y, Henion J, Bateman KP. Intact Protein Mass Spectrometry for Therapeutic Protein Quantitation, Pharmacokinetics, and Biotransformation in Preclinical and Clinical Studies: An Industry Perspective. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:1886-1900. [PMID: 32869982 DOI: 10.1021/jasms.0c00270] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Recent advancements in immunocapture methods and mass spectrometer technology have enabled intact protein mass spectrometry to be applied for the characterization of antibodies and other large biotherapeutics from in-life studies. Protein molecules have not been traditionally studied by intact mass or screened for catabolites in the same manner as small molecules, but the landscape has changed. Researchers have presented methods that can be applied to the drug discovery and development stages, and others are exploring the possibilities of the new approaches. However, a wide variety of options for assay development exists without clear recommendation on best practice, and data processing workflows may have limitations depending on the vendor. In this perspective, we share experiences and recommendations for current and future application of mass spectrometry for biotherapeutic molecule monitoring from preclinical and clinical studies.
Collapse
Affiliation(s)
- John F Kellie
- Bioanalysis, Immunogenicity & Biomarkers, GlaxoSmithKline, Collegeville, Pennsylvania 19426, United States
| | - John C Tran
- Biochemical & Cellular Pharmacology, Genentech Inc., South San Francisco, California 94080, United States
| | - Wenying Jian
- DMPK, Janssen Research & Development, Johnson & Johnson, Spring House, Pennsylvania 19477, United States
| | - Barry Jones
- Q Squared Solutions, 19 Brown Road, Ithaca, New York 14850, United States
| | - John T Mehl
- Bioanalytical Research, Bristol-Myers Squibb, Princeton, New Jersey 08648, United States
| | - Ying Ge
- Department of Cell and Regenerative Biology, Department of Chemistry, Human Proteomics Program, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Jack Henion
- Advion, Inc., 61 Brown Road, Ithaca, New York 14850, United States
| | - Kevin P Bateman
- PPDM, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
11
|
The role of ligand-binding assay and LC-MS in the bioanalysis of complex protein and oligonucleotide therapeutics. Bioanalysis 2021; 13:931-954. [PMID: 33998268 DOI: 10.4155/bio-2021-0009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ligand-binding assay (LBA) and LC-MS have been the preferred bioanalytical techniques for the quantitation and biotransformation assessment of various therapeutic modalities. This review provides an overview of the applications of LBA, LC-MS/MS and LC-HRMS for the bioanalysis of complex protein therapeutics including antibody-drug conjugates, fusion proteins and PEGylated proteins as well as oligonucleotide therapeutics. The strengths and limitations of LBA and LC-MS, along with some guidelines on the choice of appropriate bioanalytical technique(s) for the bioanalysis of these therapeutic modalities are presented. With the discovery of novel and more complex therapeutic modalities, there is an increased need for the biopharmaceutical industry to develop a comprehensive bioanalytical strategy integrating both LBA and LC-MS.
Collapse
|
12
|
Huang Y, Mou S, Wang Y, Mu R, Liang M, Rosenbaum AI. Characterization of Antibody-Drug Conjugate Pharmacokinetics and in Vivo Biotransformation Using Quantitative Intact LC-HRMS and Surrogate Analyte LC-MRM. Anal Chem 2021; 93:6135-6144. [PMID: 33835773 DOI: 10.1021/acs.analchem.0c05376] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Antibody-drug conjugates (ADCs) pose challenges to bioanalysis because of their inherently intricate structures and potential for very complex catabolism. Common bioanalysis strategy is to measure the concentration of ADCs and Total Antibody (Ab) as well as deconjugated warhead in circulation. The ADCs and the Total Ab can be quantified with ligand binding assays (LBA) or with hybrid immunocapture-liquid chromatography coupled with multiple reaction monitoring mass spectrometry (LBA-LC-MRM). With the LBA-LC-MRM approach, a surrogate analyte, often the signature peptide, and released warhead can be used for the quantification of the Total Ab and ADCs, respectively. Recent advances in analytical instrumentation, especially the development of high resolution mass spectrometers (HRMS), have enabled characterization and quantification of intact macromolecules such as ADCs. The LBA-LC-HRMS approach employs immunocapture, followed by chromatographic separation at the macromolecule level and detection of the intact analyte. We developed an intact quantification method with 1-10 μg/mL linear dynamic range using 25 μL of plasma sample volume. This method was qualified for the measurement of naked monoclonal antibody (mAb), a site-specific cysteine-conjugated ADC with drug to antibody ratio ∼2 (DAR2) and a site-nonspecific cysteine-conjugated ADC (DAR8) in rat plasma. Samples from a rat pharmacokinetic (PK) study were analyzed with both methods. For the naked mAb, the results from both assays matched well. For ADCs, new species were observed from the LBA-HRMS method. The results demonstrated that potential biotransformation of the ADC was unveiled using the intact quantification approach while not being observed with traditional LBA-LC-MRM approach. Our work demonstrated an application of novel intact quantification by supporting animal PK studies. Moreover, our results suggest that the intact quantification method can provide novel perspectives on ADC in vivo characterization and quantification, which can benefit future drug candidate optimization as well as the immunogenicity impact evaluation and safety assessment.
