What are protein therapeutics?

Protein therapeutics are a specific class of biotherapeutics and in their simplest form include genetically engineered versions of naturally occurring human proteins.  Protein therapeutics have several advantages over small-molecule drugs, providing the means to target highly specific, complex, and dynamic functions. Due to their specificity, protein-based therapies have less potential to cause adverse effects, are often well tolerated and are less likely to elicit immune responses.

 

Protein therapeutics such as monoclonal antibodies, biosimilars, bi-specifics, antibody-drug conjugates, and fusions proteins are made via recombinant DNA technology. Here recombinant proteins are produced in host cells, in which recombinant DNA, encoding the protein therapy, has previously been inserted thus instructing the host cells ribosomal machinery to express the desired recombinant protein-based therapeutic.

 

In comparison to chemical drug production, the protein-based therapeutic production process is complex, variable, and subject to elevated risk of product degradation and contamination. Process losses can be high, and the final product is fragile and can be negatively impacted by several factors including temperature, prolonged storage, denaturants, organic solvents, oxygen and changes in pH.

 

As a result, protein biologics require strict controls in manufacturing, transport and storage. An array of analytical approaches and workflows facilitate comprehensive characterization, allowing for full product understanding which can save significant time and development costs. Throughout process development and manufacturing, these analytical tools are required for reliable monitoring of product microheterogeneity ensuring drug safety and efficacy.

Consistent results for peptide mapping and monitoring across three systems of the Vanquish UHPLC platform

In collaboration with NIBRT.

 

Protein-based therapeutics workflows.


Intact mass analysis

Intact mass analysis is the assessment of the molecular weight of an intact protein sample using mass spectrometry (MS). Upon data processing, intact mass analysis allows for the determination of the average molecular mass or the monoisotopic mass of the protein of interest, depending on the resolving power of MS and the size of the protein. Typically, for large proteins such as monoclonal antibodies (mAbs), the average mass is obtained.

 

The observed mass can be compared to the expected theoretical mass for a given amino acid sequence to confirm protein identity, structural integrity, and presence and relative abundance of isoforms (e.g., glycoforms). Intact mass analysis provides information on antibody drug conjugates (ADCs), drug to antibody ratios (DAR), mAb sequence variations, impurities, and degradation products.

Intact protein mass analysis workflow using LC-MS.

Comparing biosimilars using intact mass analysis under denaturing and native conditions

Collaborator: NIBRT

Robust and reproducible peptide mapping and intact mass analysis workflows on a single instrument platform

Collaborator: NIBRT

Leaders in characterization

Collaborator: Bio-Techne Corporation

Simple, robust, high quality intact mass analysis—A biosimilars case study

Collaborator: NIBRT


Native intact analysis

Unlike in standard intact mass analysis, during native intact analysis, also known as native mass spectrometry (MS), protein analytes are sprayed from a non-denaturing solvent in a process called electrospray ionization. The term ‘native’ describes the biological status of the analytes in solution prior to the ionization event. It is imperative that conditions such as pH and ion source parameters are controlled to maintain the native folded state of the biologic protein.

 

Protein analysis in native or native-like conditions decreases charge state values, resulting in detection at higher m/z ranges with more spatial resolution. Native mass spectrometry conditions also allow the preservation of structurally critical non-covalent bonds, providing the mass of biomolecules that associate noncovalently. Furthermore, limited sample preparation is needed in exchange for a high level of information.

Native intact mass analysis workflow.

Comparing biosimilars using intact mass analysis under denaturing and native conditions

Collaborator: NIBRT

Seamless LC-MS method transfer in a biopharmaceutical development laboratory

Collaborator: Symphogen

Characterizing therapeutic proteins for drug discovery and development

Collaborator: LifeArc

Driving native mass spectrometry of membrane proteins for pharmaceutical research

Collaborator: Oxford Mass Technologies


Subunit analysis

Enzymatic digestion and reduction of monoclonal antibodies facilitates analysis at the subunit level. A commonly used approach utilizes the cysteine protease produced by S.pyogenes, known as IdeS, which is highly specific. IdeS digestion followed by reduction generates three subunits that are readily separated by reversed phase (RP) UHPLC.

 

Subunit analysis involves faster sample preparation, and simplified data interpretation compared to peptide mapping or bottom-up characterization approaches and provides simultaneous characterization of multiple attributes of antibodies at domain levels.

Subunit mass analysis workflow.

IdeS-cleaved mAb subunit analysis with LC-HRAM-MS: a quick and accurate comparison of biosimilar and originator biotherapeutics

Collaborator: NIBRT

Proton transfer charge reduction (PTCR) improves spectral matching and sequence coverage in middle-down analysis of monoclonal antibodies

Collaborator: University of Oklahoma

Subunits analysis approach for the determination of fucosylation levels in monoclonal antibodies using LC-HRAM-MS

Collaborator: NIBRT

Characterizing therapeutic proteins for drug discovery and development

Collaborator: LifeArc


Peptide mapping analysis

Peptide mapping provides a comprehensive and accurate representation of the landscape of a particular protein. With peptide mapping you create full sequence information, displaying each amino acid constituent and where they are in relation to each other. Peptide mapping should provide information on all possible post-translational modifications and sequence variants, illustrating the many possible instances in which a single protein molecule can be modified. The technique should be reproducible and reliable.

