webinar
Research Into Polysaccharide-Based Vaccine and Drug Delivery
Prof. Nikolai Petrovsky | Australian Respiratory and Sleep Medicine Institute
Register

Controlling Glycan Structure in Protein Glycosylation

Achieving control over glycan structure is no longer an exercise in downstream optimization—it is becoming a primary factor that determines whether a drug product acts like a single uniform molecule or a heterogeneous mixture of glycoforms with variable PK, function and immunogenicity potential. By defining sugar identity, linkage and branch location via enzyme editing, chemoenzymatic synthesis or glyco-engineering of expression platforms, drug developers can turn glycosylation into a designed feature that helps meet regulatory standards and support product distinctiveness.

Why Glycan Structure Control Is Critical

Precise control of glycan structure is essential since a single shift of one monosaccharide or inversion of an anomeric center can divert a therapeutic antibody from FcRn-mediated pro-longevity recycling towards asialoglycoprotein-receptor-mediated rapid clearance, or transform an anti-inflammatory IgG into an immune-complex activator that exacerbates cytokine release syndrome; without locked structure, downstream PK and immunogenicity are left to chance.

Chemoenzymatic synthesis of complex-type N-glycansFig. 1 Chemoenzymatic synthesis of complex-type N-glycans.1,5

Functional Impacts of Glycan Structural Variations

Alterations in glycan structure can drastically affect downstream protein-protein interactions. Small perturbations propagate allosteric forces throughout the glycoprotein-protein interface modifying binding kinetics and/or changing trafficking through the cell. Terminal sialic acid groups can conceal recognition motifs, allowing evasion of clearance through asialoglycoprotein receptors on hepatocytes, extending plasma half-life. Deglycosylation of sialic acid residues exposes a recognition motif for galactose-specific phagocytic receptors, which results in accelerated clearance from the bloodstream and necessitates more frequent dosing. The addition or removal of core fucose modifies affinity for FcγRIIIa and alters ADCC activity. Addition of core fucose sterically blocks interaction with FcγRIIIa and decreases ADCC. Removal of fucose flips the Fc domain into a high-affinity conformation for binding to FcγRIIIa. This increased ADCC can be beneficial in cancer therapies, but detrimental if treating autoimmune diseases. Addition of bisecting GlcNAc causes a conformational shift that decreases protein flexibility and protects against degradation by endoglycosidases. Removal of bisecting GlcNAc creates a flexible glycan branch that can be trimmed by glycosidases rapidly lowering protein half-life. Clusters of mannose residues are recognized by mannose receptors on macrophages draining the lymph node into which they were injected, significantly lowering circulation half-life to less than 24 hours. Glycosylation can also influence protein folding efficiency within the ER. An oligomannose-type glycan serves as a recognition motif for binding to chaperone proteins calnexin/calreticulin, allowing time for proper folding of the aglycone. Complex type glycans function as a sorting signal that dissociates from calnexin/calreticulin, allowing transport to the Golgi apparatus. Overproduction of improperly folded glycoforms can lead to protein aggregation if co-purified with drug substance. Sialic acid contains negatively charged groups. Addition of sialic acid creates a patch of negative charge on the protein surface that may help prevent nonspecific interactions with container surfaces.

Table 1 Glycan architectural motifs and their pharmacological echo

Motif alterationFunctional echoClinical ramificationAnalytical sentinel
Loss of terminal sialic acidFaster ASGPR uptakeShorter half-lifeIon-pair LC–MS
Absence of core fucoseHigher FcγRIIIa affinityPotentiated ADCCMiddle-up LC–MS
Addition of bisecting GlcNAcEndoglycosidase resistanceExtended exposureHILIC-FLR
High-mannose prevalenceMannose-receptor clearanceRapid systemic lossCapillary electrophoresis
Non-human α-Gal epitopeAnti-drug antibody riskNeutralizationMALDI-TOF with antibody overlay

