1
|
Ieki K, Fukuda S, Miyawaki S, Hirowatari K. Regulated bioanalysis of antibody-drug conjugates using LC-MS. Bioanalysis 2025; 17:549-560. [PMID: 40205765 DOI: 10.1080/17576180.2025.2490468] [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: 11/28/2024] [Accepted: 04/03/2025] [Indexed: 04/11/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are emerging as powerful tools in cancer therapy. Evaluating their drug disposition requires the development and validation of analytical methods to obtain accurate quantitative results, which depend on understanding the ADC structural properties and selecting appropriate analytical platforms. Liquid chromatography-mass spectrometry (LC-MS) is a key technology for ADC bioanalysis, enabling the quantification of payloads, linkers, total antibodies, ADCs, and drug-to-antibody ratio (DAR). This review highlights the strategies and challenges in developing analytical methods for quantifying ADC components in biological samples using LC-MS with a focus on their constituent units. In addition, it addresses the validation requirements of these quantitative analytical methods during drug development.
Collapse
Affiliation(s)
- Katsunori Ieki
- Pharmacokinetics & Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan
| | - Suguru Fukuda
- Pharmacokinetics & Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan
| | - Shiori Miyawaki
- Pharmacokinetics & Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan
| | - Kazunari Hirowatari
- Pharmacokinetics & Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan
| |
Collapse
|
2
|
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
|
3
|
De Cristofaro M, Lenzi A, Ghimenti S, Biagini D, Bertazzo G, Vivaldi FM, Armenia S, Pugliese NR, Masi S, Di Francesco F, Lomonaco T. Decoding the Challenges: navigating Intact Peptide Mass Spectrometry-Based Analysis for Biological Applications. Crit Rev Anal Chem 2024:1-23. [PMID: 39556023 DOI: 10.1080/10408347.2024.2427140] [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: 11/19/2024]
Abstract
Quantitative analysis of peptides in biological fluids offers a high diagnostic and prognostic tool to reflect the pathophysiological condition of the patient. Recently, methods based on liquid chromatography coupled with mass spectrometry (LC-MS) for the quantitative determination of intact peptides have been replacing traditionally used ligand-binding assays, which suffer from cross-reactivity issues. The use of "top-down" analysis of peptides is rapidly increasing since it does not undergo incomplete or non-reproducible digestion like "bottom-up" approaches. However, the low abundance of peptides and their peculiar characteristics, as well as the complexity of biological fluids, make their quantification challenging. Herein, the analytical pitfalls that may be encountered during the development of an LC-MS method for the analysis of intact peptides in biological fluids are discussed. Challenges in the pre-analytical phase, stability after sampling and sample processing, significantly impact the accuracy of peptide quantification. Emerging techniques, such as microextractions, are becoming crucial for improved sample cleanup and enrichment of target analytes. A comparison between the roles of high-resolution and low-resolution mass spectrometry in the quantification of intact peptides, as well as the introduction of supercharging reagents to enhance ionization, will be discussed.
Collapse
Affiliation(s)
| | - Alessio Lenzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Silvia Ghimenti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Giulia Bertazzo
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | | | - Silvia Armenia
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Fabio Di Francesco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| |
Collapse
|
4
|
Sleumer B, van Faassen M, Vos MJ, den Besten G, Kema IP, van de Merbel NC. Simultaneous quantification of the 22-kDa isoforms of human growth hormone 1 and 2 in human plasma by multiplexed immunocapture and LC-MS/MS. Clin Chim Acta 2024; 554:117736. [PMID: 38142804 DOI: 10.1016/j.cca.2023.117736] [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: 11/08/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
An LC-MS/MS method is presented for the simultaneous quantification of two structurally closely related protein biomarker isoforms, the 22-kDa isoforms of human growth hormone 1 and human growth hormone 2, in human plasma. It is based on multiplexed immunocapture using two monoclonal antibodies immobilized on magnetic beads, tryptic digestion and quantification of two specific signature peptides plus an additional peptide for estimation of total growth hormone related concentrations. A full validation according to international guidelines was performed across the clinically relevant concentration ranges of 0.5 to 50 ng/mL for growth hormone 1, and 2 to 50 ng/mL for growth hormone 2 and demonstrated satisfactory method performance in terms of accuracy, precision, stability and absence of interference. The method's applicability for routine analysis and its ability to effectively distinguish between GH1 and GH2 was demonstrated by the analysis of plasma samples from pregnant individuals to study the changes in growth hormone levels during pregnancy.
Collapse
Affiliation(s)
- Bas Sleumer
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK Assen, the Netherlands; Department of Analytical Biochemistry University of Groningen, A. Deusinglaan 1, 9700 AV Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, P.O. Box 30.001, 9700 RB Groningen, the Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, P.O. Box 30.001, 9700 RB Groningen, the Netherlands
| | - Michel J Vos
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, P.O. Box 30.001, 9700 RB Groningen, the Netherlands
| | - Gijs den Besten
- Department of Clinical Chemistry, Isala, Dr. Van Heesweg 2, 8025 AB Zwolle, the Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, P.O. Box 30.001, 9700 RB Groningen, the Netherlands
| | - Nico C van de Merbel
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK Assen, the Netherlands; Department of Analytical Biochemistry University of Groningen, A. Deusinglaan 1, 9700 AV Groningen, the Netherlands.
| |
Collapse
|
5
|
Sleumer B, Kema IP, van de Merbel NC. Quantitative bioanalysis of proteins by digestion and LC-MS/MS: the use of multiple signature peptides. Bioanalysis 2023; 15:1203-1216. [PMID: 37724471 DOI: 10.4155/bio-2023-0129] [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: 09/20/2023] Open
Abstract
The use of multiple signature peptides for the quantification of proteins by digestion and LC-MS/MS is reviewed and evaluated here. A distinction is made based on the purpose of the use of multiple peptides: confirmation of the protein concentration, discrimination between different protein forms or species and in vivo biotransformation. Most reports that describe methods with at least two peptides use these for confirmation, but it is not always mentioned how the peptides are used and how possible differences in concentration between the peptides are handled. Differences in concentration are often reported in the case of monitoring different protein forms or in vivo biotransformation, and this offers insight into the biological fate of the protein.