Collapse
Affiliation(s)
- Yue Huang
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Si Mou
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Yadi Wang
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Ruipeng Mu
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Meina Liang
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Anton I Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
13
|
Lee BI, Park SJ, Park Y, Shin SH, Choi JM, Park MJ, Lim JH, Kim SY, Lee H, Shin YG. Assessments of the In Vitro and In Vivo Linker Stability and Catabolic Fate for the Ortho Hydroxy-Protected Aryl Sulfate Linker by Immuno-Affinity Capture Liquid Chromatography Quadrupole Time-of-Flight Mass Spectrometric Assay. Pharmaceutics 2021; 13:pharmaceutics13010125. [PMID: 33478046 PMCID: PMC7836004 DOI: 10.3390/pharmaceutics13010125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/20/2022] Open
Abstract
Antibody-drug conjugate (ADC) linkers play an important role in determining the safety and efficacy of ADC. The Ortho Hydroxy-Protected Aryl Sulfate (OHPAS) linker is a newly developed linker in the form of a di-aryl sulfate structure consisting of phenolic payload and self-immolative group (SIG). In this study, using two bioanalytical approaches (namely "bottom-up" and "middle-up" approaches) via the liquid chromatography-quadrupole time-of-flight mass spectrometric (LC-qTOF-MS) method, in vitro and in vivo linker stability experiments were conducted for the OHPAS linker. For comparison, the valine-citrulline-p-aminobenzyloxycarbonyl (VC-PABC) linker was also evaluated under the same experimental conditions. In addition, the catabolite identification experiments at the subunit intact protein level were simultaneously performed to evaluate the catabolic fate of ADCs. As a result, the OHPAS linker was stable in the in vitro mouse/human plasma as well as in vivo pharmacokinetic studies in mice, whereas the VC-PABC linker was relatively unstable in mice in vitro and in vivo. This is because the VC-PABC linker was sensitive to a hydrolytic enzyme called carboxylesterase 1c (Ces1c) in mouse plasma. In conclusion, the OHPAS linker appears to be a good linker for ADC, and further experiments would be warranted to demonstrate the efficacy and toxicity related to the OHPAS linker.
Collapse
Affiliation(s)
- Byeong ill Lee
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea; (B.i.L.); (S.-j.P.); (Y.P.); (S.-H.S.); (J.-m.C.); (M.-j.P.); (J.-h.L.)
| | - Seo-jin Park
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea; (B.i.L.); (S.-j.P.); (Y.P.); (S.-H.S.); (J.-m.C.); (M.-j.P.); (J.-h.L.)
| | - Yuri Park
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea; (B.i.L.); (S.-j.P.); (Y.P.); (S.-H.S.); (J.-m.C.); (M.-j.P.); (J.-h.L.)
| | - Seok-Ho Shin
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea; (B.i.L.); (S.-j.P.); (Y.P.); (S.-H.S.); (J.-m.C.); (M.-j.P.); (J.-h.L.)