Relevance to Therapeutic and Research Proteins

Biopharmaceutical proteins are under glycan editing regulations enforced by Pharmacopoeias. A lack of desired glycans or heterogeneity in glycan profiles can result in loss of potency or product rejection from regulatory agencies. Monoclonal antibodies (mAbs) are the most prevalent therapeutic protein. Desirability of glycoforms for mAbs depend on its intended use; cancer therapeutics favor molecules with greater antibody-dependent cellular cytotoxicity (ADCC) while immunosuppressants require lower ADCC. Regulatory agencies require sponsors to define acceptance criteria of afucosylation, galactosylation, and sialylation levels with accompanying validated assays that link glycan % abundance to activity. For example, erythropoietin with low levels of sialic acid will have reduced half-life in the blood stream. Patients require higher doses to achieve the same effect which increases both COGs and pill burden. Too much sialylation can decrease efficacy by overly camouflaging the protein. Factor VIII activity relies on tyrosine-sulphated O-glycans for secretion. Without these glycans Factor VIII cannot be trafficked out of the ER leading to low overall titers. Biosimilar manufacturers must match glycans of the innovator product. If the overall glycan profile looks similar, but the galactosylation Peak is right-shifted, regulatory agencies may ask for PK bridging studies, delaying time to market. Glycosylation of research proteins can also lead to misinterpretation of results. Structural biologists see poor crystals if heterogeneity in glycans prevents the protein from crystalizing. Glycan heterogeneity seen in cell-based assays between runs will cause cell biologists to see different signaling events if glycan structure alters internalization rates.

Factors Influencing Glycan Structure

Synthesis of glycans relies on a template-less biosynthetic machinery; the net activities of glycosyl-transferases, glycosidases, nucleotide-sugar transporters and spatial orientation of the peptide dictate glycan structure. These factors exist in fluid membranous environments; thus, changes in enzyme levels/distribution, availability of sugar donors, and folding of the protein can divert the glycosylation pathway from production of high mannose glycans to production of immunogenic hybrid-type glycans. Therefore, controlling these factors are very important.

Biosynthetic Pathways and Enzyme Specificity

N-linked synthesis begins in the ER with a preformed oligosaccharide donor transferred en bloc to an asparagine side-chain. In contrast, O-linked synthesis begins in later Golgi compartments and proceeds sugar-by-sugar without a template. Therefore, O-linkage responds greatly to competition between locally available glycosyltransferases at that compartment. Individual glycosyltransferases preferentially use certain donor anomeric configuration, arm configurations and acceptor substrates; should two enzymes overlap in specificity, they will compete for usage of a particular sugar-linkage resulting in a random mixture weighted by their relative expression rather than a programmed addition. Competition is further affected by availability of substrates: CMP-sialic acid at low concentrations can limit sialyltransferases but not galactosyltransferases which results in increased galactose on the glycan antenna and reduces its circulatory lifespan. Enzymes often recognize alternative substrates when their 'primary' nucleotide-sugar is depleted, leading to incorporation of uncommon sugars that may not be picked up by standard LC–MS methods but can modulate binding to receptors. Likewise, localization of glycosyltransferases within Golgi cisternae provides an additional layer of selectivity: a 'late' glycosyltransferase that is artificially increased in an 'early' compartment will encounter incomplete glycans and add its sugar truncating many downstream structures. Changes in pH through the Golgi can also affect enzyme activity: slight acidification can protonate the catalytic aspartate of one enzyme but not another, biasing micro-heterogeneity. For bacteria, phase-variable expression of glycosyltransferases provides 'switch' loci that turn on and off entire suites of enzymes at high frequency, producing populations of glycoforms even in a clonal culture.

