1
|
Buitelaar PLM, de Jong KAM, Aardenburg L, van der Heijden MS, Huitema ADR, Beijnen JH, Rosing H. A multiplex UPLC-MS/MS method for the quantification of three PD-L1 checkpoint inhibitors, atezolizumab, avelumab, and durvalumab, in human serum. J Pharm Biomed Anal 2024; 243:116108. [PMID: 38522382 DOI: 10.1016/j.jpba.2024.116108] [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: 01/22/2024] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND AND AIM To support pharmacokinetic studies, a multiplex UPLC-MS/MS assay was developed and validated to quantify PD-L1 checkpoint inhibitors atezolizumab, avelumab, and durvalumab in serum. METHODS A bottom-up sample pre-treatment procedure was developed to determine atezolizumab, avelumab, and durvalumab in serum. This procedure consisted of (1) precipitation of the monoclonal antibody with ammonium sulfate, (2) reduction with dithiothreitol, (3) denaturation with methanol, and (4) tryptic digestion of the protein. The unique signature peptides resulting after sample pre-treatment of the antibodies were measured using UPLC-MS/MS with a total run time of 11 minutes. The clinical application was evaluated by analyzing 114 atezolizumab patient samples. RESULTS The developed method was found to be accurate and precise for all three analytes over a concentration range of 3.00-150 µg/mL. No endogenous interference was present in serum samples. Cross-interference experiments showed no cross-analyte interference and acceptable cross-internal standard interference. In addition, no substantial carry-over was observed. The stable isotopically labeled signature peptides were most effective in compensating for matrix effects. Recovery based on back-calculated concentrations of calibration standards and quality control samples was found to be high. The analytes were stable for at least three freeze-thaw cycles, for 42 hours at processing conditions, for at least two days at 2-8°C in the final extract, for five days before re-injection analysis at 4°C, and long-term for at least 11 months at -70°C. The assay was tested for its applicability in clinical practice. For this purpose, 114 atezolizumab patient samples were measured. CONCLUSION A multiplex UPLC-MS/MS assay was developed and validated to quantify atezolizumab, avelumab, and durvalumab in human serum. The applicability of this method was demonstrated by the analysis of clinical atezolizumab samples. The method is suitable to support clinical pharmacokinetic studies involving atezolizumab, avelumab, or durvalumab.
Collapse
Affiliation(s)
- Pauline L M Buitelaar
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Karen A M de Jong
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Leon Aardenburg
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Pharmacology, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek Hospital, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Werth EG, Roos D, Philip ET. Immunocapture LC-MS methods for pharmacokinetics of large molecule drugs. Bioanalysis 2024; 16:165-177. [PMID: 38348660 PMCID: PMC11845110 DOI: 10.4155/bio-2023-0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/31/2024] [Indexed: 03/26/2024] Open
Abstract
Implementation of immunocapture LC-MS methods to characterize the pharmacokinetic profile of large molecule drugs has become a widely used technique over the past decade. As the pharmaceutical industry strives for speediness into clinical development without jeopardizing quality, robust assays with generic application across the pipeline are becoming instrumental in bioanalysis, especially in early-stage development. This review highlights the capabilities and challenges involved in hybrid immunocapture LC-MS techniques and its continued applications in nonclinical and clinical pharmacokinetic assay design. This includes a comparison of LC-MS-based approaches to conventional ligand-binding assays and the driving demands in large molecule drug portfolios including growing sensitivity requirements and the unique challenges of new modalities requiring innovation in the bioanalytical laboratory.
Collapse
Affiliation(s)
- Emily G Werth
- Drug Metabolism and Pharma cokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT06877, USA
| | - David Roos
- Drug Metabolism and Pharma cokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT06877, USA
| | - Elsy T Philip
- Drug Metabolism and Pharma cokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT06877, USA
| |
Collapse
|
3
|
Holbrook JH, Kemper GE, Hummon AB. Quantitative mass spectrometry imaging: therapeutics & biomolecules. Chem Commun (Camb) 2024; 60:2137-2151. [PMID: 38284765 PMCID: PMC10878071 DOI: 10.1039/d3cc05988j] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
Mass spectrometry imaging (MSI) has become increasingly utilized in the analysis of biological molecules. MSI grants the ability to spatially map thousands of molecules within one experimental run in a label-free manner. While MSI is considered by most to be a qualitative method, recent advancements in instrumentation, sample preparation, and development of standards has made quantitative MSI (qMSI) more common. In this feature article, we present a tailored review of recent advancements in qMSI of therapeutics and biomolecules such as lipids and peptides/proteins. We also provide detailed experimental considerations for conducting qMSI studies on biological samples, aiming to advance the methodology.
Collapse
Affiliation(s)
- Joseph H Holbrook
- Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio 43210, USA.
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, USA
| | - Gabrielle E Kemper
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, USA
| | - Amanda B Hummon
- Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio 43210, USA.