| | - Jang-mi Choi
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea; (B.i.L.); (S.-j.P.); (Y.P.); (S.-H.S.); (J.-m.C.); (M.-j.P.); (J.-h.L.)
| | - Min-jae Park
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea; (B.i.L.); (S.-j.P.); (Y.P.); (S.-H.S.); (J.-m.C.); (M.-j.P.); (J.-h.L.)
| | - Jeong-hyeon Lim
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea; (B.i.L.); (S.-j.P.); (Y.P.); (S.-H.S.); (J.-m.C.); (M.-j.P.); (J.-h.L.)
| | - Sun Young Kim
- IntoCell Inc., 101, Sinildong-ro, Daedeok-gu, Daejeon 34324, Korea; (S.Y.K.); (H.L.)
| | - Hyangsook Lee
- IntoCell Inc., 101, Sinildong-ro, Daedeok-gu, Daejeon 34324, Korea; (S.Y.K.); (H.L.)
| | - Young G. Shin
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon 34134, Korea; (B.i.L.); (S.-j.P.); (Y.P.); (S.-H.S.); (J.-m.C.); (M.-j.P.); (J.-h.L.)
- Correspondence: ; Tel.: +82-42-821-5931
| |
Collapse
|
14
|
Cahuzac H, Devel L. Analytical Methods for the Detection and Quantification of ADCs in Biological Matrices. Pharmaceuticals (Basel) 2020; 13:ph13120462. [PMID: 33327644 PMCID: PMC7765153 DOI: 10.3390/ph13120462] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 12/27/2022] Open
Abstract
Understanding pharmacokinetics and biodistribution of antibody–drug conjugates (ADCs) is a one of the critical steps enabling their successful development and optimization. Their complex structure combining large and small molecule characteristics brought out multiple bioanalytical methods to decipher the behavior and fate of both components in vivo. In this respect, these methods must provide insights into different key elements including half-life and blood stability of the construct, premature release of the drug, whole-body biodistribution, and amount of the drug accumulated within the targeted pathological tissues, all of them being directly related to efficacy and safety of the ADC. In this review, we will focus on the main strategies enabling to quantify and characterize ADCs in biological matrices and discuss their associated technical challenges and current limitations.
Collapse
|
15
|
SHIBATA Y, YAMADA T, SUGIYAMA E, MIZUNO H, TODOROKI K. Sensitive Method for LC Analysis of Therapeutic Monoclonal Antibodies Using a Centrifugal Filtration Device with Adsorption Suppression Treatment. CHROMATOGRAPHY 2020. [DOI: 10.15583/jpchrom.2020.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Yosuke SHIBATA
- School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Eiji SUGIYAMA
- School of Pharmaceutical Sciences, University of Shizuoka
| | - Hajime MIZUNO
- School of Pharmaceutical Sciences, University of Shizuoka
| | | |
Collapse
|
16
|
Zhu L, Glick J, Flarakos J. Bioanalytical Challenges in Support of Complex Modalities of Antibody-Based Therapeutics. AAPS JOURNAL 2020; 22:130. [PMID: 33037499 DOI: 10.1208/s12248-020-00517-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/28/2020] [Indexed: 01/23/2023]
Abstract
Antibody-based therapeutic classes are evolving from monoclonal antibodies to antibody derivatives with complex structures to achieve advanced therapeutic effect. These antibody derivatives may contain multiple functional domains and are often vulnerable to in vivo biotransformation. Understanding the pharmacokinetics of these antibody derivatives requires a sophisticated bioanalytical approach to carefully characterize the whole drug and each functional domain with respect to quantity, functionality enabled by biotransformation, and corresponding immune responses. Ligand binding assays and liquid chromatography-mass spectrometry assays are predominantly used in bioanalytical support of monoclonal antibodies and are continuously used for antibody derivatives such as antibody drug conjugate and bispecific antibodies. However, they become increasingly cumbersome in coping with increased complexity of drug modality and associated biotransformation. In this mini-review, we examined the current pharmacokinetic assays in the literature for antibody drug conjugate and bispecific antibodies, and presented our view of promising bioanalytical technologies to address the distinct bioanalytical needs of complex modalities.
