At Elise Biopharma, we recognize that the sugar chains attached to your therapeutic proteins—known as glycans—are far more than decorations. They govern half-life, efficacy, immunogenicity, and even safety. Our Glycoengineering & Glycoform Optimization services transform host cells into precision machines, sculpting glycan profiles to your exact specifications. Whether you’re developing next-generation antibodies, enzyme replacements, or Fc-fusion decoys, our integrated glycosylation engineering platform ensures optimal glycoform distributions, consistent batch-to-batch quality, and a fast track to clinical success.
Why Glycoengineering Matters
- Extend Serum Half-Life
- Sialylation and terminal galactose residues engage with hepatic lectin receptors to delay clearance.
- Optimized Fc glycoforms exploit FcRn recycling for multi-week dosing intervals.
- Tune Effector Functions
- Afucosylation enhances ADCC by increasing affinity to FcγRIIIa on NK cells.
- Controlled bisecting N-acetylglucosamine (GlcNAc) modulates CDC and ADCP.
- Reduce Immunogenicity
- Human-like glycan patterns minimize anti-drug antibody (ADA) responses.
- Eliminate non-human structures (α-Gal, Neu5Gc) that trigger hypersensitivity.
- Improve Stability & Solubility
- Certain glycoforms shield hydrophobic patches, reducing aggregation.
- Glycan–protein interactions can stabilize tertiary structure under stress.
Our Glycoengineering CDMO Platform
Elise Biopharma’s proprietary platform integrates molecular design, cell line engineering, process development, and advanced analytics into a seamless glycoform optimization workflow:
Module | Capabilities |
---|---|
Host Cell Engineering | CHO, HEK293, Pichia pastoris with targeted knock-ins/knock-outs (GNTI, FUT8, ST3GAL, B4GALT) |
Vector & Expression | Glyco-optimized expression cassettes, signal peptide tuning, promoter selection for balanced expression |
Upstream Process Design | Media supplementation with Mn²⁺, uridine; feed strategy for glycan precursors; temperature and pH modulation |
Downstream Purification | Lectin affinity chromatography, mixed-mode resins for glycoform fractionation, size-exclusion polishing |
Analytical Characterization | LC-MS/MS glycopeptide mapping, HILIC-FLD glycan profiling, CE-SDS for charge variants |
Regulatory Support | CMC documentation for glycan critical quality attributes (CQAs), comparability protocols, IND/BLA filing support |
Host Cell Engineering & Strain Selection
3.1 Choosing the Right Host
- CHO Cells
- Gold standard for human-like N-glycans; robust growth and regulatory familiarity.
- HEK293 Cells
- Superior sialylation machinery; valuable for highly sialylated glycoforms.
- P. pastoris (Glycoengineered Strains)
- Rapid growth and simple media; custom strains programmed for human-type glycosylation.
3.2 Genetic Modifications
- Knock-Outs
- GNTI-KO: Produces high-mannose glycoforms for rapid clearance models or specific enzyme products.
- FUT8-KO: Eliminates core fucose for enhanced ADCC.
- Knock-Ins
- ST3GAL1/4, B4GALT1/2: Increase terminal sialylation and galactosylation.
- Human α-2,6-sialyltransferase to achieve human-specific sialic acid linkages.
3.3 Single-Cell Cloning & Screening
Using FACS and automated imaging, we isolate monoclonal cell lines with optimal growth, productivity, and glycoform consistency. Stability studies confirm > 90% desired glycoform retention over 60+ population doublings.
Upstream Process Development for Glycoform Control
4.1 Media Design & Feed Strategies
- Media Supplements:
- Manganese (Mn²⁺): Cofactor for galactosyltransferases.
- Uridine & Galactose: Precursor sugars for glycan extension.
- Feed Timing & Composition:
- Bolus vs. continuous feed to maintain steady‐state sugar levels.
- Custom feed blends to balance growth vs. glycosylation pathways.
4.2 Bioreactor Parameter Optimization
- pH Shifts:
- Slight alkaline shift (pH 7.2–7.4) favors galactosylation.
- Temperature Reduction:
- Lowering from 37 °C to 30–32 °C post-induction extends glycosyltransferase half-life.
- Dissolved Oxygen (DO):
- High DO (> 40% saturation) ensures energy for glycan biosynthesis.
4.3 High-Throughput DoE Screening
Using micro-bioreactors (AMBR®), we run factorial designs to pinpoint critical process parameters (CPPs) that drive glycoform distributions. Typical screening matrix:
Variable | Levels |
---|---|
Temperature (°C) | 30, 32, 34 |
pH Setpoint | 7.0, 7.2, 7.4 |
Mn²⁺ (µM) | 10, 20, 40 |
Feed Rate (mL/L/h) | 0.5, 1.0, 1.5 |
Downstream Purification & Glycoform Fractionation
5.1 Lectin Affinity Chromatography
- Concanavalin A (ConA): Captures high-mannose and hybrid glycans.