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| |
Collapse
|
4
|
Dong L, Chen S, Piatkov K, Wei D, Qian MG. Quantifying LAGA mutated mouse IgG2a monoclonal antibody with a rapid pepsin digestion enabled immunoaffinity LC/MS/MS assay. MAbs 2024; 16:2379903. [PMID: 39077932 PMCID: PMC11290748 DOI: 10.1080/19420862.2024.2379903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024] Open
Abstract
A sensitive and specific bioanalytical method was required to measure the exposure of a LAGA-mutated surrogate mouse IgG2a monoclonal antibody in mouse plasma, but the lack of highly specific reagents for the LAGA mutant hindered the development of a ligand-binding assay. Equally problematic is that no sensitive unique tryptic peptides suitable for quantitative mass spectrometric analysis could be identified in the mIgG2a complementarity-determining regions. To overcome these challenges, a trypsin alternative pepsin, an aspartic protease, was systematically investigated for its use in digesting the mutated mIgG2a antibody to allow generation of signature peptides for the bioanalytical quantification purpose. After a series of evaluations, a rapid one-hour pepsin digestion protocol was established for the mutated Fc backbone. Consequently, a new pepsin digestion-based liquid chromatography-tandem mass spectrometry (LC/MS/MS) method was successfully developed to support the mouse pharmacokinetic (PK) sample analysis. In brief, robust and reproducible C-terminal cleavage of both leucine and phenylalanine near the double mutation site of the mutated mIgG2a was accomplished at pH ≤2 and 37°C. Combined with a commercially available rat anti-mIgG2a heavy-chain antibody, the established immunoaffinity LC/MS/MS assay achieved a limit of quantitation of 20 ng/mL in the dynamic range of interest with satisfactory assay precision and accuracy. The successful implementation of this novel approach in discovery PK studies eliminates the need for tedious and costly generation of specific immunocapturing reagents for the LAGA mutants. The approach should be widely applicable for developing popular LAGA mutant-based biological therapeutics.
Collapse
Affiliation(s)
- Linlin Dong
- Department of Drug Metabolism, Pharmacokinetics & Modeling, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Susan Chen
- Department of Drug Metabolism, Pharmacokinetics & Modeling, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Konstantin Piatkov
- Department of Drug Metabolism, Pharmacokinetics & Modeling, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Dong Wei
- Department of Drug Metabolism, Pharmacokinetics & Modeling, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Mark G. Qian
- Department of Drug Metabolism, Pharmacokinetics & Modeling, Takeda Development Center Americas, Inc., Cambridge, MA, USA
| |
Collapse
|
5
|
Song JG, Baral KC, Kim GL, Park JW, Seo SH, Kim DH, Jung DH, Ifekpolugo NL, Han HK. Quantitative analysis of therapeutic proteins in biological fluids: recent advancement in analytical techniques. Drug Deliv 2023; 30:2183816. [PMID: 36880122 PMCID: PMC10003146 DOI: 10.1080/10717544.2023.2183816] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/06/2023] [Indexed: 03/08/2023] Open
Abstract
Pharmaceutical application of therapeutic proteins has been continuously expanded for the treatment of various diseases. Efficient and reliable bioanalytical methods are essential to expedite the identification and successful clinical development of therapeutic proteins. In particular, selective quantitative assays in a high-throughput format are critical for the pharmacokinetic and pharmacodynamic evaluation of protein drugs and to meet the regulatory requirements for new drug approval. However, the inherent complexity of proteins and many interfering substances presented in biological matrices have a great impact on the specificity, sensitivity, accuracy, and robustness of analytical assays, thereby hindering the quantification of proteins. To overcome these issues, various protein assays and sample preparation methods are currently available in a medium- or high-throughput format. While there is no standard or universal approach suitable for all circumstances, a liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay often becomes a method of choice for the identification and quantitative analysis of therapeutic proteins in complex biological samples, owing to its high sensitivity, specificity, and throughput. Accordingly, its application as an essential analytical tool is continuously expanded in pharmaceutical R&D processes. Proper sample preparation is also important since clean samples can minimize the interference from co-existing substances and improve the specificity and sensitivity of LC-MS/MS assays. A combination of different methods can be utilized to improve bioanalytical performance and ensure more accurate quantification. This review provides an overview of various protein assays and sample preparation methods, with particular emphasis on quantitative protein analysis by LC-MS/MS.
Collapse
Affiliation(s)
- Jae Geun Song
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Kshitis Chandra Baral
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Gyu-Lin Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Ji-Won Park
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Soo-Hwa Seo
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Da-Hyun Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Dong Hoon Jung
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Nonye Linda Ifekpolugo
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| | - Hyo-Kyung Han
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Korea
| |
Collapse
|
6
|
Shi M, Duan X, Zheng X, Lu D, Ge Y, Zhang N, Liu Y, You J, Xue H, Yin L. Quantification of human serum albumin by combining chymotrypsin/trypsin digestion coupled with LC-MS/MS technique. Anal Biochem 2023; 680:115316. [PMID: 37689095 DOI: 10.1016/j.ab.2023.115316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
The quantification of albumin is important in clinical medicine because the concentration of albumin in biological fluids is closely related to human health. In this study, we developed a highly selective and robust assay to determine human serum albumin (HSA) in human plasma by combining chymotrypsin/trypsin digestion coupled with targeted LC-MS/MS technique. Human plasma samples were denatured, reduced, alkylated, and digested with both chymotrypsin and trypsin to generate surrogate peptides. A unique chymotryptic peptide (NAETF) arising from human serum albumin was finally selected for targeted LC-MS/MS detection and quantification. Numerous parameters related to the targeted LC-MS/MS assay were evaluated, including lower limit of quantitation (LLOQ), linearity range, enzyme digestion efficiency, accuracy and precision. The LC-MS/MS assay was linear in the concentration range 0.05-1 mg/mL with intra-day and inter-day precision <10.2% and accuracy ranging from -3.94% to 4.89%. The assay was successfully applied to determine HSA in 148 human plasma samples.
