Monoclonal antibodies – definition, benefits & application

August 5, 2024
Approximately 160 Monoclonal antibodies or mAbs have been approved by regulatory agencies worldwide with 112 in the US and 114 in the EU.(1) These protein therapies are recognized as efficacious treatments for cancer and various ailments, such as migraines, multiple sclerosis, rheumatoid arthritis, and Crohn's disease.(2) But what exactly are monoclonal antibodies, and how are they characterized?

What are monoclonal antibodies? Brief definition and types
Monoclonal antibodies are laboratory-produced antibodies designed to mimic the immune system's ability to target pathogens.(2) Monoclonal antibodies contain a specialized binding site (Fab) responsible for the precise targeting of a particular antigen, a molecule inducing an immune response. Identification of this binding site, or epitope, is key to monoclonal antibody drug design.
Monoclonal antibodies are divided into four types according to the proportion of human amino acid sequences. Additionally, there are five immunoglobulin classes: IgG, IgA,IgD, IgE, and IgM, that are distinguished by their heavy chain. These proteins are notorious for undergoing post-translational modifications (PTMs) like glycosylation, which is critical to their efficacy.
How are monoclonal antibodies produced?
The production processes for biopharmaceuticals have evolved over time. Earlier methods focused on extracting and purifying natural sources. Contemporary methods are more precise, beginning with identifying, isolating, and expressing of the gene of interest.(3) The production process can be broken down into two stages: upstream and downstream, but before these steps and commercial manufacture, significant time is spent evaluating the developability, characterization and process development.
The upstream process spans approximately 2-3 weeks for a batch or fed-batch bioreactor, starting with a working cell bank, progressing to a seed train, then the inoculation phase, and finally production in a bioreactor. A typical bioreactor campaign will last ~2 weeks for a fed-batch or batch process or longer if continuous processing is deployed. Process engineers closely monitor the bioreactor to maintain cell viability of 70% or higher to maintain yield and minimize impurities released by deteriorated cells. During upstream production, some key product attributes are monitored, including viable cell count, relevant cell culture media components, titer, and critical process parameters like pH, evolved gases and temperature are closely monitored.
The downstream process focuses on the preparation of the final product. It starts with clarification, followed by virus activation, purification and polishing, viral clearance, concentrating the product, final formulation, sterile filtration, fill-finish, and finally, lot release. In between these steps, the samples are gathered and assessed in an analytical laboratory for titer, aggregation, purity, the presence of proteases, and identity verification. Verification of post-translation modifications (PTMs) like charge variants and glycosylation are of specific interest as they can significantly alter the efficacy and safety of the final product. Each facility differs what is monitored between each step with many if not all the attributes listed, executed in the QC labs, that focus on safety, efficacy, integrity, and purity.

What solutions does Waters | Wyatt offer in relation to monoclonal antibodies, and how can you benefit from them?
To produce effective monoclonal antibody therapies, it is necessary to precisely analyze their properties. The benefits of adding dynamic light scattering (DLS) and multi-angle light scattering (MALS) in conjunction with separation technologies, such as size exclusion chromatography (SEC) and field flow fractionation (FFF), can be broken down into the following main points:
- Comprehensive Characterization of Identity: SEC-MALS (DAWN™ MALS instrument) enables absolute determination of properties such as molar mass, size, and conformation, even for complex glycosylated monoclonal antibodies. It can also provide drug-antibody ratios for antibody-drug conjugates (ADC) and antibody-oligonucleotide conjugates (AOC).
- Enhanced Separation: FFF-MALS (Eclipse™ FFF Instrument with MALS) excels in characterizing larger protein aggregates and is well-suited for analyzing large complex samples like IgM.
- Formulation Development: High-throughput DLS (DynaPro™ Plate Reader) provides comprehensive stability screening, including aggregation and thermal stability, while Electrophoretic Light Scattering (ELS, DynaPro ZetaStar™ DLS Instrument) assesses molecular charge and turbidity.
- Viscosity and Interactions: Techniques like CG-MALS provide antibody-antigen binding kinetics and distinguish between attractive and repulsive interactions (A2, B22) impacting stability, while DLS assesses viscosity, essential for optimizing high-concentration formulations.
- Process Optimization: Light scattering measurements aid process development by determining purification endpoints in downstream processes (ultraDAWN™ MALS instrument) and facilitate high-throughput stability screening for optimal process conditions (DynaPro Plate Reader DLS).
- Aggregation and Particle Analysis: Light scattering techniques are recognized to provide robust analysis of aggregates and particle concentrations, crucial for regulatory filings.
In summary, light scattering technologies are indispensable for comprehensive characterization, facilitating biopharmaceutical development and regulatory approval.
References:
- Lyu X, Zhao Q, Hui J, Wang T, Lin M, Wang K, Zhang J, Shentu J, Dalby PA, Zhang H, Liu B. The global landscape of approved antibody therapies. Antib Ther. 2022 Sep 6;5(4):233-257. doi: 10.1093/abt/tbac021. PMID: 36213257; PMCID: PMC9535261
- Lu, RM., Hwang, YC., Liu, IJ. et al. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci 27, 1 (2020). https://doi.org/10.1186/s12929-019-0592-z
- Biopharmaceutical Processing: Development, Design and Implementation of Manufacturing Processes, Edited by Jagschies, Lindskog, Lacki, and Galliher. Copyright © 2018 Elsevier Ltd. All rights reserved. https://doi.org/10.1016/C2014-0-01092-1