1
|
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
|
2
|
Ayurini M, Chandler PG, O’Leary PD, Wang R, Rudd D, Milewska KD, Malins LR, Buckle AM, Hooper JF. Polymer End Group Control through a Decarboxylative Cobalt-Mediated Radical Polymerization: New Avenues for Synthesizing Peptide, Protein, and Nanomaterial Conjugates. JACS AU 2022; 2:169-177. [PMID: 35098233 PMCID: PMC8790747 DOI: 10.1021/jacsau.1c00453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Indexed: 06/14/2023]
Abstract
Cobalt-mediated radical polymerizations (CMRPs) have been initiated by the radical decarboxylation of tetrachlorophthalimide activated esters. This allows for the controlled radical polymerization of activated monomers across a broad temperature range with a single cobalt species, with the incorporation of polymer end groups derived from simple carboxylic acid derivatives and termination with an organozinc reagent. This method has been applied to the synthesis of a polymer/graphene conjugate and a water-soluble protein/polymer conjugate, demonstrating the first examples of CMRP in graphene and protein conjugation.
Collapse
Affiliation(s)
- Meri Ayurini
- Department
of Chemistry, Monash University, Clayton, 3800 Victoria, Australia
- Chemistry
Department, Universitas Pertamina, South Jakarta 12220, Indonesia
| | - Peter G. Chandler
- Department
of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, 3800 Victoria, Australia
| | - Paul D. O’Leary
- Department
of Chemistry, Monash University, Clayton, 3800 Victoria, Australia
| | - Ruoxin Wang
- Department
of Chemical Engineering, Monash University, Clayton, 3800 Victoria, Australia
| | - David Rudd
- Monash
Institute of Pharmaceutical Science, Parkville, 3052 Victoria, Australia
| | - Karen D. Milewska
- Research
School of Chemistry, Australian National
University, Acton, 2601 Australian Capital Territory, Australia
| | - Lara R. Malins
- Research
School of Chemistry, Australian National
University, Acton, 2601 Australian Capital Territory, Australia
| | - Ashley M. Buckle
- Department
of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, 3800 Victoria, Australia
| | - Joel F. Hooper
- Department
of Chemistry, Monash University, Clayton, 3800 Victoria, Australia
| |
Collapse
|
3
|
Hyung SJ, Leipold DD, Lee DW, Kaur S, Saad OM. Multiplexed Quantitative Analysis of Antibody-Drug Conjugates with Labile CBI-Dimer Payloads In Vivo Using Immunoaffinity LC-MS/MS. Anal Chem 2021; 94:1158-1168. [PMID: 34958550 DOI: 10.1021/acs.analchem.1c04338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Quantitative analysis of antibody-drug conjugates (ADCs) involves cleavage of ADCs into smaller analytes representing different components and subsequent measurements from multiple assays for a more comprehensive pharmacokinetic (PK) assessment. Multiple PK analytes including the drug remaining conjugated to the antibody (or antibody-conjugated drug, acDrug) and total antibody can be accessed simultaneously using a multiplex assay by proteolytic digestion of an ADC, if the sites of conjugation are homogeneous for an ADC and the linker drug is stable to proteases. Herein, a multiplexed immunoaffinity liquid chromatography-mass spectrometry (LC-MS)/MS PK assay is described involving immunoaffinity enrichment, enzymatic conversion of prodrug, trypsin digestion, and LC-MS/MS as applied to next-generation ADCs constructed from linker drugs bearing dimeric cyclopropabenzindole (CBI) payloads (duocarmycin analogues). The cytotoxic payload is chemically labile, requiring extensive optimization in sample preparation steps to stabilize the drug without ex vivo modification and to convert the prodrug into a single active form of the drug. The qualification data for this assay format showed that this approach provides robust acDrug and total antibody data and can be extended to ADCs with different monoclonal antibody frameworks and linker chemistries. Applications of this multiplexed assay to support preclinical studies are presented.