Collapse
Affiliation(s)
- Liang Zhu
- PK Sciences, Novartis Institute for Biomedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA.
| | - Jim Glick
- PK Sciences, Novartis Institute for Biomedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA
| | - Jimmy Flarakos
- PK Sciences, Novartis Institute for Biomedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
17
|
Bottom-up sample preparation for the LC-MS/MS quantification of anti-cancer monoclonal antibodies in bio matrices. Bioanalysis 2020; 12:1405-1425. [PMID: 32975434 DOI: 10.4155/bio-2020-0204] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are rapidly taking over the treatment of many malignancies, and an astonishing number of mAbs is in development. This causes a high demand for quantification of mAbs in biomatrices both for measuring therapeutic mAb concentrations and to support pharmacokinetics and pharmacodynamics studies. Conventionally, ligand-binding assays are used for these purposes, but LC-MS is gaining popularity. Although intact (top-down) and subunit (middle-down) mAb quantification is reported, signature peptide (bottom-up) quantification is currently most advantageous. This review provides an overview of the reported bottom-up mAb quantification methods in biomatrices as well as general recommendations regarding signature peptide and internal standard selection, reagent use and optimization of digestion in bottom-up quantification methods.
Collapse
|
18
|
Zhu X, Huo S, Xue C, An B, Qu J. Current LC-MS-based strategies for characterization and quantification of antibody-drug conjugates. J Pharm Anal 2020; 10:209-220. [PMID: 32612867 PMCID: PMC7322744 DOI: 10.1016/j.jpha.2020.05.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 01/28/2023] Open
Abstract
The past few years have witnessed enormous progresses in the development of antibody-drug conjugates (ADCs). Consequently, comprehensive analysis of ADCs in biological systems is critical in supporting discovery, development and evaluation of these agents. Liquid chromatography-mass spectrometry (LC-MS) has emerged as a promising and versatile tool for ADC analysis across a wide range of scenarios, owing to its multiplexing ability, rapid method development, as well as the capability of analyzing a variety of targets ranging from small-molecule payloads to the intact protein with a high, molecular resolution. However, despite this tremendous potential, challenges persist due to the high complexity in both the ADC molecules and the related biological systems. This review summarizes the up-to-date LC-MS-based strategies in ADC analysis and discusses the challenges and opportunities in this rapidly-evolving field.
Collapse
Affiliation(s)
- Xiaoyu Zhu
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
| | - Shihan Huo
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
| | - Chao Xue
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
- Department of Chemical and Biological Engineering, School of Engineering and Applied Science, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Bo An
- Exploratory Biomarker, In-vitro/In-vivo Translation, R&D Research, GlaxoSmithKline Pharmaceuticals, 1250 South Collegeville Rd, Collegeville, PA, 19426, USA
| | - Jun Qu
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
| |
Collapse
|
19
|
IQ consortium perspective: complementary LBA and LC–MS in protein therapeutics bioanalysis and biotransformation assessment. Bioanalysis 2020; 12:257-270. [DOI: 10.4155/bio-2019-0279] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Increasingly diverse large molecule modalities have driven the need for complex bioanalysis and biotransformation assessment involving both traditional ligand-binding assays (LBA) and more recent hybrid immunoaffinity LC–MS platforms. Given the scientific expertise in LBA and LC–MS typically resides in different functions within the industry, this has presented operational challenges for an integrated approach for bioanalysis and biotransformation assessment. Encouragingly, over time, the industry has recognized the complementary value of the two platforms. This has not been an easy transition as organizational structures vary widely within the industry. However, there are tremendous benefits in adopting fully integrated strategies for biopharma. This IQ consortium paper presents current perspectives across the biopharma industry. It highlights the technical and operational challenges in current large molecule bioanalysis, the value of collaborations across LBA and LC–MS, and scientific expertise for fully integrated strategies for bioanalysis and biotransformation.