Protein Sequence and Glycosylation Sites

The polypeptide substrate itself filters which Asn or Ser residues become glycosylated through rules of primary sequence consensus, secondary structure accessibility, and tertiary flexibility. An N-X-S/T sequon is required, but not sufficient, as secondary modifications such as proline-induced backbone rigidity at X+1 sterically prevents OTase binding even at an optimal sequon. Adjacent cysteines that are available for disulfide bond formation can occlude or stabilize the Gly-X-Ser motif depending on the redox environment. Accessibility to flexible loop regions with Ser/Thr residues facing the solvent will also dictate modification, while residues deep within beta sheets will not be modified even if they are located at an optimal sequon with favorable surrounding basic residues. Positive charge clustering around the modification site can increase occupancy as it will bring the negatively charged sugar donor closer to the site. Negative charge clustering will decrease occupancy as it will push away the sugar donor. Glycosylation sites which experience higher levels of dynamics (root-mean-square-fluctuation) are more likely to be glycosylated and conversely sites that reside in stable regions are out competed for glycosylation by neighboring sites that have higher dynamics. Sites can also be affected by mutations that are not near the modification site. Changing a single residue that is located far away from the glycosylation site can alter protein structure and expose a new glycosylation site.

Chemo-enzymatic approach to prepare homogeneous antibodiesFig. 2 Chemo-enzymatic approach to prepare homogeneous antibodies.2,5

Strategies for Glycan Structure Control

Modification of glycan structure follows a two-prong playbook: anchoring the addition of sugars to genomically encoded addresses and modifying glycans after their biosynthesis using sequence-defined enzymes that trim, replace or add onto existing glycans; these approaches transform cellular diversity into a titratable phenotype akin to any modular operation.

Site-Specific Glycosylation Approaches

Rewriting of the underlying protein sequence itself provides orthogonal means to address occupancy at desired sites. Asn-X-Ser/Thr sequons can be added or removed via site-directed mutagenesis so glycosylation occurs only at desired addresses. Residues flanking the sequon can be mutated to modulate local loop entropy: insertion of a proline at position X+1 reduces backbone flexibility and sterically inhibits OST binding quenching an otherwise active site. Expanding on this idea, insertion of flexible glycine-rich residues increases conformational entropy which increases local occupancy. Alternatively, silent codons can be mutated to incorporate cysteine residues which can be chemically or enzymatically converted to form glycosylation acceptor sites. This "tag-and-glycosylate" strategy allows for glycosylation to occur independent of the cell's native machinery. Occupancy can also be increased by preventing further modifications to the glycan after it has been added. Fusion of short retention signals are known to sequester proteins within early Golgi compartments preventing access of processing enzymes and resulting in high-mannose glycans. Competition from cryptic sequons elsewhere in the protein can be reduced using CRISPR base-editing to selectively mutate additional addresses. This redistribution of donor substrate funnels it toward the desired address and increases occupancy at that specific site. To demonstrate that glycosylation occurs at the desired address, an isotopically labeled amino acid can be added adjacent to the glycosylation site and later identified by LC–MS/MS analysis as a sort of barcode.

Enzymatic Remodeling and Extension

Remodeling enzymes then remove sugars and install defined replacements into the resulting glycoprotein, analogous to trimming and restuccoing a sponge cake. IgG-specific EndoS endoglycosidases release sugar chain heterogeneity by clipping off entire antennae between two N-acetylglucosamine residues (GlcNAcs) of the glycan core, yielding a uniform stub terminating in a GlcNAc acceptor substrate for any desired transglycosylation. Modified versions of these enzymes (wild-type endoglycosidases tend to hydrolyze sugars) known as "glycosynthases" can be used to load homogeneous sugars that have been activated as unstable oxazolines or electrophilic glycosyl fluorides. Sugar oxazolines or fluorides donated by these activated donors provide galactose β1-4-GlcNAc (lactosamine) repeats with defined terminal modifications including sialic acid, afucosylation, or bisecting GlcNAc. Mutant EndoS glycosynthases have been engineered to efficiently accept oligosaccharides activated as aldonolactones, thus bypassing altogether the sugar oxazoline step under acidic conditions. Reaction conditions are chosen to preserve the native fold of the antibody protein while enabling transglycosylation by adding the sugar donor typically in slight excess to shift the reaction equilibrium towards complete remodeling. If necessary, the size of heterogenous glycans can be reduced with other endoglycosidases prior to transglycosylation to step-wise build larger glycans. By repeating this trimming and rebuilding process with a set of orthogonal enzymes, sugars of arbitrary complexity can be trimmed away and rebuilt with single residue control.

in vitro and Chemoenzymatic Solutions

Both in vitro and chemoenzymatic approaches decouple glycosylation from cellular logistical constraints by using purified enzymes and synthetic donors in stoichiometric ratios; this allows gram-scale production of homogeneous glycoforms without host cell glycan heterogeneity, regulatory concerns about adventitious agents, or metabolic interactions that can obscure structure–function relationships.