Collapse
Affiliation(s)
- Meiyun Shi
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 1242(21), PR China; Aim Honesty Biopharmaceutical Co. LTD, Dalian, 116600, PR China
| | - Xujian Duan
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 1242(21), PR China
| | - Xinyue Zheng
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 1242(21), PR China
| | - Di Lu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 1242(21), PR China
| | - Yuncheng Ge
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 1242(21), PR China
| | - Ning Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 1242(21), PR China
| | - Yajun Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 1242(21), PR China
| | - Jiansong You
- Aim Honesty Biopharmaceutical Co. LTD, Dalian, 116600, PR China
| | - Hongyu Xue
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 1242(21), PR China.
| | - Lei Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, 1242(21), PR China.
| |
Collapse
|
7
|
Gautam SS, Singh SP. Immunopurification Reagents and Their Application in Biologics and Biomarker Quantitation Using LC-MS/MS in Drug Discovery. J Chromatogr Sci 2023; 61:799-805. [PMID: 36469494 DOI: 10.1093/chromsci/bmac096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Indexed: 10/01/2023]
Abstract
The LC-MS/MS technology is one of the most utilized bio-analytical tools owing to its advantage of selectivity, sensitivity and multitasking. The advent of novel biological therapies and increasing demand for protein biomarker identification and quantitation have put the LC-MS/MS technology at the forefront. The questions which are been posed to the LC-MS/MS scientist are complex. The complexity of the question increases further with the matrices in which these questions need to be answered. To bring down the complexity of the analysis, LC-MS/MS technology is utilizing the immunopurification (IP) technique as the new sample preparation technique. The IP reagents are the most common reagents which are used to decrease the matrices' complexity and allow the LC-MS/MS system to reach greater sensitivity. The utilization of these reagents is increasing every day, but the proper utilization of these reagents is still unknown to the common analyst in drug discovery. The present review throws light on the utilization aspect of these reagents, as we have classified these reagents on basis of their utilization, which will allow the readers to gain an understanding of these reagents. This review will also talk about the merits and the demerits of each approach and the current understanding of utilizing these reagents.
Collapse
Affiliation(s)
- Shashyendra Singh Gautam
- Toxicokinetics Laboratory/Analytical Chemistry Laboratory, Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 200102, India
- Biocon-Bristol-Myers Squibb Research Centre, Syngene International Ltd, Bangalore 560100, India
| | - Sheelendra Pratap Singh
- Toxicokinetics Laboratory/Analytical Chemistry Laboratory, Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 200102, India
| |
Collapse
|
8
|
Massonnet P, Grifnée E, Farré-Segura J, Demeuse J, Huyghebaert L, Dubrowski T, Dufour P, Schoumacher M, Peeters S, Le Goff C, Cavalier E. Concise review on the combined use of immunocapture, mass spectrometry and liquid chromatography for clinical applications. Clin Chem Lab Med 2023; 61:1700-1707. [PMID: 37128992 DOI: 10.1515/cclm-2023-0253] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
Immunocapture is now a well-established method for sample preparation prior to quantitation of peptides and proteins in complex matrices. This short review will give an overview of some clinical applications of immunocapture methods, as well as protocols with and without enzymatic digestion in a clinical context. The advantages and limitations of both approaches are discussed in detail. Challenges related to the choice of mass spectrometer are also discussed. Top-down, middle-down, and bottom-up approaches are discussed. Even though immunocapture has its limitations, its main advantage is that it provides an additional dimension of separation and/or isolation when working with peptides and proteins. Overall, this short review demonstrates the potential of such techniques in the field of proteomics-based clinical medicine and paves the way for better personalized medicine.
Collapse
Affiliation(s)
- Philippe Massonnet
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Elodie Grifnée
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Jordi Farré-Segura
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Justine Demeuse
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Loreen Huyghebaert
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Thomas Dubrowski
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Patrice Dufour
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | | | - Stéphanie Peeters
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
| | - Caroline Le Goff
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, CHU de Liège, Centre de Recherche Intégré sur les Médicaments (CIRM), Liège, Belgium
- Clinical Chemistry, CIRM, University of Liège, Liège, Belgium
| |
Collapse
|
9
|
Qin Q, Gong L. Current Analytical Strategies for Antibody-Drug Conjugates in Biomatrices. Molecules 2022; 27:6299. [PMID: 36234836 PMCID: PMC9572530 DOI: 10.3390/molecules27196299] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/27/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a new class of biotherapeutics, consisting of a cytotoxic payload covalently bound to an antibody by a linker. Ligand-binding assay (LBA) and liquid chromatography-mass spectrometry (LC-MS) are the favored techniques for the analysis of ADCs in biomatrices. The goal of our review is to provide current strategies related to a series of bioanalytical assays for pharmacokinetics (PK) and anti-drug antibody (ADA) assessments. Furthermore, the strengths and limitations of LBA and LC-MS platforms are compared. Finally, potential factors that affect the performance of the developed assays are also provided. It is hoped that the review can provide valuable insights to bioanalytical scientists on the use of an integrated analytical strategy involving LBA and LC-MS for the bioanalysis of ADCs and related immunogenicity evaluation.
