GAA Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Function

The GAA recombinant monoclonal antibody is a laboratory-generated molecule that combines high specificity for the GAA protein with recombinant production methods. Unlike conventional monoclonal antibodies derived from hybridoma cells, this antibody is synthesized in vitro using phage display technology or similar platforms . Its primary function is to detect and quantify GAA protein levels in human tissues or samples, aiding in studies related to glycogen storage disorders like Pompe disease .

Production Process

The antibody is manufactured through a multi-step process:

  1. Gene Extraction: GAA-specific antibody genes are isolated from rabbit B cells .

  2. Cloning: These genes are amplified and inserted into phage vectors, which are then introduced into mammalian cell lines for protein expression .

  3. Purification: Affinity chromatography is used to isolate the antibody, ensuring high purity and activity .

ParameterDetail
HostMammalian cell lines
ClonalityMonoclonal
Purification MethodAffinity chromatography
ApplicationsIHC, ELISA

Immunohistochemistry (IHC)

The antibody is validated for IHC, with recommended dilutions ranging from 1:50 to 1:200 . It enables precise localization of GAA within lysosomes or extracellular compartments in tissue samples.

Enzyme-Linked Immunosorbent Assay (ELISA)

In ELISA, the antibody detects recombinant GAA with high sensitivity, making it suitable for quantifying enzyme levels in biological fluids or cell lysates .

Comparative Analysis of GAA Antibodies

While multiple GAA antibodies exist, the recombinant monoclonal variant distinguishes itself through:

FeatureRecombinant Monoclonal AntibodyConventional Monoclonal Antibodies
ProductionIn vitro synthesisHybridoma cell culture
SpecificityEngineered for high affinityVariable based on hybridoma selection
PurityHigh (affinity chromatography)Moderate to high
ReactivityHuman GAAHuman GAA (e.g., mouse/rabbit origin)

Research Findings and Limitations

  • Sensitivity: Detects recombinant GAA at concentrations as low as 0.3 ng/mL in ELISA .

  • Cross-reactivity: Specific to human GAA, with no reported cross-reactivity with non-human orthologs .

  • Limitations: Primarily used in research; not validated for therapeutic or diagnostic use in humans .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The GAA recombinant monoclonal antibody is produced through an in vitro synthetic process. It begins with the extraction of GAA antibody genes from B cells isolated from immunoreactive rabbits. These genes are then amplified and cloned into suitable phage vectors. The vectors are subsequently introduced into mammalian cell lines, enabling the production of functional antibodies. Following this, the GAA recombinant monoclonal antibody undergoes affinity chromatography purification. This antibody exhibits excellent performance in the detection of human GAA protein in ELISA and IHC applications.

Lysosomal alpha-glucosidase (GAA) is an indispensable enzyme involved in the breakdown of glycogen within lysosomes. Its primary role is to hydrolyze glycogen into glucose, thereby contributing to glucose homeostasis and preventing glycogen accumulation in various tissues. Dysfunction or deficiency of this enzyme can lead to severe health conditions, such as Pompe disease.

Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. The delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery time information.
Synonyms
Lysosomal alpha-glucosidase (EC 3.2.1.20) (Acid maltase) (Aglucosidase alfa) [Cleaved into: 76 kDa lysosomal alpha-glucosidase, 70 kDa lysosomal alpha-glucosidase], GAA
Target Names
GAA
Uniprot No.

Target Background

Function

Essential for the degradation of glycogen in lysosomes. Exhibits the highest activity on alpha-1,4-linked glycosidic linkages, but can also hydrolyze alpha-1,6-linked glucans.