Collapse
|
20
|
Bioanalytical methods for therapeutic monoclonal antibodies and antibody–drug conjugates: A review of recent advances and future perspectives. J Pharm Biomed Anal 2020; 179:112991. [DOI: 10.1016/j.jpba.2019.112991] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/01/2019] [Accepted: 11/13/2019] [Indexed: 11/23/2022]
|
21
|
Schadt S, Hauri S, Lopes F, Edelmann MR, Staack RF, Villaseñor R, Kettenberger H, Roth AB, Schuler F, Richter WF, Funk C. Are Biotransformation Studies of Therapeutic Proteins Needed? Scientific Considerations and Technical Challenges. Drug Metab Dispos 2019; 47:1443-1456. [PMID: 31748266 DOI: 10.1124/dmd.119.088997] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/09/2019] [Indexed: 02/13/2025] Open
Abstract
For therapeutic proteins, the currently established standard development path generally does not foresee biotransformation studies by default because it is well known that the clearance of therapeutic proteins proceeds via degradation to small peptides and individual amino acids. In contrast to small molecules, there is no general need to identify enzymes involved in biotransformation because this information is not relevant for drug-drug interaction assessment and for understanding the clearance of a therapeutic protein. Nevertheless, there are good reasons to embark on biotransformation studies, especially for complex therapeutic proteins. Typical triggers are unexpected rapid clearance, species differences in clearance not following the typical allometric relationship, a mismatch in the pharmacokinetics/pharmacodynamics (PK/PD) relationship, and the need to understand observed differences between the results of multiple bioanalytical methods (e.g., total vs. target-binding competent antibody concentrations). Early on during compound optimization, knowledge on protein biotransformation may help to design more stable drug candidates with favorable in vivo PK properties. Understanding the biotransformation of a therapeutic protein may also support designing and understanding the bioanalytical assay and ultimately the PK/PD assessment. Especially in cases where biotransformation products are pharmacologically active, quantification and assessment of their contribution to the overall pharmacological effect can be important for establishing a PK/PD relationship and extrapolation to humans. With the increasing number of complex therapeutic protein formats, the need for understanding the biotransformation of therapeutic proteins becomes more urgent. This article provides an overview on biotransformation processes, proteases involved, strategic considerations, regulatory guidelines, literature examples for in vitro and in vivo biotransformation, and technical approaches to study protein biotransformation. SIGNIFICANCE STATEMENT: Understanding the biotransformation of complex therapeutic proteins can be crucial for establishing a pharmacokinetic/pharmacodynamic relationship. This article will highlight scientific, strategic, regulatory, and technological features of protein biotransformation.
Collapse
Affiliation(s)
- Simone Schadt
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Simon Hauri
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Filipe Lopes
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Martin R Edelmann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Roland F Staack
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Roberto Villaseñor
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Hubert Kettenberger
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Adrian B Roth
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Franz Schuler
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Wolfgang F Richter
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| | - Christoph Funk
- Roche Pharma Research and Early Development, Pharmaceutical Sciences (S.S., S.H., F.L., R.V., A.B.R., F.S., W.F.R., C.F.) and Roche Pharma Research and Early Development, Therapeutic Modalities (M.R.E.), Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland; and Roche Pharma Research and Early Development, Pharmaceutical Sciences (R.F.S.) and Roche Pharma Research and Early Development, Therapeutic Modalities (H.K.), Roche Innovation Center Munich, Roche Diagnostics, Penzberg, Germany
| |
Collapse
|
22
|
SOTOMATSU S, YAMADA T, MIZUNO H, HAYASHI H, TOYO’OKA T, TODOROKI K. High-Temperature Reversed-Phase LC Separation of Heavy and Light Chain Fragments of Therapeutic Monoclonal Antibodies and Antibody-Drug Conjugate Produced by Chemical Reduction. CHROMATOGRAPHY 2019. [DOI: 10.15583/jpchrom.2019.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Sae SOTOMATSU
- School of Pharmaceutical Sciences, University of Shizuoka
| | | | - Hajime MIZUNO
- School of Pharmaceutical Sciences, University of Shizuoka