Collapse
Affiliation(s)
- Qiuping Qin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Likun Gong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 101408, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| |
Collapse
|
10
|
Lu L, Liu X, Zuo C, Zhou J, Zhu C, Zhang Z, Fillet M, Crommen J, Jiang Z, Wang Q. In vitro/in vivo degradation analysis of trastuzumab by combining specific capture on HER2 mimotope peptide modified material and LC-QTOF-MS. Anal Chim Acta 2022; 1225:340199. [DOI: 10.1016/j.aca.2022.340199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/12/2022] [Accepted: 07/22/2022] [Indexed: 11/28/2022]
|
11
|
Kramlinger VM, Dalvie D, Heck CJS, Kalgutkar AS, O'Neill J, Su D, Teitelbaum AM, Totah RA. Future of Biotransformation Science in the Pharmaceutical Industry. Drug Metab Dispos 2022; 50:258-267. [PMID: 34921097 DOI: 10.1124/dmd.121.000658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/23/2021] [Indexed: 11/22/2022] Open
Abstract
Over the past decades, the number of scientists trained in departments dedicated to traditional medicinal chemistry, biotransformation and/or chemical toxicology have seemingly declined. Yet, there remains a strong demand for such specialized skills in the pharmaceutical industry, particularly within drug metabolism/pharmacokinetics (DMPK) departments. In this position paper, the members of the Biotransformation, Mechanisms, and Pathways Focus Group (BMPFG) steering committee reflect on the diverse roles and responsibilities of scientists trained in the biotransformation field in pharmaceutical companies and contract research organizations. The BMPFG is affiliated with the International Society for the Study of Xenobiotics (ISSX) and was specifically created to promote the exchange of ideas pertaining to topics of current and future interest involving the metabolism of xenobiotics (including drugs). The authors also delve into the relevant education and diverse training skills required to successfully nurture the future cohort of industry biotransformation scientists and guide them toward a rewarding career path. The ability of scientists with a background in biotransformation and organic chemistry to creatively solve complex drug metabolism problems encountered during research and development efforts on both small and large molecular modalities is exemplified in five relevant case studies. Finally, the authors stress the importance and continued commitment to training the next generation of biotransformation scientists who are not only experienced in the metabolism of conventional small molecule therapeutics, but are also equipped to tackle emerging challenges associated with new drug discovery modalities including peptides, protein degraders, and antibodies. SIGNIFICANCE STATEMENT: Biotransformation and mechanistic drug metabolism scientists are critical to advancing chemical entities through discovery and development, yet the number of scientists academically trained for this role is on the decline. This position paper highlights the continuing demand for biotransformation scientists and the necessity of nurturing creative ways to train them and guarantee the future growth of this field.
Collapse
Affiliation(s)
- Valerie M Kramlinger
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Deepak Dalvie
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Carley J S Heck
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Amit S Kalgutkar
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - James O'Neill
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Dian Su
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Aaron M Teitelbaum
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Rheem A Totah
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| |
Collapse
|
12
|
Non-Antibody-Based Binders for the Enrichment of Proteins for Analysis by Mass Spectrometry. Biomolecules 2021; 11:biom11121791. [PMID: 34944435 PMCID: PMC8698613 DOI: 10.3390/biom11121791] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/24/2021] [Accepted: 11/27/2021] [Indexed: 02/07/2023] Open
Abstract
There is often a need to isolate proteins from body fluids, such as plasma or serum, prior to further analysis with (targeted) mass spectrometry. Although immunoglobulin or antibody-based binders have been successful in this regard, they possess certain disadvantages, which stimulated the development and validation of alternative, non-antibody-based binders. These binders are based on different protein scaffolds and are often selected and optimized using phage or other display technologies. This review focuses on several non-antibody-based binders in the context of enriching proteins for subsequent liquid chromatography-mass spectrometry (LC-MS) analysis and compares them to antibodies. In addition, we give a brief introduction to approaches for the immobilization of binders. The combination of non-antibody-based binders and targeted mass spectrometry is promising in areas, like regulated bioanalysis of therapeutic proteins or the quantification of biomarkers. However, the rather limited commercial availability of these binders presents a bottleneck that needs to be addressed.
Collapse
|
13
|
Affinity capture in bottom-up protein analysis - Overview of current status of proteolytic peptide capture using antibodies and molecularly imprinted polymers. Anal Chim Acta 2021; 1182:338714. [PMID: 34602193 DOI: 10.1016/j.aca.2021.338714] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022]
Abstract
Antibody-based affinity capture has become the gold standard in sample preparation for determination of low-abundance protein biomarkers in biological matrices prior to liquid chromatography-mass spectrometry (LC-MS) determination. This comprises both capture of intact proteins prior to the digestion step and capture of proteolytic peptides after digestion of the sample. The latter can be performed both using antibodies specifically developed to capture target proteolytic peptides, as well as by the less explored use of anti-protein antibodies to capture the proteolytic epitope peptide. Molecularly imprinted polymers (MIPs), also called plastic antibodies are another affinity-based approach emerging as sample preparation technique in LC-MS based protein biomarker analysis. The current review gives a critical and comprehensive overview of proteolytic peptide capture using antibodies and MIPs in LC-MS based protein biomarker determination during the last five years. The main emphasis is on capture of non-modified peptides, while a brief overview of affinity capture of peptides containing post-translational modifications (PTMs) is provided.