Collapse
Affiliation(s)
- Bas Sleumer
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK, Assen, The Netherlands
- Department of Analytical Biochemistry, University of Groningen, A Deusinglaan 1, 9700 AV Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Nico C van de Merbel
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK, Assen, The Netherlands
- Department of Analytical Biochemistry, University of Groningen, A Deusinglaan 1, 9700 AV Groningen, The Netherlands
| |
Collapse
|
6
|
Krombholz S, Thomas A, Delahaut P, Bidlingmaier M, Schilbach K, Miller G, Thevis M. A combined top-down and bottom-up LC-HRMS/MS method for the quantification of human growth hormone in plasma and serum. Growth Horm IGF Res 2023; 72-73:101560. [PMID: 37995539 DOI: 10.1016/j.ghir.2023.101560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/03/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023]
Abstract
OBJECTIVE The precise and accurate quantification of human growth hormone (GH) in plasma/ serum is crucial for the diagnosis and treatment of diseases like GH deficiency or acromegaly. However, the ligand-binding assays (LBAs) currently used for routine testing show considerable methodological variability. Here, we present a complementary, combined top-down and bottom-up LC-MS-based method to quantify (intact) GH in plasma and serum, which concurrently provides a basis for a MS-based analysis of GH in doping controls. DESIGN Extraction of GH from plasma/ serum was accomplished by protein precipitation, followed by an immunocapture step using protein A-coupled magnetic beads and a polyclonal anti-GH antibody. The intact protein was subsequently analyzed top-down on a 2D-LC-HRMS/MS system. In addition, sample extracts were digested with trypsin and analyzed for signal peptides corresponding to 'total', 22 kDa and 20 kDa GH (bottom-up). Both assays were validated according to current guidelines and compared to the GH isoform differential immunoassay used in routine doping control analysis. GH concentrations in serum samples of healthy adults, patients with acromegaly, and in samples obtained after administration of recombinant GH were analyzed as proof-of-principle. RESULTS The intact monomeric 22 kDa isoform of GH was selectively quantified in a representative working range of 0.5 to 10 ng/ml by top-down LC-HRMS/MS. Subsequent bottom-up analysis provided additional data on 'total' and 20 kDa GH. Top-down and bottom-up assay results for the 22 kDa isoform correlated well with the corresponding immunoassay results (R2 > 0.95). For a possible application of the method in an anti-doping context, the ratio between 22 kDa and 'total' GH was evaluated, indicating differences between the various donor groups, but only with limited significance. CONCLUSION The top-down and bottom-up LC-HRMS/MS method developed here presents a valuable tool for the quantification of GH in plasma/ serum complementary to established LBAs used at present in clinical measurements. Albeit the examination of the GH isoform proportions by the LC-MS method does not yet allow for the assessment of GH abuse, the obtained findings provide an important basis to enable LC-MS-based GH analysis of doping control samples in the future.
Collapse
Affiliation(s)
- Sophia Krombholz
- Center for Preventive Doping Research - Institute of Biochemistry, German Sport University Cologne, Germany
| | - Andreas Thomas
- Center for Preventive Doping Research - Institute of Biochemistry, German Sport University Cologne, Germany
| | | | - Martin Bidlingmaier
- Endocrine Research Laboratories, Department of Medicine IV, LMU University Hospital, Munich, Germany
| | - Katharina Schilbach
- Endocrine Research Laboratories, Department of Medicine IV, LMU University Hospital, Munich, Germany
| | - Geoffrey Miller
- Sports Medicine Research and Testing Laboratory, Salt Lake City, UT, USA
| | - Mario Thevis
- Center for Preventive Doping Research - Institute of Biochemistry, German Sport University Cologne, Germany; European Monitoring Center for Emerging Doping Agents (EuMoCEDA), Cologne, Bonn, Germany.
| |
Collapse
|
7
|
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
|
8
|
Kim H, Cheon DH, Yang WS, Baek JH. Simultaneous Quantification of Apolipoprotein C-III O-Glycoforms by Protein-MRM. J Proteome Res 2023; 22:91-100. [PMID: 36412001 DOI: 10.1021/acs.jproteome.2c00490] [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: 11/23/2022]
Abstract
Apolipoprotein C-III (APOC-III) regulates triglyceride levels, associated with a risk of cardiovascular disease. One gene generates several proteoforms, each with a different molecular mass and a unique function. Unlike peptide multiple reaction monitoring (MRM), protein-MRM without digestion is required to analyze clinically relevant individual proteoforms. We developed a protein-MRM method without digestion to individually quantify APOC-III proteoforms in human serum. We optimized the protein-MRM method following 60% acetonitrile extraction with C18 filtration. Bovine serum and myoglobin served as supporting cushions and the internal standard during sample preparation, respectively. Furthermore, we evaluated the LOD, lower limit of quantification, linearity, accuracy, and precision. Good correlation compared with turbidimetric immunoassay (TIA) and peptide-MRM was observed using 30 clinical sera. Individual APOC-III O-glycoforms were identified by top-down proteomics and simultaneously quantified using the protein-MRM method. The sum abundance of APOC-III proteoforms was significantly correlated with TIA and peptide-MRM. Our protein-MRM method provides an affordable and rapid quantification of potential disease-specific proteoforms. Precise quantification of each proteoform allows investigators to identify novel biological roles potentially related to cardiovascular disease or novel biomarkers. We expect our protein-oriented method to be more clinically useful than antibody-based immunoassays and peptide-oriented MRM analysis, especially for quantification of a biomarker proteoform with certain post-translational modifications.
Collapse
Affiliation(s)
- Hyojin Kim
- R&D Center for Clinical Mass Spectrometry, Seegene Medical Foundation, Seongdong-gu, Seoul 04805, Korea
| | - Dong Huey Cheon
- R&D Center for Clinical Mass Spectrometry, Seegene Medical Foundation, Seongdong-gu, Seoul 04805, Korea
| | - Won Suk Yang
- R&D Center for Clinical Mass Spectrometry, Seegene Medical Foundation, Seongdong-gu, Seoul 04805, Korea
| | - Je-Hyun Baek
- R&D Center for Clinical Mass Spectrometry, Seegene Medical Foundation, Seongdong-gu, Seoul 04805, Korea
| |
Collapse
|
9
|
Sun L, Xu Y, Dube N, Anderson M, Breidinger S, Vaddady P, Thornton B, Morrow L, Matthews RP, Stoch SA, Woolf EJ. Incorporating protein precipitation to resolve hybrid IP-LC-MS assay interference for ultrasensitive quantification of intact therapeutic insulin dimer in human plasma. J Pharm Biomed Anal 2022; 212:114639. [DOI: 10.1016/j.jpba.2022.114639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/15/2021] [Accepted: 01/29/2022] [Indexed: 10/19/2022]
|
10
|
Amrani ME, Gerencser L, Huitema ADR, Hack CE, van Luin M, van der Elst KCM. A generic sample preparation method for the multiplex analysis of seven therapeutic monoclonal antibodies in human plasma or serum with liquid chromatography-tandem mass spectrometry. J Chromatogr A 2021; 1655:462489. [PMID: 34509691 DOI: 10.1016/j.chroma.2021.462489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/03/2021] [Accepted: 08/18/2021] [Indexed: 11/26/2022]
Abstract
Due to the increasing number of therapeutic monoclonal antibodies (mAbs) used in the clinic, there is an increasing need for robust analytical methods to quantify total mAb concentrations in human plasma for clinical studies and therapeutic drug monitoring. We developed an easy, rapid, and robust sample preparation method for liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. The method was validated for infliximab (IFX), rituximab (RTX), cetuximab (CTX), dupilumab (DPL), dinutuximab (DNX), vedolizumab (VDZ), and emicizumab (EMZ). Saturated ammonium sulfate (AS) was used to precipitate immunoglobulins in human plasma. After centrifugation, supernatant containing albumin was decanted, and the precipitated immunoglobulin fraction was re-dissolved in buffer containing 6M guanidine. This fraction was then completely denatured, reduced, alkylated, and trypsin digested. Finally, signature peptides from the seven mAbs were simultaneously quantified on LC-MS/MS together with their internal standards stable isotopically labeled peptide counterparts. The linear dynamic ranges (1 - 512 mg/L) of IFX, CTX, RTX, and EMZ showed excellent (R2 > 0.999) linearity and those of DPL, DNX, and VDZ showed good (R2 > 0.995) linearity. The method was validated in accordance with the EMA guidelines. EDTA plasma, sodium citrate plasma, heparin plasma, and serum yielded similar results. Prepared samples were stable at room temperature (20°C) and at 5°C for 3 days, and showed no decline in concentration for all tested mAbs. This described method, which has the advantage of an easy, rapid, and robust pre-analytical sample preparation, can be used as a template to quantify other mAbs in human plasma or serum.