| | | | | | | |
Collapse
|
23
|
Kang L, Weng N, Jian W. LC–MS bioanalysis of intact proteins and peptides. Biomed Chromatogr 2019; 34:e4633. [DOI: 10.1002/bmc.4633] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/19/2019] [Accepted: 06/25/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Lijuan Kang
- Drug Metabolism and Pharmacokinetics (DMPK)Janssen Pharmaceutical Companies of Johnson and Johnson Spring House PA
| | - Naidong Weng
- Drug Metabolism and Pharmacokinetics (DMPK)Janssen Pharmaceutical Companies of Johnson and Johnson Spring House PA
| | - Wenying Jian
- Drug Metabolism and Pharmacokinetics (DMPK)Janssen Pharmaceutical Companies of Johnson and Johnson Spring House PA
| |
Collapse
|
24
|
Ren C, Bobst CE, Kaltashov IA. Exploiting His-Tags for Absolute Quantitation of Exogenous Recombinant Proteins in Biological Matrices: Ruthenium as a Protein Tracer. Anal Chem 2019; 91:7189-7198. [PMID: 31083917 DOI: 10.1021/acs.analchem.9b00504] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Metal labeling and ICP MS detection offer an alternative to commonly accepted techniques that are currently used to quantitate exogenous proteins in vivo, but modifying the protein surface with metal-containing groups inevitably changes its biophysical properties and is likely to affect trafficking and biodistribution. The approach explored in this work takes advantage of the presence of hexa-histidine tags in many recombinant proteins, which have high affinity toward a range of metals. While many divalent metals bind to poly histidine sequences reversibly, oxidation of imidazole-bound CoII or RuII is known to result in a dramatic increase of the binding strength. In order to evaluate the feasibility of using imidazole-bound metal oxidation as a means of attaching permanent tags to polyhistidine segments, a synthetic peptide YPDFEDYWMKHHHHHH was used as a model. RuII can be oxidized under ambient (aerobic) conditions, allowing any oxidation damage to the peptide beyond the metal-binding site to be avoided. The resulting peptide-RuIII complex is very stable, with the single hexa-histidine segment capable of accommodating up to three metal ions. Localization of RuIII within the hexa-histidine segment of the peptide was confirmed by tandem mass spectrometry. The RuIII/peptide binding appears to be irreversible, with both low- and high-molecular weight biologically relevant scavengers failing to strip the metal from the peptide. Application of this protocol to labeling a recombinant form of an 80 kDa protein transferrin allowed RuIII to be selectively placed within the His-tag segment. The metal label remained stable in the presence of ubiquitous scavengers and did not interfere with the receptor binding, while allowing the protein to be readily detected in serum at sub-nM concentrations. The results of this work suggest that ruthenium lends itself as an ideal metal tag for selective labeling of His-tag containing recombinant proteins to enable their sensitive detection and quantitation with ICP MS.
Collapse
Affiliation(s)
- Chengfeng Ren
- Department of Chemistry , University of Massachusetts-Amherst , Amherst , Massachusetts 01003 , United States
| | - Cedric E Bobst
- Department of Chemistry , University of Massachusetts-Amherst , Amherst , Massachusetts 01003 , United States
| | - Igor A Kaltashov
- Department of Chemistry , University of Massachusetts-Amherst , Amherst , Massachusetts 01003 , United States
| |
Collapse
|
25
|
Protein quantification by LC–MS: a decade of progress through the pages of Bioanalysis. Bioanalysis 2019; 11:629-644. [DOI: 10.4155/bio-2019-0032] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Over the past 10 years, there has been a remarkable increase in the use of LC–MS for the quantitative determination of proteins, and this technique can now be considered an established bioanalytical platform for the quantification of macromolecular drugs and biomarkers, next to the traditional ligand-binding assays. Many researchers have contributed to the field and helped improve both the technical possibilities of LC–MS-based workflows and our understanding of the meaning of the results that are obtained. As a tribute to Bioanalysis, which has published many important contributions, this report gives a high-level overview of the most important trends in the field of protein LC–MS, as published in this journal since its inauguration a decade ago. It describes the major technical developments with regard to sample handling, separation and MS detection of both digested and intact protein analysis. In addition, the relevance of the complex structure and in vivo behavior of proteins is discussed and the effect of protein–protein interactions, biotransformation and the occurrence of isoforms on the analytical result is addressed.