Collapse
|
14
|
Olaleye O, Spanov B, Ford R, Govorukhina N, van de Merbel NC, Bischoff R. Enrichment and Liquid Chromatography-Mass Spectrometry Analysis of Trastuzumab and Pertuzumab Using Affimer Reagents. Anal Chem 2021; 93:13597-13605. [PMID: 34582688 PMCID: PMC8515351 DOI: 10.1021/acs.analchem.1c02807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Trastuzumab and pertuzumab are monoclonal antibodies used in the treatment of human epidermal growth factor receptor-2 (HER2)-positive breast cancer. Therapeutic proteins may undergo chemical modifications that may affect the results of bioanalytical assays, as well as their therapeutic efficacy. Modifications may arise during production and storage, as well as after administration to patients. Studying in vivo biotransformation of monoclonal, therapeutic antibodies requires their enrichment from plasma to discriminate them from endogenous antibodies, as well as from other plasma proteins. To this end, we screened Affimer reagents for selectivity toward trastuzumab or pertuzumab. Affimer reagents are alternative binding proteins possessing two variable binding loops that are based on the human protease inhibitor stefin A or phytocystatin protein scaffolds. Affimer reagents were selected from an extensive library by phage display. The four best-performing binders for each therapeutic antibody were prioritized using a microtiter plate-based approach combined with liquid chromatography-mass spectrometry (LC-MS) in the selected reaction monitoring (SRM) mode. These Affimer reagents were immobilized via engineered 6-His or Cys tags to Ni2+- or maleimide beads, respectively. Recovery values of 70% and higher were obtained for both trastuzumab and pertuzumab when spiked at 100, 150, and 200 μg/mL concentrations in human plasma followed by trypsin digestion in the presence of 0.5% sodium deoxycholate and 10 mM dithiothreitol (DTT). Notably, the maleimide beads showed undetectable unspecific binding to endogenous immunoglobulin G (IgGs) or other plasma proteins when analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). The enrichment method was applied to samples from stress tests of the antibodies at 37 °C to mimic in vivo conditions.
Collapse
Affiliation(s)
- Oladapo Olaleye
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Baubek Spanov
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Robert Ford
- Avacta Life Sciences Limited, Unit 20, Ash Way, Thorp Arch Estate, Wetherby LS23 7FA, United Kingdom
| | - Natalia Govorukhina
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Nico C van de Merbel
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.,Bioanalytical Laboratory, PRA Health Sciences, Early Development Services, Amerikaweg 18, Assen 9407 TK, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|
15
|
Rashid F, Baghla R, Kale P, Shah M, Malakar D, Pillai M. Absolute Quantification of Follicle Stimulating Hormone (FSH) Using Its Signature Peptides and Enzymatic Digestion in Human Serum by UPLC/LC–MS/MS. Chromatographia 2021. [DOI: 10.1007/s10337-021-04057-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
16
|
Bioanalysis of therapeutic monoclonal antibody by peptide adsorption-controlled LC-MS. Bioanalysis 2021; 13:265-276. [PMID: 33543661 DOI: 10.4155/bio-2020-0262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: We aimed to develop an easy, low-cost and versatile mass spectrometric method for the bioanalysis of a therapeutic monoclonal antibody (mAb) in human serum that employs peptide adsorption-controlled (PAC)-LC/MS using selected reaction monitoring mode (LC-MS/MS-SRM). Materials & methods: Rituximab was used as a model mAb. To apply the method to human serum samples, a peptide of the complementarity-determining region was selected as the surrogate peptide. The usefulness of PAC-LC-MS/MS-SRM was evaluated by a collaborative study. Results: The calibration curve ranged from 0.5 (or 1.0) to 1000.0 μg/ml. The selectivity, linearity, accuracy and precision met the predefined acceptance criteria. Conclusion: Our method could be a useful bioanalytical method for the quantification of mAbs in clinical samples.
Collapse
|
17
|
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
|
18
|
Levernæs MCS, Moe AU, Bøe SL, Paus E, Reubsaet L, Halvorsen TG. Liquid chromatography mass spectrometry based characterization of epitope configurations. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2020; 12:5476-5484. [PMID: 33141131 DOI: 10.1039/d0ay01283a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Here we evaluate a quick and easy tool for determination of epitope configuration using immunocapture and liquid chromatography mass spectrometry (LC-MS) subsequent to pre-treatment of the target protein to disrupt its three-dimensional structure. The approach can be a valuable screening tool to identify antibodies that can be used in peptide capture by anti-protein antibodies. The experimental set-up was established using seven monoclonal antibodies (mAbs) with known linear or conformational epitope recognition. The mAbs were developed to target either of the two biomarkers, progastrin releasing peptide (ProGRP) or human chorionic gonadotropin (hCG). Best coherence with established epitope configuration was seen when using both denaturation, reduction and alkylation as pre-treatment method of the proteins (≥70% reduction in MS signal intensity compared to control) prior to immunocapture and LC-MS determination. The final method was used to determine the epitope configuration of four anti-thyroglobulin mAbs with unknown epitope configuration; all four mAbs showed configurational epitope recognition. These results were also supported by western blots of native, and reduced and alkylated protein using three of the evaluated mAbs, and by analysis native, and reduced and alkylated protein in a routine immunofluorometric assay employing the four evaluated antibodies.