Collapse
Affiliation(s)
- Mohsin El Amrani
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands.
| | - Laszlo Gerencser
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Alwin D R Huitema
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands; Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - C Erik Hack
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Matthijs van Luin
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Kim C M van der Elst
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands
| |
Collapse
|
11
|
Thakur A, Tan Z, Kameyama T, El-Khateeb E, Nagpal S, Malone S, Jamwal R, Nwabufo CK. Bioanalytical strategies in drug discovery and development. Drug Metab Rev 2021; 53:434-458. [PMID: 34310243 DOI: 10.1080/03602532.2021.1959606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A reliable, rapid, and effective bioanalytical method is essential for the determination of the pharmacokinetic, pharmacodynamic, and toxicokinetic parameters that inform the safety and efficacy profile of investigational drugs. The overall goal of bioanalytical method development is to elucidate the procedure and operating conditions under which a method can sufficiently extract, qualify, and/or quantify the analyte(s) of interest and/or their metabolites for the intended purpose. Given the difference in the physicochemical properties of small and large molecule drugs, different strategies need to be adopted for the development of an effective and efficient bioanalytical method. Herein, we provide an overview of different sample preparation strategies, analytical platforms, as well as procedures for achieving high throughput for bioanalysis of small and large molecule drugs.
Collapse
Affiliation(s)
- Aarzoo Thakur
- Innovations in Food and Chemical Safety, Agency for Science, Technology, and Research, Singapore, Singapore.,Skin Research Institute of Singapore, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Zhiyuan Tan
- Department of Early Clinical Development, dMed-Clinipace, Shanghai, China
| | - Tsubasa Kameyama
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Eman El-Khateeb
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK.,Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Shakti Nagpal
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | | | - Rohitash Jamwal
- College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | | |
Collapse
|
12
|
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
|
13
|
Lai X, Tang J, ElSayed MEH. Recent advances in proteolytic stability for peptide, protein, and antibody drug discovery. Expert Opin Drug Discov 2021; 16:1467-1482. [PMID: 34187273 DOI: 10.1080/17460441.2021.1942837] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: To discover and develop a peptide, protein, or antibody into a drug requires overcoming multiple challenges to obtain desired properties. Proteolytic stability is one of the challenges and deserves a focused investigation.Areas covered: This review concentrates on improving proteolytic stability by engineering the amino acids around the cleavage sites of a liable peptide, protein, or antibody. Peptidases are discussed on three levels including all peptidases in databases, mixtures based on organ and tissue types, and individual peptidases. The technique to identify cleavage sites is spotlighted on mass spectrometry-based approaches such as MALDI-TOF and LC-MS. For sequence engineering, the replacements that have been commonly applied with a higher chance of success are highlighted at the beginning, while the rarely used and more complicated replacements are discussed later. Although a one-size-fits-all approach does not exist to apply to different projects, this review provides a 3-step strategy for effectively and efficiently conducting the proteolytic stability experiments to achieve the eventual goal of improving the stability by engineering the molecule itself.Expert opinion: Improving the proteolytic stability is a spiraling up process sequenced by testing and engineering. There are many ways to engineer amino acids, but the choice must consider the cost and properties affected by the changes of the amino acids.
Collapse
Affiliation(s)
- Xianyin Lai
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jason Tang
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mohamed E H ElSayed
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
14
|
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
|
15
|
Moran AB, Domínguez-Vega E, Nouta J, Pongracz T, de Reijke TM, Wuhrer M, Lageveen-Kammeijer GSM. Profiling the proteoforms of urinary prostate-specific antigen by capillary electrophoresis - mass spectrometry. J Proteomics 2021; 238:104148. [PMID: 33618028 DOI: 10.1016/j.jprot.2021.104148] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/26/2021] [Accepted: 01/31/2021] [Indexed: 01/07/2023]
Abstract
Early detection of prostate cancer may lead to the overdiagnosis and overtreatment of patients as well as missing significant cancers. The current diagnostic approach uses elevated serum concentrations of prostate-specific antigen (PSA) as an indicator of risk. However, this test has been widely criticized as it shows poor specificity and sensitivity. In order to improve early detection and diagnosis, several studies have investigated whether different PSA proteoforms are correlated to prostate cancer. Until now, studies and methodologies for the comprehensive characterization of PSA proteoforms from biofluids are scarce. For this purpose, we developed an intact protein assay to analyze PSA by capillary electrophoresis-electrospray ionization-mass spectrometry after affinity purification from patients' urine. Here, we determined six proteolytic cleavage variants. In regard to glycosylation, tri-, di-, mono- and non-sialylated complex-type N-glycans were found on non-cleaved PSA, as well as the non-glycosylated variant. The performance of the intact protein assay was assessed using a pooled sample, obtaining an inter-day variability of 15%. Furthermore, urinary patient samples were analyzed by intact protein analysis and a bottom-up approach (glycopeptide analysis). This combined approach revealed complimentary information on both levels, demonstrating the benefit of using two orthogonal techniques to provide a thorough profile of urinary PSA. SIGNIFICANCE: The detection of clinically relevant prostate cancer requires a more specific and sensitive biomarker and, in this case, several PSA proteoforms may be able to aid or improve the current PSA test. However, a comprehensive analysis of the intact PSA proteoform profile is still lacking. This study investigated the PSA proteoforms present in urine and, in particular, determined the relative contribution of cleaved PSA and non-cleaved PSA forms to the total glycosylation profile. Importantly, intact protein analysis did not require further sample treatment before being measured by CE-ESI-MS. Furthermore, its glycosylation was also assessed in a bottom-up approach to provide complementary information. Overall, these results represent an important basis for future characterization and biomarker studies.