Advantages of Cell-Free Glycosylation Systems

Cell-free media are defined, and the lack of membranes enables continuous titration of nucleotide-sugar donors, pH, and metal cofactors so that each reaction cycle can be recorded and reproduced. There are no competing biosynthetic pathways to divert expensive donors away from the desired glycosidic bond, improving atom economy and reducing purification burden. Reactions volumes can be easily amplified linearly because mass-transfer effects associated with whole organelles are supplanted by turbulence in a well-mixed solution; increasing reaction volume 500-fold maintains identical kcat/Km observed during microliter-scale experimentation, reducing the scale-up period from months to days. The reactions are run in open vessels allowing for in situ analytics—inline Raman spectroscopy or micro-pH electrodes can be used to control PID loops that adjust for slowly changing boundary conditions before product heterogeneity has a chance to develop, which would be impossible within the confines of an intact Golgi. Enzyme mixtures can also be replaced via tangential-flow ultrafiltration, allowing for catalyst deactivation to be addressed by simple diafiltration rather than time-consuming cell-line re-development. Additionally, since there is no host organism, there is no concern for endotoxin, viral or adventitious-agent, and host-cell protein contamination, allowing for a reduced downstream toxicology burden and shifts regulatory risk to chemistry-centric development.

Table 2 Comparative advantage matrix: cell-free vs cellular systems

ParameterCell-free platformCellular platformFunctional leverage
Donor stoichiometryUser-definedMetabolically competedCost control
Volume scalabilityLinearMembrane-limitedSpeed to gram
Real-time correctionInline probesGolgi-inaccessibleVariance compression
Catalyst refreshDiafiltrationRe-clone / re-transfectTimeline saving
Adventitious burdenNoneViral / endotoxinTox package slim
Glycoform scopeAny synthetic donorHost enzymatic repertoireStructure freedom

Combining Chemical and Enzymatic Methods

Chemical synthesis and enzymatic glycosylation are often combined in what are known as chemoenzymatic methods. Synthetic chemistry is used to prepare activated sugars (often high-energy oxazolines or fluorides) with non-natural, chemoselective handles such as azido groups for click chemistry, photolabile groups for reversible protection, and alkyne or acetal groups for chemoselective ligation. These activated sugars can then be transferred to proteins by engineered glycosyltransferases or endoglycosidases. Such chemoenzymatic methods have been used to chemically ligate complex glycans onto proteins in a single, aqueous step without the need for orthogonal protection strategies required for chemical glycan synthesis. If the desired glycan is too complex to be prepared enzymatically, the glycan portion can be chemically prepared with a small disaccharide handle. Glycosyltransferases can then be used to enzymatically grow the glycan antenna from this handle. Synthetic chemistry steps can be performed on resin allowing purification of the donor by filtration prior to enzymatic transfer. This is advantageous since often conditions required for chemical synthesis are toxic to enzymes. When a glycoprotein has been trimmed enzymatically to a homogeneous GlcNAc stub, any protein can be provided with a common handle for subsequent chemical modification.

Analytical Tools for Glycan Structure Verification

Structure confirmation of glycans requires an orthogonal tool set consisting of high resolution MS, separation and statistical matching; this should provide evidence for all linkage points, anomeric positions, and occupancy on each linkage point with a certainty beyond the analytical instrument/system. Only when structure data at all these levels agrees can software vendors confirm that a synthetic or biologically derived glycoform is the same from batch to batch.