Collapse
|
19
|
Kellie JF, Pannullo KE, Li Y, Fraley K, Mayer A, Sychterz CJ, Szapacs ME, Karlinsey MZ. Antibody Subunit LC-MS Analysis for Pharmacokinetic and Biotransformation Determination from In-Life Studies for Complex Biotherapeutics. Anal Chem 2020; 92:8268-8277. [DOI: 10.1021/acs.analchem.0c00520] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
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
|
Fan PR, Zhao X, Wei ZH, Huang YP, Liu ZS. Robust immobilized enzyme reactor based on trimethylolpropane trimethacrylate organic monolithic matrix through “thiol-ene” click reaction. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2019.109456] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Generic MS-based method for the bioanalysis of therapeutic monoclonal antibodies in nonclinical studies. Bioanalysis 2020; 12:231-243. [DOI: 10.4155/bio-2019-0253] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: A generic bioanalytical method was developed to quantify therapeutic IgG1 monoclonal antibodies (mAbs) in mouse sera by combining an easy sample preparation method with LC/MS using selected reaction monitoring. Materials & methods: Rituximab and trastuzumab were used as model mAbs. A synthetic stable isotope-labeled peptide or a stable isotope-labeled mAb was used as an internal standard. The method feasibility was evaluated by a collaborative study involving six laboratories. Results: The calibration curve ranged from 1.0 to 1000.0 μg/ml (correlation coefficient >0.99). The validation parameters including selectivity, linearity of calibration curve, accuracy and precision met the predefined acceptance criteria. Conclusion: Our method is a useful bioanalytical method for the quantification of therapeutic IgG mAbs in nonclinical animal studies.
Collapse
|
23
|
Fluoride reactivation-enabled sensitive quantification of tabun adducts on human serum albumin by GC-MS/MS via isotope dilution. Bioanalysis 2019; 11:2145-2159. [PMID: 31729243 PMCID: PMC6923783 DOI: 10.4155/bio-2019-0161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Organophosphorus nerve agents inhibit the cholinesterase activity by phosphylation of the active site serine. The resulting phosphylated cholinesterase and adducts on human serum albumin (HSA) are appropriate biomarkers for nerve agents exposure. Several methods have been developed for the detection of nerve agents, including fluoride reactivation or alkaline cleavage. It was previously thought that some nerve agents adducts to HSA could not be detected via fluoride regeneration. In our study, the results showed that tabun (GA) adducts of HSA could be detected by fluoride regeneration. The sample preparation included acetone precipitation, washing and SPE. Deuterated tabun (d5-GA) was applied as the internal standard. The product of regenerated fluorotabun is detected with a good linearity (R2 > 0.997) in the concentration range from 0.02 to 100.0 ng/ml, small relative standard deviation (≤6.89%) and favorable recoveries between 94.8 and 106.3%. The established preparation confirmed the fluorotabun was regenerated from the GA-HSA adducts.
Collapse
|
24
|
Dong S, Zhang A, Gu Y, Lu S, Teng L, Wang R, Liu J, Fan H, Si D. Direct quantification of intact FIM in monkey plasma using a selective chromatography-tandem mass spectrometry method: Application in a pharmacokinetic study. Biomed Chromatogr 2019; 34:e4729. [PMID: 31656040 DOI: 10.1002/bmc.4729] [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: 06/04/2019] [Revised: 10/09/2019] [Accepted: 10/19/2019] [Indexed: 11/06/2022]
Abstract
FIM protein, which consists of 155 amino acids, was developed as a novel GLP-1 analog to reduce blood glucose, and pharmacodynamic results showed that it had a certain effect when used in treating Alzheimer's disease. The molecular weight of FIM is 16,304 Da. In theory, the concentration of FIM in biological samples should be determined by the ligand binding assay method or indirectly quantified using LC-MS/MS instrumentation. However, the above methods are complex and time-consuming. In this study, we successfully developed a simpler LC-MS/MS method for directly quantifying the intact FIM protein in monkey plasma for the first time. The chromatographic separation of FIM was achieved using an InertSustain Bio C18 column with a mobile phase of acetonitrile containing 0.1% formic acid (A)-water containing 0.1% formic acid (B) at a flow rate of 0.3 ml/min. Good linearity was observed in the concentration range of 5-500 ng/ml (r2 > 0.99). The intra- and inter-day precisions (expressed as relative standard deviation, RSD) of FIM were 2.30-12.8 and 7.30-13.2%, respectively. The intra- and inter-day accuracies (expressed as a relative error, RE) were -12.7-6.55 and - 10.1-0.892%, respectively. This method was successfully applied for a pharmacokinetic study of the FIM protein in four monkeys after subcutaneous administration.