 

Peptide mapping involves the digestion of a therapeutic protein into its constituent peptides via a chemical or enzymatic reaction. Separation and subsequent identification of these peptides provides the details necessary to elucidate a protein’s full sequence information. Structural characterization at this level provides information on post translational modifications (PTMs) such as site-specific glycosylation, amino acid substitutions (sequence variants) and/or truncations.

 

Advances in protein sample preparation chemistries, ultra-high performance liquid chromatography (UHPLC), mass spectrometry (MS) hardware, and intuitive software facilitate the generation of comprehensive, confident peptide maps.

End-to-end peptide mapping solution for complete protein characterization.

Confident peptide mapping and disulfide bond analysis of an IgG2 monoclonal antibody

Collaborator: NIBRT

High-throughput peptide mapping of trastuzumab using a tandem LC-MS workflow

Collaborator: NIBRT

Peptide mapping of challenging monoclonal antibodies

Collaborator: Symphogen

Characterizing therapeutic proteins for drug discovery and development

Collaborator: LifeArc

See more resources


Charge variant analysis

Charge variant analysis is used to elicit the heterogeneity of charge variant forms or protein-based biologics.

 

Biotherapeutic proteins such as monoclonal antibodies (mAbs) are far more heterogeneous than small-molecule drugs. The presence of the charged state can significantly impact the structure, stability, binding affinity, and efficacy of the biotherapeutic drug. It is therefore necessary to understand the profile of the drug so that charge variants are identified and removed if necessary.

 

Charge variants of mAbs are due to modifications such as sialylation, deamidation, and C-terminal lysine truncation. Salt gradient cation-exchange chromatography has commonly been used with some success in characterizing mAb charge variants. However, significant effort is often required to tailor the salt gradient method for each individual mAb. In the fast-paced drug development environment, a quick and robust platform method is desirable to accommodate the majority of the mAb analyses. 

Charge variant analysis workflow.

Evaluation and application of salt- and pH-based ion-exchange chromatography gradients for analysis of therapeutic monoclonal antibodies

Collaborator: NIBRT

Simple charge variant profile comparison of an innovator monoclonal antibody and a biosimilar candidate

Collaborator: NIBRT

Bringing biosimilar therapeutics to market faster – A CRO case study

Collaborator: Sartorius Stedim BioOutsource Ltd.


Multi-Attribute Method (MAM)

What is the Multi-Attribute Method?

The Multi-Attribute Method (MAM) is a peptide mapping-based method which takes advantage of high-resolution mass spectrometric data. High-resolution mass spectrometry provides the accurate mass information aiding identification of product quality attributes and consequent quantitation and monitoring, which leads to a more comprehensive biomolecule process understanding, allowing for better process control for manufacturing.

 

Modern biopharmaceutical drugs, such as monoclonal antibodies (mAbs) and antibody drug conjugates (ADCs), are tremendously complex molecules manufactured inside living cells. Characterizing and monitoring these compounds in chemistry, manufacturing, and controls (CMC), and quality control environments can present a significant challenge, but the use of high-resolution accurate mass (HRAM) mass spectrometry offers a powerful way of overcoming these challenges.

 

The additional dimension of separation combined with the sensitivity and specificity of Orbitrap MS yields deeper product knowledge than a combined battery of conventional characterization tests, eliminating many traditional lot release tests.

 

By directly measuring critical quality attributes at the individual residue level, understanding how changes in process or production affect a drug product becomes part of a "quality by design" approach. Together, the results provided by HRAM MS enable safer and more potent drugs to be manufactured, delivering an incredibly powerful research-to-routine workflow.

Multi-attribute method workflow.

Deploying the multi-attribute method (MAM) across sites at Pfizer

Collaborator: Pfizer

Meet the expert: Rich Rogers discusses his vision of what the future may hold for the multi-attribute method (MAM) in the biopharmaceutical industry

Collaborator: Rich Rogers

Multi-Attribute Method (MAM) offers an ideal solution for development and release of safe and effective biotherapeutics

Collaborator: BATL


Aggregate analysis

Therapeutic protein aggregation is the process in which protein molecules assemble into stable complexes composed of two or more proteins. The manufacturing and purification processes used to make protein therapeutics typically yield high purity, single protein molecules called monomers. Following production, therapeutic protein monomers can form aggregates during storage and/or transportation. Protein aggregation in biotherapeutics can lead to incorrect drug dosage and/or undesired, potentially fatal immune responses in patients. Consequently, careful monitoring of protein aggregates is important for safety and quality assurance.