Structural Characterization of Glycans

Released glycans are fluorescently or isotopically labelled prior to LC-MS for ionisation equalisation. They are then separated on porous graphitised carbon (PGC) LC-MS which can resolve α2,3- from α2,6-sialylation or β1,3- from β1,4-galactose linkages. Ion-mobility spectrometry inserts an additional orthogonal gas-phase shape metric that provides collision-cross-section values to discriminate isobaric isomers without chemical derivatisation. When permethylation precedes MALDI-TOF it both stabilises labile sialic acids and equalises mass-response factors, allowing relative peak areas to be reported as molar ratios without external calibration. Software such as GlycoWorkbench or SimGlycan can automatically score observed fragments against theoretical B/Y and C/Z ion libraries and rank candidates by number of matching fragment ions and mass error. Transferring this same workflow to automated 96-well microplates allows 100's of samples to be run per day, enabling glycan mapping to become high-throughput QC compatible with Quality by Design initiatives. Multivariate statistics (PCA, OPLS-DA) can reduce complex peak tables to single-score vectors which collapse lot-to-lot similarity into one overlay plot, streamlining regulatory justification of process drift or biosimilar comparability.

Site-Specific Glycosylation Analysis

Site-specific occupancy determines which Asn or Ser residue is modified by which glycoform. This information can be obtained through middle-down or bottom-up proteomics techniques which preserve the glycopeptide bond during fragmentation. Typically, controlled proteolysis is performed on a glycoprotein digest using Glu-C or trypsin at low pH and low temperature to prevent glycan loss prior to enrichment of glycopeptides by zwitterionic hydrophilic interaction chromatography (HILIC) or lectin weak-affinity chromatography to sufficiently concentrate the otherwise low-abundance glycopeptides that are lost in bulk protein signal. These glycopeptides can then be separated via nano-flow LC–MS/MS. Stepped-energy collision will fragment both the peptide backbone and glycan, producing oxonium ions in the same acquisition. Spectra are then searched against databases like GlycoSpectrumScan or SimGlycan, which create theoretical m/z values for all possible glycopeptide combinations, resulting in a list of identified glycopeptides ranked by probability. Additionally, to break any remaining isobaric overlaps, electron-transfer dissociation (ETD) fragmentation can be used. ETD leaves the glycan intact and fragments the peptide. The resulting c- and z-ion ladder allow the user to determine which residue was modified to within one amino acid. The parallel acquisition of glycan oxonium ions such as m/z 204 for HexNAc or 366 for Hex-HexNAc can rapidly identify glycan class without complete de novo sequencing. Finally, by isobarically tagging glycans with a quaternary amine label, site-specific glycosylation occupancy can be quantified across bioreactor feeds to track changes over time and potentially catch upstream deviations before they are observed in bulk assays.

When Structural Control Requires Custom Solutions

Developers reach a point where cell-culture glycosylation by routine means can't provide the atomic-level control, non-human glycans or batch-to-batch consistency regulators demand. This point is reached when your target glycoprotein needs to be sulfated-Lewis-x, contain C-glycosidic linkers or dual-site antibody signatures (afucosylated at one Fc, hyper-sialylated at the other) that aren't found in CHO supernatants. When you get here, bespoke chemoenzymatic methods are your only realistic path to a patentable molecule, fileable as a single LC-MS peak and manufacturable at scale without tweaking every new bioreactor run.

Complex or High-Precision Glycosylation Projects

Indications where α2,6-only sialylation is required for anti-inflammatory signaling; click-enabled glycans needed for downstream PEGylation; multispecifics that need their two Fc moieties to bear distinct glyco-signatures (such as afucosylated vs. sialylated for eff/longevity balance); Biosimilars that need to shoehorn within sub-% of an originator glycotope... all of these processes are beyond what can be dialed into most cell lines (even the most engineered of them). The examples above each represent a situation where either the homogeneous nature of cell line biology imposes a bottleneck, or regulatory/legal concerns require a specific glyco-footprint be matched and repeatedly met. When two Fcs on the same molecule need distinct glycosylation patterns (one afucosylated for maximum ADCC, the other sialylated for optimal half-life), traditional mammalian expression isn't possible since both polypeptides pass through the same Golgi apparatus. When a biologic needs to exactly match a competitor's glycosylation profile year after year, inherent kit-to-kit variability (i.e. "antenna drift") that may be tolerated for a trailblazing small molecule becomes critical. And when vaccines must retain activity after months/year of room-temp storage as a dry powder, chemically heterogeneous cell extracts are less desirable than homogeneous glyco-conjugates. In each case the game plan becomes either allocate financial resources towards building/chScaling your own chemoenzymatic toolkit; or contract a company that already has validated enzymes, rare sugars, and GMP ready analytics.