Collapse
Affiliation(s)
- Shiqi Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Aijie Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yuan Gu
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Shuixiu Lu
- Prosperous Biopharma Co. Ltd, Beijing, China
| | | | - Rui Wang
- Prosperous Biopharma Co. Ltd, Beijing, China
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Huirong Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Duanyun Si
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| |
Collapse
|
25
|
Label-free protein quantification after ultrafast digestion of complex proteomes using ultrasonic energy and immobilized-trypsin magnetic nanoparticles. Talanta 2019; 196:262-270. [DOI: 10.1016/j.talanta.2018.12.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/27/2022]
|
26
|
Development of two complementary LC–HRMS methods for analyzing sotatercept in dried blood spots for doping controls. Bioanalysis 2019; 11:923-940. [DOI: 10.4155/bio-2018-0313] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: sotatercept is a therapeutic Fc-fusion protein with erythropoiesis-stimulating activity. Due to a potential abuse of the drug by athletes in professional sports, a sensitive detection method is required. In sports drug testing, alternative matrices such as dried blood spots (DBS) are gaining increasing attention as they can provide several advantages over conventional matrices. Materials & methods: Herein, two complementary LC–high-resolution mass spectrometry (HRMS) detection methods for sotatercept from DBS, an initial testing procedure (ITP) and a confirmation procedure (CP) were developed and validated for the first time. Both methods comprise an ultrasonication-assisted extraction, affinity enrichment, proteolytic digestion and HRMS detection. Results & conclusion: For the multianalyte ITP, artificial samples fortified with sotatercept, luspatercept and bimagrumab, and authentic specimens containing bimagrumab were successfully analyzed as proof-of-concept. The validated detection methods for sotatercept are fit for purpose and the ITP was shown to be suitable for the detection of novel IgG-based pharmaceuticals in doping control DBS samples.
Collapse
|
27
|
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
|
28
|
Levernæs MCS, Farhat B, Oulie I, Abdullah SS, Paus E, Reubsaet L, Halvorsen TG. Immunocapture sample clean-up in determination of low abundant protein biomarkers – a feasibility study of peptide capture by anti-protein antibodies. RSC Adv 2019; 9:34902-34911. [PMID: 35702551 PMCID: PMC9097496 DOI: 10.1039/c9ra05071j] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/21/2019] [Indexed: 01/04/2023] Open
Abstract
Immunocapture in mass spectrometry based targeted protein analysis using a bottom-up workflow is nowadays mainly performed by target protein extraction using anti-protein antibodies followed by tryptic digestion. Already available monoclonal antibodies (mAbs) which were developed against intact target proteins (anti-protein antibodies) can capture proteotypic epitope containing peptides after tryptic digestion of the sample. In the present paper considerations when developing a method for targeted protein quantitation through capture of epitope containing peptides are discussed and a method applying peptide capture by anti-protein antibodies is compared with conventional immunocapture MS. The model protein used for this purpose was progastrin releasing peptide (ProGRP), a validated low abundant biomarker for Small Cell Lung Cancer with reference values in serum in the pg mL−1 range. A set of mAbs which bind linear epitopes of ProGRP are available, and after a theoretical consideration, three mAbs (E146, E149 and M18) were evaluated for extraction of proteotypic epitope peptides from a complex sample. M18 was the best performing mAb for peptide capture by anti-protein antibodies, matching the LOD (54 pg mL−1) and LOQ (181 pg mL−1) of the existing conventional immunocapture LC-MS/MS method for determination of ProGRP. Peptide and protein capture using the same mAb were also compared with respect to sample clean-up, and the peptide capture workflow yielded cleaner extracts and therewith less complex chromatograms. Analysis of five patient samples demonstrated that peptide capture by anti-protein antibodies can be used for the determination of various levels of endogenously present ProGRP. Targeted protein biomarker determination by immunocapture LC-MS/MS: comparison of peptide and protein capture using anti-protein antibodies.![]()
Collapse
Affiliation(s)
| | - Bassem Farhat
- Department Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | - Inger Oulie
- Department Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | | | - Elisabeth Paus
- Department of Medical Biochemistry
- Norwegian Radium Hospital
- Oslo University Hospital
- Norway
| | - Léon Reubsaet
- Department Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | | |
Collapse
|
29
|
Irie K, Okada A, Yamasaki Y, Kokan C, Hata A, Kaji R, Fukushima K, Sugioka N, Okada Y, Katakami N, Fukushima S. An LC-MS/MS Method for Absolute Quantification of Nivolumab in Human Plasma: Application to Clinical Therapeutic Drug Monitoring. Ther Drug Monit 2018; 40:716-724. [DOI: 10.1097/ftd.0000000000000558] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
30
|
Levernæs MCS, Brandtzaeg OK, Amundsen SF, Reubsaet L, Lundanes E, Halvorsen TG, Wilson SR. Selective Fishing for Peptides with Antibody-Immobilized Acrylate Monoliths, Coupled Online with NanoLC-MS. Anal Chem 2018; 90:13860-13866. [DOI: 10.1021/acs.analchem.8b00935] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Maren C. S. Levernæs
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, Oslo NO-0316, Norway
| | | | - Sunniva Furre Amundsen
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, Oslo NO-0316, Norway
| | - Léon Reubsaet
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, Oslo NO-0316, Norway
| | - Elsa Lundanes
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
| | - Trine G. Halvorsen
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, Oslo NO-0316, Norway
| | - Steven R. Wilson
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, NO-0315 Oslo, Norway
| |
Collapse
|
31
|
Review of approaches and examples for monitoring biotransformation in protein and peptide therapeutics by MS. Bioanalysis 2018; 10:1877-1890. [PMID: 30325207 DOI: 10.4155/bio-2018-0113] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Biotherapeutic drugs have emerged in quantity in pharmaceutical pipelines, and increasingly diverse biomolecules are progressed through preclinical and clinical development. As purification, separation, mass spectrometer detection and data processing capabilities improve, there is opportunity to monitor drug concentration by traditional ligand-binding assay or MS measurement and to monitor metabolism, catabolism or other biomolecular mass variants present in circulation. This review highlights approaches and examples of monitoring biotransformation of biotherapeutics by MS as these techniques are poised to add value to drug development in years to come. The increased use of such approaches, and the successful quantitation of biotherapeutic structural modifications, will provide insightful data for the benefit of both researchers and patients.