Collapse
Affiliation(s)
- Suk-Joon Hyung
- BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Douglas D Leipold
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Donna W Lee
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Surinder Kaur
- BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Ola M Saad
- BioAnalytical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
4
|
The role of ligand-binding assay and LC-MS in the bioanalysis of complex protein and oligonucleotide therapeutics. Bioanalysis 2021; 13:931-954. [PMID: 33998268 DOI: 10.4155/bio-2021-0009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ligand-binding assay (LBA) and LC-MS have been the preferred bioanalytical techniques for the quantitation and biotransformation assessment of various therapeutic modalities. This review provides an overview of the applications of LBA, LC-MS/MS and LC-HRMS for the bioanalysis of complex protein therapeutics including antibody-drug conjugates, fusion proteins and PEGylated proteins as well as oligonucleotide therapeutics. The strengths and limitations of LBA and LC-MS, along with some guidelines on the choice of appropriate bioanalytical technique(s) for the bioanalysis of these therapeutic modalities are presented. With the discovery of novel and more complex therapeutic modalities, there is an increased need for the biopharmaceutical industry to develop a comprehensive bioanalytical strategy integrating both LBA and LC-MS.
Collapse
|
5
|
Deonarain MP, Yahioglu G. Current strategies for the discovery and bioconjugation of smaller, targetable drug conjugates tailored for solid tumor therapy. Expert Opin Drug Discov 2021; 16:613-624. [PMID: 33275475 DOI: 10.1080/17460441.2021.1858050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Introduction: Antibody-Drug Conjugates (ADCs) have undergone a recent resurgence with 5 product approvals over the last 2 years but for those close to the field, it's been repeated cycles of setbacks and new innovations. A new wave of innovation is in the type of format used to deliver the cytotoxic payloads, with smaller bio-molecules being designed to have more optimal penetration and elimination properties tailored for solid tumors.Areas covered: In this review, the authors cover many of the recently described smaller-format drug conjugates (including formats such as diabodies, Fabs, scFvs, domain antibodies) with an emphasis on the types of conjugation technologies used to attach the chemical linker-payload.Expert opinion: Smaller formats are highly influenced by the structure of the linker-payload, arguably more-so than larger ADCs, so careful consideration is needed where solublising and pharmacokinetic modulation is required. High-quality conjugates are being developed with in vivo tumor efficacy and tolerability properties competitive with ADCs and with a few formats already in clinical development, we expect the pipeline to expand and to reach the market.
Collapse
Affiliation(s)
- Mahendra P Deonarain
- Antikor Biopharma Ltd, Stevenage Bioscience Catalyst, Hertfordshire, UK.,Department of Chemistry, Imperial College London, London, UK
| | - Gokhan Yahioglu
- Antikor Biopharma Ltd, Stevenage Bioscience Catalyst, Hertfordshire, UK.,Department of Chemistry, Imperial College London, London, UK
| |
Collapse
|
6
|
Deonarain MP, Xue Q. Tackling solid tumour therapy with small-format drug conjugates. Antib Ther 2020; 3:237-245. [PMID: 33928231 DOI: 10.1093/abt/tbaa024] [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: 08/26/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023] Open
Abstract
The pharmacokinetic-pharmacodynamic relationship is extremely complex and tumour drug penetration is one key parameter influencing therapeutic efficacy. In the context of antibody-drug conjugates (ADCs), which has undergone many innovation cycles and witnessed many failures, this feature is being addressed by a number of alternative technologies. Immunoglobulin-based ADCs continue to dominate the industrial landscape, but smaller formats offer the promise of more-effective cytotoxic payload delivery to solid tumours, with a higher therapeutic window afforded by the more rapid clearance. To make these smaller formats viable as delivery vehicles, a number of strategies are being employed, which will be reviewed here. These include identifying the most-appropriate size to generate the larger therapeutic window, increasing the amount of functional, cytotoxic payload delivered through conjugation or half-life extending technologies or other ways of extending the dosing without inducing toxicity.
Collapse
Affiliation(s)
- Mahendra P Deonarain
- Antikor Biopharma Ltd, Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage, Hertfordshire SG12FX, UK
| | - Quinn Xue
- Essex Biotechnology Ltd, Shun Tak Centre, Room 2818, China Merchants Tower, Connaught Road Central, Hong Kong 168-200, SAR China
| |
Collapse
|
7
|
Zhu X, Huo S, Xue C, An B, Qu J. Current LC-MS-based strategies for characterization and quantification of antibody-drug conjugates. J Pharm Anal 2020; 10:209-220. [PMID: 32612867 PMCID: PMC7322744 DOI: 10.1016/j.jpha.2020.05.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 01/28/2023] Open
Abstract
The past few years have witnessed enormous progresses in the development of antibody-drug conjugates (ADCs). Consequently, comprehensive analysis of ADCs in biological systems is critical in supporting discovery, development and evaluation of these agents. Liquid chromatography-mass spectrometry (LC-MS) has emerged as a promising and versatile tool for ADC analysis across a wide range of scenarios, owing to its multiplexing ability, rapid method development, as well as the capability of analyzing a variety of targets ranging from small-molecule payloads to the intact protein with a high, molecular resolution. However, despite this tremendous potential, challenges persist due to the high complexity in both the ADC molecules and the related biological systems. This review summarizes the up-to-date LC-MS-based strategies in ADC analysis and discusses the challenges and opportunities in this rapidly-evolving field.