Collapse
Affiliation(s)
- Alan B Moran
- Leiden University Medical Center, Center for Proteomics and Metabolomics, 2300 RC Leiden, the Netherlands
| | - Elena Domínguez-Vega
- Leiden University Medical Center, Center for Proteomics and Metabolomics, 2300 RC Leiden, the Netherlands
| | - Jan Nouta
- Leiden University Medical Center, Center for Proteomics and Metabolomics, 2300 RC Leiden, the Netherlands
| | - Tamas Pongracz
- Leiden University Medical Center, Center for Proteomics and Metabolomics, 2300 RC Leiden, the Netherlands
| | - Theo M de Reijke
- Amsterdam UMC, location Academic Medical Center, Department of Urology, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Manfred Wuhrer
- Leiden University Medical Center, Center for Proteomics and Metabolomics, 2300 RC Leiden, the Netherlands
| | | |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Bults P, Sonesson A, Knutsson M, Bischoff R, van de Merbel NC. Intact protein quantification in biological samples by liquid chromatography - high-resolution mass spectrometry: somatropin in rat plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1144:122079. [PMID: 32247186 DOI: 10.1016/j.jchromb.2020.122079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/20/2020] [Accepted: 03/22/2020] [Indexed: 01/28/2023]
Abstract
The quantitative determination of intact proteins in biological samples by LC with high-resolution MS detection can be a useful alternative to ligand-binding assays or LC-MS-based quantification of a surrogate peptide after protein digestion. The 22-kDa biopharmaceutical protein somatropin (recombinant human growth hormone) was quantified down to 10 ng/mL (0.45 nM) in 75 μL of rat plasma by the combination of an immunocapture step using an anti-somatropin antibody and LC-MS on a quadrupole-time of flight instrument. Accuracy and precision of the method as well as its selectivity and sensitivity did not depend on the width of the mass extraction window nor on whether only one or a summation of multiple charge states of the protein analyte were used as the detection response. Quantification based on deconvoluted mass spectra showed equally acceptable method performance but with a less favorable lower limit of quantification of 30 ng/mL. Concentrations in plasma after dosing of somatropin to rats correlated well for the deconvolution approach and the quantification based on the summation of the response of the four most intense charge states (14+ to 17+) of somatropin.
Collapse
Affiliation(s)
- Peter Bults
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands; Bioanalytical Laboratory, PRA Health Sciences, Amerikaweg 18, 9407 TK Assen, the Netherlands
| | - Anders Sonesson
- Ferring Pharmaceuticals, Kay Fiskers Plads 11, DK-2300 Copenhagen, Denmark
| | - Magnus Knutsson
- Ferring Pharmaceuticals, Kay Fiskers Plads 11, DK-2300 Copenhagen, Denmark
| | - Rainer Bischoff
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Nico C van de Merbel
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands; Bioanalytical Laboratory, PRA Health Sciences, Amerikaweg 18, 9407 TK Assen, the Netherlands.
| |
Collapse
|
19
|
Maráková K, Rai AJ, Schug KA. Effect of difluoroacetic acid and biological matrices on the development of a liquid chromatography-triple quadrupole mass spectrometry method for determination of intact growth factor proteins. J Sep Sci 2020; 43:1663-1677. [PMID: 32052929 DOI: 10.1002/jssc.201901254] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 12/17/2022]
Abstract
In biological systems, variable protein expression is a crucial marker for numerous diseases, including cancer. The vast majority of liquid chromatography-triple quadrupole mass spectrometry-based quantitative protein assays use bottom-up methodologies, where proteins are subjected to proteolytic cleavage prior to analysis. Here, the effect of difluoroacetic acid and biological matrices on the developement of a multiple reaction monitoring based top-down reversed-phase liquid chromatography-triple quadrupole mass spectrometry method for analysis of cancer-related intact proteins was evaluated. Seven growth factors (5.5-26.5 kDa; isoelectric points: 4.6-9.9) were analyzed on a wide-pore C4 column. The optimized method was performed at 30°C, using a 0.2 mL/min flow rate, a 10 %B/min gradient slope, and 0.05% v/v difluoroacetic acid as a mobile phase modifier. The increase of mass spectrometry sensitivity due to the difluoroacetic acid (estimated limits of detection in biological matrices 1-500 ng/mL) significantly varied for proteins with lower and higher charge state distributions. Matrix effects, as well as the specificity of the method were assessed for variable biological samples and pretreatment methods. This work demonstrates method development to improve the ability to target intact proteins directly by more affordable triple quadrupole mass spectrometry instrumentation, which could be beneficial in many application fields.
Collapse
Affiliation(s)
- Katarína Maráková
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovakia
| | - Alex J Rai
- Department of Pathology and Cell Biology Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA
| | - Kevin A Schug
- Department of Chemistry & Biochemistry, The University of Texas at Arlington, Arlington, Texas, USA
| |
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
|
Neubert H, Shuford CM, Olah TV, Garofolo F, Schultz GA, Jones BR, Amaravadi L, Laterza OF, Xu K, Ackermann BL. Protein Biomarker Quantification by Immunoaffinity Liquid Chromatography–Tandem Mass Spectrometry: Current State and Future Vision. Clin Chem 2020; 66:282-301. [DOI: 10.1093/clinchem/hvz022] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022]
Abstract
Abstract
Immunoaffinity–mass spectrometry (IA-MS) is an emerging analytical genre with several advantages for profiling and determination of protein biomarkers. Because IA-MS combines affinity capture, analogous to ligand binding assays (LBAs), with mass spectrometry (MS) detection, this platform is often described using the term hybrid methods. The purpose of this report is to provide an overview of the principles of IA-MS and to demonstrate, through application, the unique power and potential of this technology. By combining target immunoaffinity enrichment with the use of stable isotope-labeled internal standards and MS detection, IA-MS achieves high sensitivity while providing unparalleled specificity for the quantification of protein biomarkers in fluids and tissues. In recent years, significant uptake of IA-MS has occurred in the pharmaceutical industry, particularly in the early stages of clinical development, enabling biomarker measurement previously considered unattainable. By comparison, IA-MS adoption by CLIA laboratories has occurred more slowly. Current barriers to IA-MS use and opportunities for expanded adoption are discussed. The path forward involves identifying applications for which IA-MS is the best option compared with LBA or MS technologies alone. IA-MS will continue to benefit from advances in reagent generation, more sensitive and higher throughput MS technologies, and continued growth in use by the broader analytical community. Collectively, the pursuit of these opportunities will secure expanded long-term use of IA-MS for clinical applications.