 

Investigators routinely use size-exclusion chromatography (SEC), which separates particles according to hydrodynamic size, in order to determine aggregation levels in therapeutic protein samples. Recently, the use of hydrophobic interaction chromatography (HIC) as an orthogonal technique to SEC has also been a topic of great interest. HIC separates proteins based on hydrophobicity in their native state and can often detect changes in protein structure as well as aggregation.

Robust, reproducible workflow for UHPLC-based protein aggregate monitoring.

Lifetime stability of size exclusion chromatography columns for protein aggregate analysis

Collaborator: NIBRT

A universal chromatography method for aggregate analysis of monoclonal antibodies

Collaborator: NIBRT

The importance of correct UHPLC instrument setup for protein aggregate analysis by size-exclusion chromatography

Collaborator: NIBRT

High-throughput protein aggregate analysis of monoclonal antibodies using a novel dual-channel UHPLC instrument

Collaborator: NIBRT


Host cell protein (HCP) analysis

Recombinant biotherapeutics are generally produced using non-human host cells, with Chinese hamster ovary (CHO), murine myeloma, and Escherichia coli cell-lines most commonly used. During cell growth and harvest, endogenous proteins from these host cells are released and can detrimentally affect final drug product safety and efficacy.

 

Purification of drug products from host cell protein contaminants can be challenging, with low-level contamination remaining after purification. The detection and quantification of residual HCPs as potential process-related impurities is critical for biopharmaceutical companies in accordance with regulatory agency guidelines (ICH Q6B).

Host cell protein analysis workflow.

Comprehensive identification and label-free quantitation of host cell protein contaminants using BioPharma Finder 4.1 software

Collaborator: NIBRT

Easy, fast and reproducible analysis of host cell protein (HCP) in monoclonal antibody preparations

Collaborator: NIBRT


Glycan analysis

Glycan analysis involves several approaches to investigate the sequence, structure, and location of carbohydrate molecules modifications on proteins or protein residues in biotherapeutics.

 

Glycans serve a variety of structural and functional roles in membrane and secreted proteins, with most proteins undergoing some degree of glycosylation during their synthesis.

 

Changes in the glycosylation pattern of protein biotherapeutics have been shown to impact their half-life, stability, safety, and efficacy. In general, glycans are divided into two main groups: O-linked and N-linked glycans. O-linked glycosylation involves the attachment of oligosaccharides to serine or threonine amino acid residues through an oxygen atom, and N-linked glycosylation involves the attachment of oligosaccharides to asparagine amino acid residues through a nitrogen atom.

 

More than 60% of therapeutic proteins are post translationally modified following biosynthesis by the addition of N- or O-linked glycans. Glycosylation of biotherapeutics can be influenced by a multitude of process related factors, such as pH, carbon source, dissolved oxygen, temperature during manufacture, and by the choice of expression system.

 

While glycosylation is the most common post-translational modification of proteins, it is also the most demanding from an analytical point of view. Biotherapeutic glycoproteins with complex glycosylation patterns have the potential to easily fall out of specification with changes in biomanufacturing processes. To meet regulatory demands such as ICH Q5E and ICH Q6B, manufacturers must carefully characterize glycosylation of proteins and its relation to the clinical activity of the therapeutic. The complete analysis of a glycoprotein provides information on the primary structure of the oligosaccharides as well as their variation at individual glycosylation sites.

Intact glycoprotein analysis workflow.

What is my glycosylation pattern?

Glycan structure determination workflow.

Which glycans are in my drug and what is their concentration?

Glycopeptide profiling workflow.

Where do my glycans reside?

Monosaccharide and sialic acid analysis workflow.

What is the composition of sugars in my drug entity?

Comprehensive protein glycosylation comparison of an innovator monoclonal antibody to a candidate biosimilar by HILIC UHPLC analysis

Collaborator: NIBRT

Evaluation of chromatographic phases for separation of differentially labeled glycans from erythropoietin and trastuzumab

Collaborator: NIBRT

Fast profiling of the N-glycan population in biotherapeutic antibodies by UHPLC-FLD with MS confirmation

Collaborator: NIBRT

Use of alternative chromatographic phases and LC-MS for characterization of N-glycans from NISTmAb RM 8671

Collaborator: NIBRT


Higher order structure analysis

Hydrogen deuterium exchange (HDX) mass spectrometry (MS) is a powerful tool for studying the dynamics of higher order structure of protein-based therapeutics. The rate of hydrogen-to-deuterium exchange within the amide hydrogen on the backbone of biotherapeutics provides solvent accessibility information, and thus protein structure and conformation can be inferred.

 

Full structural characterization is critical in biopharmaceutical development where the subtle but critical local conformational changes can impact safety and efficacy.

Bottom-up HDX workflow.

Intact/top-down HDX workflow.

Hydrogen deuterium exchange mass spectrometry for the masses

Hydrogen deuterium exchange mass spectrometry for protein structural characterization