Collapse
Affiliation(s)
- Xiaoyu Zhu
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
| | - Shihan Huo
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
| | - Chao Xue
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
- Department of Chemical and Biological Engineering, School of Engineering and Applied Science, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Bo An
- Exploratory Biomarker, In-vitro/In-vivo Translation, R&D Research, GlaxoSmithKline Pharmaceuticals, 1250 South Collegeville Rd, Collegeville, PA, 19426, USA
| | - Jun Qu
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
| |
Collapse
|
8
|
Chandler PG, Buckle AM. Development and Differentiation in Monobodies Based on the Fibronectin Type 3 Domain. Cells 2020; 9:E610. [PMID: 32143310 PMCID: PMC7140400 DOI: 10.3390/cells9030610] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/01/2020] [Indexed: 12/13/2022] Open
Abstract
As a non-antibody scaffold, monobodies based on the fibronectin type III (FN3) domain overcome antibody size and complexity while maintaining analogous binding loops. However, antibodies and their derivatives remain the gold standard for the design of new therapeutics. In response, clinical-stage therapeutic proteins based on the FN3 domain are beginning to use native fibronectin function as a point of differentiation. The small and simple structure of monomeric monobodies confers increased tissue distribution and reduced half-life, whilst the absence of disulphide bonds improves stability in cytosolic environments. Where multi-specificity is challenging with an antibody format that is prone to mis-pairing between chains, multiple FN3 domains in the fibronectin assembly already interact with a large number of molecules. As such, multiple monobodies engineered for interaction with therapeutic targets are being combined in a similar beads-on-a-string assembly which improves both efficacy and pharmacokinetics. Furthermore, full length fibronectin is able to fold into multiple conformations as part of its natural function and a greater understanding of how mechanical forces allow for the transition between states will lead to advanced applications that truly differentiate the FN3 domain as a therapeutic scaffold.
Collapse
Affiliation(s)
- Peter G. Chandler
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton 3800, Australia;
| | | |
Collapse
|
9
|
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
|
10
|
Beck A, D’Atri V, Ehkirch A, Fekete S, Hernandez-Alba O, Gahoual R, Leize-Wagner E, François Y, Guillarme D, Cianférani S. Cutting-edge multi-level analytical and structural characterization of antibody-drug conjugates: present and future. Expert Rev Proteomics 2019; 16:337-362. [DOI: 10.1080/14789450.2019.1578215] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Alain Beck
- Biologics CMC and Developability, IRPF - Centre d’Immunologie Pierre-Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Valentina D’Atri
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Anthony Ehkirch
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg, France
| | - Szabolcs Fekete
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Oscar Hernandez-Alba
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg, France
| | - Rabah Gahoual
- Unité de Technologies Biologiques et Chimiques pour la Santé (UTCBS), Paris 5-CNRS UMR8258 Inserm U1022, Faculté de Pharmacie, Université Paris Descartes, Paris, France
| | - Emmanuel Leize-Wagner
- Laboratoire de Spectrométrie de Masse des Interactions et des Systèmes (LSMIS), UMR 7140, Université de Strasbourg, CNRS, Strasbourg, France
| | - Yannis François
- Laboratoire de Spectrométrie de Masse des Interactions et des Systèmes (LSMIS), UMR 7140, Université de Strasbourg, CNRS, Strasbourg, France
| | - Davy Guillarme
- Biologics CMC and Developability, IRPF - Centre d’Immunologie Pierre-Fabre (CIPF), Saint-Julien-en-Genevois, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, IPHC UMR 7178, Université de Strasbourg, CNRS, Strasbourg, France
| |
Collapse
|