Collapse
|
22
|
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
|
23
|
Gajjala PR, Bruck H, Noels H, Heinze G, Ceccarelli F, Kribben A, Saez-Rodriguez J, Marx N, Zidek W, Jankowski J, Jankowski V. Novel plasma peptide markers involved in the pathology of CKD identified using mass spectrometric approach. J Mol Med (Berl) 2019; 97:1451-1463. [PMID: 31385015 PMCID: PMC6746684 DOI: 10.1007/s00109-019-01823-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/12/2019] [Accepted: 07/25/2019] [Indexed: 02/04/2023]
Abstract
Chronic kidney disease (CKD) may progress to end-stage renal disease (ESRD) at different pace. Early markers of disease progression could facilitate and improve patient management. However, conventional blood and urine chemistry have proven unable to predict the progression of disease at early stages. Therefore, we performed untargeted plasma peptidome analysis to select the peptides involved in progression, which are suitable for long prospective studies in future. The study consists of non-CKD (n = 66) and CKD (n = 106) patients with different stages. We performed plasma peptidomics on these subjects using chromatography and mass spectrometric approaches. Initially, we performed LC-ESI-MS and applied least absolute shrinkage and selection operator logistic regressions to select the peptides that are differentially expressed and we generated a peptidomic score for each subject. Later, we identified and sequenced the peptides with MALDI-MS/MS and also performed univariate and multivariate analyses with the clinical variables and peptidomic score to reveal their association with progression of renal disease. A logistic regression model selected 14 substances showing different concentrations according to renal function, of which seven substances were most likely occur in CKD patients. The peptidomic model had a global P value of < 0.01 with R2 of 0.466, and the area under the curve was 0.87 (95% CI, 0.8149-0.9186; P < 0.0001). The predicted score was significantly higher in CKD than in non-CKD patients (2.539 ± 0.2637 vs - 0.9382 ± 0.1691). The model was also able to predict stages of CKD: the Spearman correlation coefficient of the linear predictor with CKD stages was 0.83 with concordance indices of 0.899 (95% CI 0.863-0.927). In univariate analysis, the most consistent association of peptidomic score in CKD patients was with C-reactive protein, sodium level, and uric acid, which are unanticipated substances. Peptidomic analysis enabled to list some unanticipated substances that have not been extensively studied in the context of CKD but were associated with CKD progression, thus revealing interesting candidate markers or mediators of CKD of potential use in CKD progression management. KEY MESSAGES: • Conventional blood and urine chemistry have proven unable to predict the progression of disease at early stages of chronic kidney disease (CKD). • We performed untargeted plasma peptidome analysis to select the peptides involved in progression. • A logistic regression model selected 14 substances showing different concentrations according to renal function. • These peptides are unanticipated substances that have not been extensively studied in the context of CKD but were associated with CKD progression, thus revealing markers or mediators of CKD of potential use in CKD progression management.
Collapse
Affiliation(s)
- Prathibha R Gajjala
- Institute of Molecular Cardiovascular Research, University Hospital RWTH Aachen, Aachen, Germany
| | - Heike Bruck
- Department of Internal Medicine, Nephrology, Rheumatology, Diabetology and Endocrinology, Helios Hospital Krefeld, Krefeld, Germany
| | - Heidi Noels
- Institute of Molecular Cardiovascular Research, University Hospital RWTH Aachen, Aachen, Germany
| | - Georg Heinze
- Section for Clinical Biometrics, Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Francesco Ceccarelli
- Joint Research Centre for Computational Biomedicine, RWTH Aachen University, Aachen, Germany
| | - Andreas Kribben
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Julio Saez-Rodriguez
- Joint Research Centre for Computational Biomedicine, RWTH Aachen University, Aachen, Germany
- Institute for Computational Biomedicine, Faculty of Medicine Bioquant, Heidelberg University, Heidelberg, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I, University Hospital Aachen, Aachen, Germany
| | - Walter Zidek
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research, University Hospital RWTH Aachen, Aachen, Germany
- Experimental Vascular Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, Netherlands
| | - Vera Jankowski
- Institute of Molecular Cardiovascular Research, University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
24
|
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
|
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
|
Direct quantitation of therapeutic antibodies for pharmacokinetic studies using immuno-purification and intact mass analysis. Bioanalysis 2019; 11:203-213. [PMID: 30734579 DOI: 10.4155/bio-2018-0240] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aim: The quantitation of therapeutic antibodies by MS often utilizes a surrogate peptide approach. Recent enhancements in instrumentation and sample preparation have enabled quantitation by detection of the intact molecule using MS. Methods & Results: A comparison of three methods for quantitative analysis of therapeutic monoclonal antibodies including analysis after deglycosylation, after hinge digestion and at the fully intact antibody level is reported. The optimized methodology provided sensitivity down to 0.1 μg/ml and a lower limit of quantitation of 0.5 ug/ml from a 30 μl sample volume. Conclusion: Application of this approach to a pharmacokinetic study compared with a conventional surrogate peptide and a ligand-binding assays provided consistent data with direct detection of the dosed molecule.
Collapse
|
27
|
Pedersen ME, Østergaard J, Jensen H. Flow-Induced Dispersion Analysis (FIDA) for Protein Quantification and Characterization. Methods Mol Biol 2019; 1972:109-123. [PMID: 30847787 DOI: 10.1007/978-1-4939-9213-3_8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Flow-Induced Dispersion Analysis (FIDA) enables characterization and quantification of proteins under native conditions. FIDA is based on measuring the change in size of a ligand as it selectively interacts with the target protein. The unbound ligand has a relatively small apparent hydrodynamic radius (size), which increase in the presence of the analyte due to binding to the analyte. The Kd of the interaction may be obtained in a titration experiment and the measurement of the apparent ligand size in an unknown sample forms the basis for determining the analyte concentration. The apparent molecular size is measured by Taylor dispersion analysis (TDA) in fused silica capillary capillaries. FIDA is a "ligand-binding" assay and has therefore certain features in common with Enzyme-Linked Immunosorbent Assay (ELISA), Surface Plasmon Resonance (SPR), and Biolayer Interferometry (BLI) based techniques. However, FIDA probes a single in-solution binding event and thus makes assay development straightforward, and the absolute size measurement enables built-in assay quality control. Further, as FIDA does not involve surface chemistries, complications related to nonspecific adsorption of analyte and assay components are minimized enabling direct measurement in, e.g., plasma and serum.
Collapse
Affiliation(s)
| | - Jesper Østergaard
- FIDA-Tech Aps, C/O University of Copenhagen, Copenhagen, Denmark
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Jensen
- FIDA-Tech Aps, C/O University of Copenhagen, Copenhagen, Denmark.