Collapse
|
26
|
Bults P, Spanov B, Olaleye O, van de Merbel NC, Bischoff R. Intact protein bioanalysis by liquid chromatography – High-resolution mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1110-1111:155-167. [DOI: 10.1016/j.jchromb.2019.01.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/20/2019] [Accepted: 01/31/2019] [Indexed: 02/07/2023]
|
27
|
Beck A, D’Atri V, Ehkirch A, Fekete S, Hernandez-Alba O, Gahoual R, Leize-Wagner E, François Y, Guillarme D, Cianférani S. Cutting-edge multi-level analytical and structural characterization of antibody-drug conjugates: present and future. Expert Rev Proteomics 2019; 16:337-362. [DOI: 10.1080/14789450.2019.1578215] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Alain Beck
- Biologics CMC and Developability, IRPF - Centre d’Immunologie Pierre-Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Valentina D’Atri
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Anthony Ehkirch
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg, France
| | - Szabolcs Fekete
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg, France
| | - Rabah Gahoual
- Unité de Technologies Biologiques et Chimiques pour la Santé (UTCBS), Paris 5-CNRS UMR8258 Inserm U1022, Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Emmanuel Leize-Wagner
- Laboratoire de Spectrométrie de Masse des Interactions et des Systèmes (LSMIS), UMR 7140, Université de Strasbourg, CNRS, Strasbourg, France
| | - Yannis François
- Laboratoire de Spectrométrie de Masse des Interactions et des Systèmes (LSMIS), UMR 7140, Université de Strasbourg, CNRS, Strasbourg, France
| | - Davy Guillarme
- Biologics CMC and Developability, IRPF - Centre d’Immunologie Pierre-Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg, France
| |
Collapse
|
28
|
ADME Considerations and Bioanalytical Strategies for Pharmacokinetic Assessments of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:antib7040041. [PMID: 31544891 PMCID: PMC6698957 DOI: 10.3390/antib7040041] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/19/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a unique class of biotherapeutics of inherent heterogeneity and correspondingly complex absorption, distribution, metabolism, and excretion (ADME) properties. Herein, we consider the contribution of various components of ADCs such as various classes of warheads, linkers, and conjugation strategies on ADME of ADCs. Understanding the metabolism and disposition of ADCs and interpreting exposure-efficacy and exposure-safety relationships of ADCs in the context of their various catabolites is critical for design and subsequent development of a clinically successful ADCs. Sophisticated bioanalytical assays are required for the assessments of intact ADC, total antibody, released warhead and relevant metabolites. Both ligand-binding assays (LBA) and hybrid LBA-liquid chromatography coupled with tandem mass spectrometry (LBA-LC-MS/MS) methods have been employed to assess pharmacokinetics (PK) of ADCs. Future advances in bioanalytical techniques will need to address the rising complexity of this biotherapeutic modality as more innovative conjugation strategies, antibody scaffolds and novel classes of warheads are employed for the next generation of ADCs. This review reflects our considerations on ADME of ADCs and provides a perspective on the current bioanalytical strategies for pharmacokinetic assessments of ADCs.
Collapse
|
29
|
Review of approaches and examples for monitoring biotransformation in protein and peptide therapeutics by MS. Bioanalysis 2018; 10:1877-1890. [PMID: 30325207 DOI: 10.4155/bio-2018-0113] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Biotherapeutic drugs have emerged in quantity in pharmaceutical pipelines, and increasingly diverse biomolecules are progressed through preclinical and clinical development. As purification, separation, mass spectrometer detection and data processing capabilities improve, there is opportunity to monitor drug concentration by traditional ligand-binding assay or MS measurement and to monitor metabolism, catabolism or other biomolecular mass variants present in circulation. This review highlights approaches and examples of monitoring biotransformation of biotherapeutics by MS as these techniques are poised to add value to drug development in years to come. The increased use of such approaches, and the successful quantitation of biotherapeutic structural modifications, will provide insightful data for the benefit of both researchers and patients.
Collapse
|