- Aleuria aurantia lectin (AAL): Binds fucosylated glycoforms for targeted removal.
5.2 Mixed-Mode & IEX Polishing
- Mixed-Mode Resins: Combine ionic and hydrophobic interactions to resolve closely related glycoforms.
- Cation/Anion Exchange: Separate charge variants arising from sialylation differences.
5.3 Size-Exclusion & HIC
- SEC: Final polishing to remove high-molecular-weight aggregates that can co-elute with certain glycoforms.
- HIC: Exploit subtle hydrophobicity changes introduced by glycan modifications.
Analytical Characterization & Quality Control
Assay | Purpose |
---|---|
LC-MS/MS Glycopeptide Mapping | Site-specific glycan composition |
HILIC-FLD Glycan Profiling | Quantitative glycan distribution (2AA labeling) |
CE-SDS Charge Variant Analysis | Detects sialylation and deamidation variants |
1D/2D-SDS-PAGE & Western Blot | Assess molecular weight shifts |
SPR/BLI FcRn Binding | Functional half-life correlate |
Cell-Based Fc Effector Assays | ADCC, CDC, and ADCP potency |
Stability Studies | Real-time & accelerated glycoform integrity |
Endotoxin & Host-Cell Protein (HCP) | Ensure safety and purity |
Each batch is benchmarked against pre-defined glycan critical quality attributes (CQAs) to guarantee consistency and regulatory compliance.
Regulatory & CMC Support
- Glycan CQA Definition: Establish glycoform ranges critical for safety and efficacy.
- Comparability Protocols: Demonstrate equivalence between R&D and GMP lots via head-to-head analytical studies.
- IND/BLA Filings: Comprehensive CMC documentation, including glycosylation pathways, CPP tables, and analytical methods validation.
- Global Compliance: Alignment with FDA, EMA, and ICH Q6B guidelines on glycan characterization.
Case Study: Afucosylated Antibody Enhancement
Challenge: A mid-stage oncology biotech needed an afucosylated monoclonal antibody to boost ADCC.
Solution Workflow:
- FUT8 Knock-Out in CHO-S host via CRISPR/Cas9.
- Screening & Cloning: 200 single clones evaluated; top line produced 4 g/L with < 5% fucosylation.
- Process Development: Optimized feed strategy with Mn²⁺ depletion post-peak to prevent residual fucosylation.
- Analytics: Achieved > 95% afucosylation confirmed by HILIC-FLD.
- GMP Manufacturing: Two 1,000 L runs delivered clinical-grade drug substance with consistent glycoform profiles.
Outcome: IND clearance in 8 months; first-in-human trial initiated on schedule.
Workflow: From Concept to Clinic
- Discovery & Feasibility (4–6 weeks)
- In silico glycosylation modeling and target glycoform specification.
- Small-scale expression screen and preliminary glycan analysis.
- Cell Line Generation (8–12 weeks)
- Host engineering, vector design, transfection, and single-cell cloning.
- Productivity and glycoform screening in multiwell plates.
- Process Definition (6–8 weeks)
- Micro-bioreactor DoE for CPP identification.
- Scale-up to bench-top bioreactors with optimized feed and control strategies.
- GMP Manufacturing (12–16 weeks)
- Technology transfer, facility qualification, and GMP campaign scheduling.
- Bulk drug substance production, downstream purification, and release testing.
- Regulatory Filing & Support (Concurrent)
- CMC dossier preparation, glycan CQA justifications, and assay validations.
- IND/BLA submission assistance and health authority interactions.
- Post-Launch Support
- Continuous process verification (CPV), comparability for lifecycle management, and process improvements.
(FAQs)
- What is the typical timeline for glycoengineering CDMO projects?
- From feasibility to IND-enabling GMP batches: ~6–9 months, depending on complexity.
- Can you customize glycoforms beyond common Fc glycan patterns?
- Yes. We engineer host pathways to produce hybrid, high-mannose, or novel glycoarchitectures on demand.
- How do you ensure batch-to-batch consistency?
- Rigorous CPP control, real-time analytics, and CPV frameworks guarantee < 5% glycoform variability.
- Are your processes cGMP-compliant?
- Absolutely. Our state-of-the-art facility follows FDA 21 CFR Part 211, EU GMP, and ICH Q6B guidelines.
- What analytical capabilities do you offer for glycan profiling?
- LC-MS/MS, HILIC-FLD, CE-SDS, SPR, cell-based assays, and more—as detailed above.
Ready to Elevate Your Glycoform Profiles?
Partner with Elise Biopharma—the premier glycoengineering CDMO—to harness the full power of glycosylation engineering.