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
28
|
Perspectives on potentiating immunocapture-LC-MS for the bioanalysis of biotherapeutics and biomarkers. Bioanalysis 2018; 10:1679-1690. [PMID: 30371100 DOI: 10.4155/bio-2018-0205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The integration of ligand-binding assay and LC-MS/MS (immunocapture-LC-MS) has unleashed the combined advantages of both powerful techniques for addressing the ever increasing bioanalytical challenges for biotherapeutics and biomarker assays. The highly specific, selective and sensitive characteristics of the immunocapture-LC-MS-based assays have enabled the determination of biotherapeutics and biomarkers in biomatrices with ease of method development, less requirements on key reagents as well as structural specificity for endogenous and engineered biomolecules. In addition, the versatile immunocapture-LC-MS technology has expanded into many challenging areas to enhance mechanistic studies of drug interactions with their targets. This paper intends to summarize our perspectives on enhancing the use of immunocapture-LC-MS in drug discovery and development for emerging new modalities.
Collapse
|
29
|
Quantitation of intact monoclonal antibody in biological samples: comparison of different data processing strategies. Bioanalysis 2018; 10:1055-1067. [DOI: 10.4155/bio-2018-0016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: Sample extraction using immuno-affinity capture coupled with LC–high-resolution mass spectrometer has recently emerged as a novel approach for the determination of concentrations of large molecules at intact level in biological matrix. Methodology: In the current work, different data processing strategies for intact protein bioanalysis, deconvoluted mass spectra or extracted ion chromatogram, were applied to quantitate monoclonal antibody in biological samples for comparison of assay performance. Conclusion: Both deconvolution and extracted ion chromatogram strategies showed similar selectivity, sensitivity, accuracy and precision. The monkey pharmacokinetics data obtained from both approaches agreed well with each other, and agreed with data obtained from surrogate peptide approach. The pros and cons, and optimal parameters of each approach were discussed.
Collapse
|
30
|
Top-down LC–MS quantitation of intact denatured and native monoclonal antibodies in biological samples. Bioanalysis 2018; 10:1039-1054. [DOI: 10.4155/bio-2017-0282] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim: The requirements for developing antibody biotherapeutics benefit from understanding the nature and relevant aspects of the entire molecule. The method presented herein employs on-line multidimensional LC–quadrupole time-of-flight (QTOF)-MS for the quantitative determination of an antibody isolated from biological samples while maintaining the intact native biologically active conformation of the antibody. Results: Following method optimization for a model antibody, an incurred biotherapeutic in cynomologus monkey was quantified in its intact top-down native conformation. A partial method validation demonstrated acceptable precision and accuracy although improved sensitivity requires further studies. Conclusion: An on-line multidimensional LC–MS approach presents a proof-of-principle example for quantifying an intact, native antibody isolated from an incurred biological sample via immunoaffinity techniques coupled with top-down QTOF LC–MS bioanalysis.
Collapse
|
31
|
LC–HRMS quantitation of intact antibody drug conjugate trastuzumab emtansine from rat plasma. Bioanalysis 2018; 10:851-862. [DOI: 10.4155/bio-2018-0003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Aim: Compared with small molecules, LC–MS quantitation of larger biotherapeutic proteins such as antibodies and antibody–drug conjugates at the intact level presents many challenges in both LC and MS due to their higher molecular weight, bigger size, structural complexity and heterogeneity. Results & conclusion: In this study, quantitation of an intact lysine-linked antibody–drug conjugate, trastuzumab emtansine is presented. Trastuzumab emtansine was extracted from rat plasma using bead-based immunoaffinity capture; after elution from the beads, it was directly analyzed on a LC–HRMS system. Quantitation using both extracted ion chromatogram and deconvoluted mass peaks was evaluated. A limit of quantitation was approximately 20 ng on column with a linear dynamic range from 5 to 100 μg/ml. In addition, the reproducibility and distribution of the drug-to-antibody ratio at different trastuzumab emtansine concentrations were discussed.
Collapse
|
32
|
Schmitz EM, Leijten NM, van Dongen JL, Broeren MA, Milroy LG, Brunsveld L, Scharnhorst V, van de Kerkhof D. Optimizing charge state distribution is a prerequisite for accurate protein biomarker quantification with LC-MS/MS, as illustrated by hepcidin measurement. ACTA ACUST UNITED AC 2018; 56:1490-1497. [DOI: 10.1515/cclm-2018-0013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/26/2018] [Indexed: 12/20/2022]
Abstract
Abstract
Background:
Targeted quantification of protein biomarkers with liquid chromatography-tandem mass spectrometry (LC-MS/MS) has great potential, but is still in its infancy. Therefore, we elucidated the influence of charge state distribution and matrix effects on accurate quantification, illustrated by the peptide hormone hepcidin.
Methods:
An LC-MS/MS assay for hepcidin, developed based on existing literature, was improved by using 5 mM ammonium formate buffer as mobile phase A and as an elution solution for solid phase extraction (SPE) to optimize the charge state distribution. After extensive analytical validation, focusing on interference and matrix effects, the clinical consequence of this method adjustment was studied by performing receiving operating characteristic (ROC)-curve analysis in patients with iron deficiency anemia (IDA, n=44), anemia of chronic disease (ACD, n=42) and non-anemic patients (n=93).
Results:
By using a buffered solution during sample preparation and chromatography, the most abundant charge state was shifted from 4+ to 3+ and the charge state distribution was strongly stabilized. The matrix effects which occurred in the 4+ state were therefore avoided, eliminating bias in the low concentration range of hepcidin. Consequently, sensitivity, specificity and positive predictive value (PPV) for detection of IDA patients with the optimized assay (96%, 97%, 91%, respectively) were much better than for the original assay (73%, 70%, 44%, respectively).
Conclusions:
Fundamental improvements in LC-MS/MS assays greatly impact the accuracy of protein quantification. This is urgently required for improved diagnostic accuracy and clinical value, as illustrated by the validation of our hepcidin assay.