Collapse
|
32
|
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
|
33
|
Development of an ELISA–LC–MS hybrid assay for quantification of biotherapeutics. Bioanalysis 2018; 10:1427-1438. [DOI: 10.4155/bio-2018-0082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Background: Magnetic bead immunocapture-LC–MS has been widely used for bioanalysis of biotherapeutic proteins. However, magnetic beads are difficult to be fully automated and more costly than ELISA plates. Aim: Develop an ELISA–LC–MS hybrid assay as an alternate platform. Results: Among seven ELISA plates tested, Pierce streptavidin plates, which did not require time-consuming capture antibody precoating steps, provided the best sensitivity and assay dynamic range (5–2500 ng/ml or 10–5000 ng/ml), similar to magnetic bead immunocapture–LC–MS assay and better than an ELISA (50–500 ng/ml). The entire procedures could be fully automated using a liquid handling system. Conclusion: This study demonstrates that ELISA–LC–MS hybrid approach using streptavidin plates represents a promising platform for bioanalysis of biotherapeutics.
Collapse
|
34
|
Dong S, Gu Y, Wei G, Si D, Liu C. Determination of liraglutide in rat plasma by a selective liquid chromatography-tandem mass spectrometry method: Application to a pharmacokinetics study. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1091:29-35. [DOI: 10.1016/j.jchromb.2018.05.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/09/2018] [Accepted: 05/15/2018] [Indexed: 02/01/2023]
|
35
|
Critical considerations for immunocapture enrichment LC–MS bioanalysis of protein therapeutics and biomarkers. Bioanalysis 2018; 10:987-995. [DOI: 10.4155/bio-2018-0062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In recent years, immunocapture enrichment coupled with LC–MS technology has seen more applications for the measurement of low abundant protein therapeutics and biomarkers in biological matrices. In this article, several critical considerations for the application of immunocapture enrichment to LC–MS bioanalysis of protein therapeutics and biomarkers, including reagent selection, reagent characterization, designing of capture format, etc. are discussed. All these considerations are critical in developing reliable and robust bioanalytical assays with high assay specificity and sensitivity. Successful examples using the immunocapture LC–MS approach in the quantification of biotherapeutic and low abundant protein biomarkers will also be discussed.
Collapse
|
36
|
HASHII N, UTOH M, OHTSU Y, KATO N, GODA R, GOTO R, SHIMIZU H, TAKAMURA F, HOSHINO M, MABUCHI M, YAMAGUCHI T, ISHII-WATABE A, KATORI N. Bioanalytical Quantification of Therapeutic Antibodies by Liquid Chromatography/mass Spectrometry. CHROMATOGRAPHY 2018. [DOI: 10.15583/jpchrom.2017.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
| | | | | | - Nozomu KATO
- Translational Research Department, Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corp
| | | | | | | | | | | | - Masanari MABUCHI
- DMPK Research Laboratories, Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corp
| | | | | | | |
Collapse
|
37
|
2D-LC–MS/MS to measure cleaved high-molecular-weight kininogen in human plasma as a biomarker for C1-INH-HAE. Bioanalysis 2017; 9:1477-1491. [DOI: 10.4155/bio-2017-0105] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: C1-INH-HAE is caused by activation of plasma kallikrein which subsequently cleaves high-molecular-weight kininogen (HMWK) to generate bradykinin and cHMWK. Materials & methods: A novel ion-pair 2D LC–MS/MS assay was developed to measure the 46 kDa cHMWK in plasma as a biomarker for C1-INH-HAE. The sample preparation included sodium dodecyl sulfate denaturation, methanol crash, chymotryptic digestion and peptide enrichment by solid phase extraction. Results: The LLOQ was 200 ng/ml. The overall cHMWK recovery combining crash and digestion was 57.5%. The precision of the method was ≤12.7% and accuracy ≤-13.8%. Conclusion: A reagent-free LC–MS assay has been developed for the quantitation of 46 kDa cHMWK, which was shown to be elevated in plasma of C1-INH-HAE patients due to C1-INH deficiency relative to that of healthy subjects.
Collapse
|
38
|
Levernæs MCS, Broughton MN, Reubsaet L, Halvorsen TG. To elute or not to elute in immunocapture bottom-up LC–MS. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1055-1056:51-60. [DOI: 10.1016/j.jchromb.2017.03.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/03/2017] [Accepted: 03/23/2017] [Indexed: 02/07/2023]
|