Gene References Into Functions
  1. PI-rhGAA may have the potential to be a useful therapeutic option for improving the treatment of Pompe disease. PMID: 29102549
  2. The most prevalent mutation in Pompe disease is c.-32-13T, G. PMID: 29181627
  3. The narrow substrate-binding pocket of rhGAA is located near the C-terminal ends of beta-strands of the catalytic (beta/alpha)8 domain. It is shaped by a loop from the N-terminal beta-sheet domain and both inserts I and II. PMID: 29061980
  4. This is the first study of rhGAA to differentiate M6P glycans and identify their attachment sites, despite rhGAA being an approved drug for Pompe disease. PMID: 29274340
  5. GAA mutations are associated with Pompe disease. PMID: 28763149
  6. Enzyme activities (acid alpha-glucosidase (GAA), galactocerebrosidase (GALC), glucocerebrosidase (GBA), alpha-galactosidase A (GLA), alpha-iduronidase (IDUA) and sphingomyeline phosphodiesterase-1 (SMPD-1)) were measured on approximately 43,000 de-identified dried blood spot (DBS) punches. Screen positive samples were submitted for DNA sequencing to obtain genotype confirmation of disease risk. PMID: 27238910
  7. Enzyme replacement therapy (ERT) (alglucosidase alfa) stabilizes respiratory function and enhances mobility and muscle strength in late-onset Pompe disease. Lysosomal glycogen in muscle biopsies from treatment-naive LOPD patients was reduced following ERT (alglucosidase alfa). PMID: 27473031
  8. In adults with Pompe disease, antibody formation does not hinder rhGAA efficacy in the majority of patients. It is associated with IARs and may be mitigated by the IVS1/delex18 GAA genotype. PMID: 27362911
  9. Reanalysis of the patient's DNA sample using next generation sequencing (NGS) of a panel of target genes causing glycogen storage disorders demonstrated compound heterozygosity for a point mutation and an exonic deletion in the GAA gene. PMID: 28657663
  10. This study identified thirteen novel and two common GAA mutations. The allelic frequency of c.2662G > T (p.Glu888X) was 23.1% in northern Chinese patients and 4.2% in southern Chinese patients, whereas the allelic frequency of c.1935C >A (p.Asp645Glu) was 20.8% in southern and 3.8% in northern Chinese patients. PMID: 28394184
  11. This is the first report of the alpha-glucosidase inhibitory activity of compounds 20, 26, and 29. The findings support the significant role of Eremanthus species as novel sources of new drugs and/or herbal remedies for the treatment of type 2 diabetes. PMID: 27322221
  12. Compared with controls, GAA gene expression levels in coronary artery disease (CAD) patients were significantly elevated, suggesting that GAA may play a role in CAD development. PMID: 26580301
  13. The study reports on the clinical, biochemical, morphological, muscle imaging, and genetic findings of six adult Pompe patients from five unrelated families with the c.-32-13T>G GAA gene mutation in a homozygous state. All patients exhibited decreased GAA activity and elevated creatine kinase levels. PMID: 26231297
  14. Glycogen storage disease type II is caused by a deficiency of GAA activity resulting from mutation of the GAA gene. PMID: 26575883
  15. RT-PCR followed by DNA sequence analysis of patients with Pompe disease revealed a new variant in the GAA gene, leading to an aberrant splicing event. PMID: 25243733
  16. Findings indicate that the GAA c.2238G > C (p.W746C) novel mutation is the most common mutation in mainland Chinese late-onset Pompe patients, as observed in Taiwanese patients, expanding the genetic spectrum of the disease. PMID: 25526786
  17. This study reveals several alterations distributed along the GAA gene in a sample of Brazilian families. PMID: 25681614
  18. Mutations in the acid alpha-glucosidase gene are associated with Pompe disease. PMID: 25026126
  19. GAA deficiency results in reduced mTORC1 activation, which is partly responsible for the skeletal muscle wasting phenotype and can be ameliorated by leucine supplementation. PMID: 25231351
  20. The phenotype LO-GSDII with a GAA mutation in the North of Italy appears not significantly different from other LO-GSDII populations in Europe or the USA. PMID: 24158270
  21. Data showcases the largest informative family with late-onset Pompe disease described in the literature, exhibiting a peculiar complex set of mutations of the GAA gene. This may partially elucidate the clinical heterogeneity of this family. PMID: 24107549
  22. 7 of 27 in: Gene. 2014 Mar 1;537(1) Novel GAA sequence variant c.1211 A>G reduces enzyme activity but not protein expression in infantile and adult onset Pompe disease. PMID: 24384324
  23. This study demonstrates that the c.-32-13T>G mutation of the GAA gene abrogates the binding of the splicing factor U2AF65 to the polypyrimidine tract of exon 2, and several splicing factors affect exon 2 inclusion. PMID: 24150945
  24. The study describes two unrelated cases affected with classical early-onset Pompe disease, both originating from the same small Mexican region, with the same novel homozygous frameshift mutation at gene GAA (c.1987delC). PMID: 24399866
  25. Mutations in the GAA gene are associated with glycogen storage disease type II. PMID: 23884227
  26. Adult patients with alpha-glucosidase mutations other than c.-32-13 T>G can have very low alpha-glucosidase activity in fibroblasts but express higher activity in muscle and store less glycogen in muscle than patients with infantile Pompe disease. PMID: 23000108
  27. The study provided an update of the Pompe disease mutation database with 60 novel GAA sequence variants and additional studies on the functional effect of 34 previously reported variants. PMID: 22644586
  28. Transcriptional response to GAA deficiency (Pompe disease) in infantile-onset patients. PMID: 22658377
  29. The report details genetic testing to identify GAA mutations in German patients with late-onset glycogen storage disease type II. PMID: 18607768
  30. This study defines a critical role for endoplasmic reticulum stress in the activation of autophagy due to the 546G>T acid alpha glucosidase mutation. PMID: 21982629
  31. No common mutation is found in association with low levels of acid alpha-glucosidase activity in late-onset Pompe disease. Most patients produce unprocessed forms of GAA protein compared with patients who have higher GAA activity. PMID: 21484825
  32. Mutation analysis of the GAA gene revealed the p.D645E in all patients with Pompe disease, suggesting it as the most common mutation in the Thai population. PMID: 21039225
  33. The enzymatic screening of Pompe disease can be justified in patients with myopathies of unknown etiology, as demonstrated in this report of a Mexican patient with late-onset glycogen-storage disease type 2. PMID: 20350966
  34. Data show that the p.R1147G missense mutation impaired glucosidase activity. PMID: 19834502
  35. Homozygosity for multiple contiguous single-nucleotide polymorphisms as an indicator of large heterozygous deletions: identification of a novel heterozygous 8-kb intragenic deletion (IVS7-19 to IVS15-17) in a patient with glycogen storage disease type II. PMID: 11854868
  36. Novel target of the Notch-1/Hes-1 signaling pathway. PMID: 12065598
  37. Two novel mutations of the acid alpha-glucosidase gene, P361L and R437C, were found in a juvenile-onset glycogen storage disease type II (GSDII) 16-year-old Chinese patient. The asymptomatic 13-year-old brother of the proband is also compound heterozygote. PMID: 12601120
  38. Mutations in the alpha glucosidase gene are associated with infantile onset glycogen storage disease type II. PMID: 12923862
  39. Childhood Pompe disease demonstrating phenotypic variability of p.Asp645Asn. PMID: 15145338
  40. Data show that the mature forms of GAA characterized by polypeptides of 76 or 70 kDa are, in fact, larger molecular mass multicomponent enzyme complexes; peptides released during proteolytic processing remained tightly associated with the major species. PMID: 15520017
  41. Two novel mutations (Ala237Val and Gly293Arg) were found in the acid alpha-glucosidase gene in a Pompe disease patient with vascular involvement. PMID: 15668445
  42. Acid-alpha-glucosidase activity and specific activity, and lysosomal glycogen content are useful predictors of age of onset in Pompe disease. PMID: 15993875
  43. Complete molecular analysis of the GAA gene of patients with late onset glycogen storage disease type II shows missense mutations and splicing mutations. PMID: 16917947
  44. From 14 Argentinean patients diagnosed with either infantile or late-onset disease, we identified 14 distinct mutations in the acid alpha-glucosidase (GAA) gene, including nine novel variants. PMID: 17056254
  45. Two new missense mutations (p.266Pro>Ser and p.439Met>Lys) were new missense mutations causing late onset GSD II. PMID: 17092519
  46. Patients with the same c.-32-13T-->G haplotype (c.q. GAA genotype) may manifest first symptoms at different ages, indicating that secondary factors may substantially influence the clinical course of patients with this mutation. PMID: 17210890
  47. This study demonstrated a significant increase of GAA activity (1.3-7.5-fold) after imino sugar treatment in fibroblasts from patients carrying the mutations L552P (three patients) and G549R (one patient). PMID: 17213836
  48. N-glycans of recombinant human GAA were expressed in the milk of transgenic rabbits. PMID: 17293352
  49. The role of autophagy in Pompe disease was examined by analyzing single muscle fibers. PMID: 17592248
  50. Mutations in glucosidase alpha are associated with glycogen storage disease type II. PMID: 17616415