Collapse
Affiliation(s)
- Ellen M.H. Schmitz
- Expert Center Clinical Chemistry Eindhoven , Eindhoven , The Netherlands
- Catharina Hospital Eindhoven , Clinical Laboratory , Eindhoven , The Netherlands
- Eindhoven University of Technology , Department of Biomedical Engineering , Laboratory of Chemical Biology and Institute for Complex Molecular Systems , Eindhoven , The Netherlands
- Máxima Medical Center Veldhoven , Clinical Laboratory , Veldhoven , The Netherlands
| | - Niels M. Leijten
- Expert Center Clinical Chemistry Eindhoven , Eindhoven , The Netherlands
- Catharina Hospital Eindhoven , Clinical Laboratory , Eindhoven , The Netherlands
- Eindhoven University of Technology , Department of Biomedical Engineering , Laboratory of Chemical Biology and Institute for Complex Molecular Systems , Eindhoven , The Netherlands
| | - Joost L.J. van Dongen
- Expert Center Clinical Chemistry Eindhoven , Eindhoven , The Netherlands
- Eindhoven University of Technology , Department of Biomedical Engineering , Laboratory of Chemical Biology and Institute for Complex Molecular Systems , Eindhoven , The Netherlands
| | - Maarten A.C. Broeren
- Expert Center Clinical Chemistry Eindhoven , Eindhoven , The Netherlands
- Máxima Medical Center Veldhoven , Clinical Laboratory , Veldhoven , The Netherlands
| | - Lech G. Milroy
- Eindhoven University of Technology , Department of Biomedical Engineering , Laboratory of Chemical Biology and Institute for Complex Molecular Systems , Eindhoven , The Netherlands
| | - Luc Brunsveld
- Expert Center Clinical Chemistry Eindhoven , Eindhoven , The Netherlands
- Eindhoven University of Technology , Department of Biomedical Engineering , Laboratory of Chemical Biology and Institute for Complex Molecular Systems , Eindhoven , The Netherlands
| | - Volkher Scharnhorst
- Expert Center Clinical Chemistry Eindhoven , Eindhoven , The Netherlands
- Catharina Hospital Eindhoven , Clinical Laboratory , Eindhoven , The Netherlands
- Eindhoven University of Technology , Department of Biomedical Engineering , Laboratory of Chemical Biology and Institute for Complex Molecular Systems , Eindhoven , The Netherlands
| | - Daan van de Kerkhof
- Expert Center Clinical Chemistry Eindhoven , Eindhoven , The Netherlands
- Catharina Hospital Eindhoven , Clinical Laboratory , Eindhoven , The Netherlands
- Algemeen Klinisch Laboratorium Catharina Ziekenhuis , Michelangelolaan 2 , 5623 EJ Eindhoven , The Netherlands
| |
Collapse
|
33
|
LC–MS Challenges in Characterizing and Quantifying Monoclonal Antibodies (mAb) and Antibody-Drug Conjugates (ADC) in Biological Samples. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s40495-017-0118-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
34
|
Vasicek LA, Spellman DS, Hsieh S, Seghezzi W, Zhang S, Santostefano M, Bateman KP. Quantitation of a Therapeutic Antibody in Serum Using Intact Sequential Affinity Capture, Trypsin Digestion, and LC-MS/MS. Anal Chem 2017; 90:866-871. [DOI: 10.1021/acs.analchem.7b03716] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Lisa A. Vasicek
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., West Point, Pennsylvania, United States
| | - Daniel S. Spellman
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., West Point, Pennsylvania, United States
| | - SuChun Hsieh
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Palo Alto, California, United States
| | - Wolfgang Seghezzi
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Palo Alto, California, United States
| | - Shuli Zhang
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Palo Alto, California, United States
| | - Michael Santostefano
- Safety
Assessment and Laboratory Animal Resources, Merck & Co., Inc., Boston, Massachusetts, United States
| | - Kevin P. Bateman
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., West Point, Pennsylvania, United States
| |
Collapse
|
35
|
Mesonzhnik NV, Postnikov PV, Appolonova SA, Krotov GI. Characterization and Detection of Erythropoietin Fc Fusion Proteins Using Liquid Chromatography–Mass Spectrometry. J Proteome Res 2017; 17:689-697. [DOI: 10.1021/acs.jproteome.7b00739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Natalia V. Mesonzhnik
- Institute
of Pharmacy and Translational Medicine, Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya Street, 119991 Moscow, Russia
| | - Pavel V. Postnikov
- Anti-Doping Center, Elizavetinskiy
per., 10/1, 105005 Moscow, Russian Federation
| | - Svetlana A. Appolonova
- Institute
of Pharmacy and Translational Medicine, Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya Street, 119991 Moscow, Russia
| | - Grigory I. Krotov
- NRC Institute of Immunology FMBA of Russia, 24 Kashirskoye Highway, Moscow 115478, Russia
| |
Collapse
|
36
|
Immunoaffinity capture coupled with capillary electrophoresis - mass spectrometry to study therapeutic protein stability in vivo. Anal Biochem 2017; 539:118-126. [DOI: 10.1016/j.ab.2017.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 01/09/2023]
|
37
|
The future of immunocapture-capillary electrophoresis-high resolution mass spectrometry. Bioanalysis 2017; 9:1627-1631. [PMID: 29095042 DOI: 10.4155/bio-2017-4977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
38
|
van den Broek I, Sobhani K, Van Eyk JE. Advances in quantifying apolipoproteins using LC-MS/MS technology: implications for the clinic. Expert Rev Proteomics 2017; 14:869-880. [PMID: 28870113 DOI: 10.1080/14789450.2017.1374859] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Apolipoproteins play a key role in pre-, pro-, and anti-atherosclerotic processes and have become important circulating biomarkers for the prediction of cardiovascular disease (CVD) risk. Whereas currently clinical immunoassays are not available for most apolipoproteins and lack the capacity for multiplexing, liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) allows simultaneous, highly-specific, and precise quantification of multiple apolipoproteins. Areas covered: We discuss LC-MS/MS methods for quantification of apolipoproteins reported in the literature and highlight key requirements for clinical use. Besides the advances in sample preparation and LC-MS/MS technologies, this overview also discusses advances in proteoform analysis and applications of dried blood/plasma collection. Expert commentary: Standardized quantification using LC-MS/MS technology has been demonstrated for apolipoprotein A-I and B. However, for implementation in clinical CVD risk assessment, LC-MS/MS must bring significant added clinical value in comparison to fast, standardized, and straightforward clinical (immuno)assays. Ongoing advances in accuracy and multiplexing capacity of LC-MS/MS, nonetheless, bear potential to enable standardized and interpretable personalized profiling of a patient's CVD risk by simultaneous quantification of multiple apolipoproteins and -variants. We, moreover, anticipate further personalization of CVD risk assessment by the potential of LC-MS/MS to enable simultaneous genotyping and remote monitoring using dried blood/plasma collection devices.