Partnering for Advanced Glycan Control

Contractors supply projects with pre-qualified glycosynthase collections, validated UDP-sugar libraries and LC-MS glyco-profile packages that are already compliant with ICH Q11 requirements for "well-characterized drug substances." In effect, this reduces years of internal reagent procurement and assay development timelines to months, while also furnishing viral-clearance packages and material-traceability documentation that clients can provide to auditors "as-is". Since the vendor aggregates anonymized batch records from multiple projects, they can also provide data-backed recommendations on which glycoform distribution maximizes half-life or which PNGS architecture is least likely to trigger an IND hold—knowledge that would otherwise cost hundreds of thousands of dollars and months of time to acquire from scratch. And if something does go wrong (e.g. a late-stage comparability batch fails due to unanticipated glyco-drift), the vendor accepts the analytical liability: they simply re-work the batch at no extra expense to the sponsor. Across all these dimensions, partnering with a custom glycosylation provider mitigates risk and translates into real-life benefits for clients: accelerated IND packages, more straightforward regulatory filings and increased chances of approval for Priority/Morediseases programs where glycan fidelity is mandatory.

Protein Glycosylation Services for Precise Glycan Structure Control

Controlling glycan structure in protein glycosylation requires coordinated control over glycosylation sites, enzyme specificity, and post-glycosylation processing. When native expression systems fail to deliver defined glycan structures, specialized protein glycosylation and engineering services provide the precision needed for structural and functional clarity.

Protein Glycosylation Services

Protein glycosylation services enable controlled attachment and modification of glycans on target proteins using enzymatic, in vitro, and chemoenzymatic strategies. By focusing on site-specific glycosylation and defined reaction conditions, these services help minimize unwanted structural variability and generate glycoproteins with consistent glycan architectures. Such approaches are particularly valuable for studies where subtle glycan structural differences have measurable effects on protein stability, activity, or biological interactions.

Glycan Engineering Services

Glycan engineering services extend beyond initial glycosylation to fine-tune glycan structures through controlled remodeling, extension, or modification. By selectively adding, removing, or modifying monosaccharide units, glycan engineering enables precise control over branching patterns, terminal residues, and functional glycan motifs. These services are especially useful for advanced protein engineering projects, including antibody optimization, structure-function studies, and applications requiring reproducible and well-defined glycan structures.

References

  1. Zhao X, Huang Y, Zhou S, et al. Recent chemical and chemoenzymatic strategies to complex-type N-glycans[J]. Frontiers in Chemistry, 2022, 10: 880128. https://doi.org/10.3389/fchem.2022.880128.
  2. Chao Q, Ding Y, Chen Z H, et al. Recent progress in chemo-enzymatic methods for the synthesis of N-glycans[J]. Frontiers in chemistry, 2020, 8: 513. https://doi.org/10.3389/fchem.2020.00513.
  3. Satoh T, Yamaguchi T, Kato K. Emerging structural insights into glycoprotein quality control coupled with N-glycan processing in the endoplasmic reticulum[J]. Molecules, 2015, 20(2): 2475-2491. https://doi.org/10.3390/molecules20022475.
  4. Edwards E, Livanos M, Krueger A, et al. Strategies to control therapeutic antibody glycosylation during bioprocessing: Synthesis and separation[J]. Biotechnology and bioengineering, 2022, 119(6): 1343-1358. https://doi.org/10.1002/bit.28066.
  5. Distributed under Open Access license CC BY 4.0, without modification.
* Only for research. Not suitable for any diagnostic or therapeutic use.
Send Inquiry
Verification code