Show More

Hide All

Database Links

HGNC: 4065

OMIM: 232300

KEGG: hsa:2548

STRING: 9606.ENSP00000305692

UniGene: Hs.1437

Involvement In Disease
Glycogen storage disease 2 (GSD2)
Protein Families
Glycosyl hydrolase 31 family
Subcellular Location
Lysosome. Lysosome membrane.

Q&A

What is the biological function of GAA and why are GAA antibodies important research tools?

Lysosomal alpha-glucosidase (GAA) is a crucial enzyme involved in the breakdown of glycogen within lysosomes. Its primary function is to hydrolyze glycogen into glucose, helping to maintain glucose homeostasis and prevent glycogen accumulation in various tissues. Dysfunction or deficiency of this enzyme leads to Pompe disease (glycogen storage disease type II) .

GAA recombinant monoclonal antibodies serve as important research tools for several reasons:

  • They enable precise detection of GAA protein expression in various tissues and experimental systems

  • They allow researchers to track the distribution and localization of GAA in cellular and animal models

  • They facilitate the development of targeted therapeutic approaches for Pompe disease

  • They can be used in analytical applications such as ELISA and immunohistochemistry to study GAA biology

The methodological value of these antibodies lies in their ability to provide specific binding to GAA protein with minimal cross-reactivity, enabling reliable experimental results in both basic and translational research contexts.

How are GAA recombinant monoclonal antibodies generated and what are the key production methods?