Collapse
Affiliation(s)
- Irene van den Broek
- a Cedars-Sinai Medical Center, The Advanced Clinical Biosystems Research Institute , The Heart Institute , Los Angeles , CA , USA
| | - Kimia Sobhani
- b Department of Pathology and Laboratory Medicine , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| | - Jennifer E Van Eyk
- a Cedars-Sinai Medical Center, The Advanced Clinical Biosystems Research Institute , The Heart Institute , Los Angeles , CA , USA
| |
Collapse
|
39
|
Kang L, Camacho RC, Li W, D’Aquino K, You S, Chuo V, Weng N, Jian W. Simultaneous Catabolite Identification and Quantitation of Large Therapeutic Protein at the Intact Level by Immunoaffinity Capture Liquid Chromatography–High-Resolution Mass Spectrometry. Anal Chem 2017; 89:6065-6075. [DOI: 10.1021/acs.analchem.7b00674] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Lijuan Kang
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Raul C. Camacho
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Wenyu Li
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Katharine D’Aquino
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Seohee You
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Vanessa Chuo
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Naidong Weng
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Wenying Jian
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| |
Collapse
|
40
|
Zhao Y, Liu G, Yuan X, Gan J, Peterson JE, Shen JX. Strategy for the Quantitation of a Protein Conjugate via Hybrid Immunocapture-Liquid Chromatography with Sequential HRMS and SRM-Based LC-MS/MS Analyses. Anal Chem 2017; 89:5144-5151. [DOI: 10.1021/acs.analchem.7b00926] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Yue Zhao
- Analytical and Bioanalytical
Operations, Research and Development, Bristol-Myers Squibb Co., Princeton, New Jersey 08543, United States
| | - Guowen Liu
- Analytical and Bioanalytical
Operations, Research and Development, Bristol-Myers Squibb Co., Princeton, New Jersey 08543, United States
| | - Xiling Yuan
- Analytical and Bioanalytical
Operations, Research and Development, Bristol-Myers Squibb Co., Princeton, New Jersey 08543, United States
| | - Jinping Gan
- Analytical and Bioanalytical
Operations, Research and Development, Bristol-Myers Squibb Co., Princeton, New Jersey 08543, United States
| | - Jon E. Peterson
- Analytical and Bioanalytical
Operations, Research and Development, Bristol-Myers Squibb Co., Princeton, New Jersey 08543, United States
| | - Jim X. Shen
- Analytical and Bioanalytical
Operations, Research and Development, Bristol-Myers Squibb Co., Princeton, New Jersey 08543, United States
| |
Collapse
|
41
|
Lanshoeft C, Cianférani S, Heudi O. Generic Hybrid Ligand Binding Assay Liquid Chromatography High-Resolution Mass Spectrometry-Based Workflow for Multiplexed Human Immunoglobulin G1 Quantification at the Intact Protein Level: Application to Preclinical Pharmacokinetic Studies. Anal Chem 2017; 89:2628-2635. [DOI: 10.1021/acs.analchem.6b04997] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Christian Lanshoeft
- Novartis Institutes for Biomedical Research, Drug
Metabolism and Pharmacokinetics, Novartis
Campus, 4056 Basel, Switzerland
- Laboratoire
de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Sarah Cianférani
- Laboratoire
de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Olivier Heudi
- Novartis Institutes for Biomedical Research, Drug
Metabolism and Pharmacokinetics, Novartis
Campus, 4056 Basel, Switzerland
| |
Collapse
|
42
|
Shah V, Lassman ME, Chen Y, Zhou H, Laterza OF. Achieving efficient digestion faster with Flash Digest: potential alternative to multi-step detergent assisted in-solution digestion in quantitative proteomics experiments. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2017; 31:193-199. [PMID: 27794205 DOI: 10.1002/rcm.7778] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 10/10/2016] [Accepted: 10/27/2016] [Indexed: 05/21/2023]
Abstract
RATIONALE In quantitative analysis of protein biomarkers and therapeutic proteins by liquid chromatography/mass spectrometry (LC/MS), it is a preferred and well-established approach to digest with proteolytic enzymes to produce smaller peptide fragments which are more suitable for LC/MS analysis than the intact protein. In-solution digestion is one widely used method for protein digestion. Proteolytically resistant proteins often require digestion times that extend beyond normal working hours and prohibit same day analysis. We evaluated the performance of an immobilized enzyme reactor (IMER) to determine if this technology could reduce method development time, digestion time and increase throughput. METHODS We digested human plasma samples using a commercially available IMER, Flash Digest, and compared it to an in-solution digestion method for analysis of three different apolipoprotein biomarkers APOE, APOC2, and APOC3. The plasma digests were analyzed via LC/MS using electrospray ionization (ESI) and multiple reaction monitoring (MRM). Value assigned calibrators were selected over a relevant physiological concentration range for each protein of interest. Quality control samples (QCs) and 'unknown' human plasma samples were analyzed with both methods. RESULTS Flash Digest significantly reduced digestion time for APOC3, the most proteolytically resistant of the three proteins, to 30 min compared with overnight used with in-solution digestion. The Flash Digest achieved comparable digestion efficiency with minimal method development and reduced sample preparation time. Both methods showed linearity over a physiologically relevant concentration range. Precision was evaluated and a percentage coefficient of variance (% CV) less than 8% was obtained during intra-day reproducibility evaluation for all three apolipoproteins with Flash Digest. Concentrations observed for QCs and unknown samples using Flash Digest were comparable to the in-solution method. CONCLUSIONS An IMER such as Flash Digest may be a potential alternative to in-solution digestion to accelerate digestion of proteolytically resistant proteins in a quantitative proteomics experiments, reduce method development time and increase throughput. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Vinit Shah
- Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Michael E Lassman
- Translational Molecular Biomarkers, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Ying Chen
- Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Haihong Zhou
- Cardiometabolic Disease, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Omar F Laterza
- Translational Molecular Biomarkers, Merck Research Laboratories, Kenilworth, NJ, USA
| |
Collapse
|
43
|
The emergence of low-cost compact mass spectrometry detectors for chromatographic analysis. Trends Analyt Chem 2016. [DOI: 10.1016/j.trac.2016.04.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
44
|
LC–MS quantification of protein drugs: validating protein LC–MS methods with predigestion immunocapture. Bioanalysis 2016; 8:1951-64. [DOI: 10.4155/bio-2016-0137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
A refinement of protein LC–MS bioanalysis is to use predigestion immunoaffinity capture to extract the drug from matrix prior to digestion. Because of their increased sensitivity, such hybrid assays have been successfully validated and applied to a number of clinical studies; however, they can also be subject to potential interferences from antidrug antibodies, circulating ligands or other matrix components specific to patient populations and/or dosed subjects. The purpose of this paper is to describe validation experiments that measure immunocapture efficiency, digestion efficiency, matrix effect and selectivity/specificity that can be used during method optimization and validation to test the resistance of the method to these potential interferences. The designs and benefits of these experiments are discussed in this report using an actual assay case study.
Collapse
|
45
|
A workflow for absolute quantitation of large therapeutic proteins in biological samples at intact level using LC-HRMS. Bioanalysis 2016; 8:1679-91. [DOI: 10.4155/bio-2016-0096] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: The commonly used LC–MS workflow to quantify protein therapeutics in biological samples is ‘bottom-up’ approach. In this study, the aim is to establish ‘top-down’ approach for absolute quantitation of therapeutic antibodies or proteins of similar sizes in biological samples at intact level. Materials & methods: Using a recombinant human monoclonal antibody as the model molecule, we present a workflow to measure large therapeutic proteins in plasma at intact level based on deconvoluted high-resolution MS (HRMS) peaks. A novel MultiQuant™ software function was developed to automatically deconvolute the peaks and process the data. Results & conclusion: The workflow showed satisfying performance. This is a proof of concept study demonstrating the feasibility of bioanalysis of large therapeutic proteins at intact level using LC-HRMS.
Collapse
|