GAA recombinant monoclonal antibodies are typically produced through a multi-step process that combines molecular biology techniques with cell culture systems. The general methodology involves:

  • Isolation of B cells from immunoreactive rabbits or other animals that have been exposed to GAA antigen

  • Extraction and amplification of GAA-specific antibody genes from these B cells

  • Cloning of the antibody genes into suitable phage vectors

  • Introduction of these vectors into mammalian cell expression systems

  • Expression of functional antibodies in culture

  • Purification via affinity chromatography to isolate the GAA-specific antibodies

Alternative production platforms include plant-based systems, which have shown promise for specialized glycoengineering applications. For instance, researchers have successfully used Arabidopsis cell cultures to produce GAA with specific glycan structures (such as M3) that are advantageous for mannose receptor-mediated cellular delivery .

The choice of production system significantly impacts the post-translational modifications of the antibody, particularly glycosylation patterns, which can affect functionality, stability, and immunogenicity of the final product.

What analytical techniques are most effective for characterizing GAA recombinant monoclonal antibodies?

Comprehensive characterization of GAA recombinant monoclonal antibodies requires multiple analytical approaches:

  • Functional Analysis:

    • ELISA for binding specificity and affinity determination

    • Immunohistochemistry (IHC) for tissue reactivity assessment

    • Enzymatic activity assays to confirm that antibody binding doesn't interfere with GAA function

  • Structural Characterization:

    • SDS-PAGE and Western blotting for molecular weight and purity assessment

    • Mass spectrometry for detailed protein sequence verification and post-translational modification mapping

    • Size-exclusion chromatography for aggregation analysis

  • Glycosylation Analysis:

    • Lectin binding assays for glycan pattern screening

    • HPLC or mass spectrometry-based glycan profiling

    • N-glycan release and analysis to determine mannose-6-phosphate content, which is crucial for lysosomal targeting

  • Stability Testing:

    • Accelerated stability studies under various storage conditions

    • Thermal shift assays to determine melting temperatures

    • Freeze-thaw stability assessment

When implementing these techniques, researchers should establish appropriate acceptance criteria based on the intended application of the antibody, whether for analytical detection or therapeutic development.

How can antibody:GAA fusion proteins overcome the limitations of conventional enzyme replacement therapy for Pompe disease?

Conventional enzyme replacement therapy (ERT) with recombinant human GAA (rhGAA) faces significant limitations, particularly poor delivery to skeletal muscle and loss of the enzyme to the liver. These challenges stem from the reliance on cation-independent mannose-6-phosphate receptor (CI-MPR) for cellular uptake. Antibody:GAA fusion proteins represent an innovative approach to address these limitations through several mechanisms:

  • Target-Specific Delivery: By fusing GAA to antibodies that recognize specific cell surface proteins (such as CD63 or ITGA7) that are preferentially expressed in affected tissues like skeletal muscle, researchers can bypass CI-MPR-dependent uptake mechanisms .

  • Enhanced Tissue Distribution: Antibody:GAA fusions demonstrate superior glycogen clearance in skeletal muscles compared to GAA alone, indicating improved delivery to the tissues most affected in Pompe disease .

  • Maintained Enzymatic Function: Critically, these fusion proteins retain the enzymatic activity of GAA while gaining the targeting benefits of the antibody moiety .

  • Lysosomal Processing: Upon internalization via antibody-mediated endocytosis, the fusion proteins undergo appropriate processing to the active lysosomal form of GAA, as demonstrated in both cellular models and in vivo studies in Pompe disease mouse models .

Experimental evidence shows that α-mCD63:GAA fusion proteins administered to Gaa 6neo/6neo mice resulted in accumulation of the lysosomal form of GAA within 6 hours of injection, with persistence for at least 48 hours post-injection. Furthermore, in skeletal muscles, α-mCD63:GAA removed more glycogen than GAA alone, demonstrating the enhanced delivery conferred by the antibody component .

What strategies are effective for inducing immune tolerance to rhGAA in the context of enzyme replacement therapy?

The development of high sustained antibody titers against rhGAA occurs in 35-40% of Pompe disease patients undergoing enzyme replacement therapy, negatively impacting therapeutic outcomes. Several immune tolerance induction (ITI) strategies have been investigated:

The optimal immune tolerance strategy should achieve long-term antigen-specific tolerance while preserving normal immune responses to other antigens, minimizing the duration of immunosuppression, and reducing infection risks, particularly in vulnerable infant populations with infantile Pompe disease.

How do post-translational modifications of GAA antibodies influence their functional properties and therapeutic efficacy?

Post-translational modifications (PTMs) of GAA antibodies, particularly glycosylation patterns, profoundly impact their functional properties and therapeutic efficacy through several mechanisms:

  • Mannose-6-Phosphate Content:

    • The mannose-6-phosphate (M6P) content of GAA is critical for CI-MPR-mediated uptake into tissues

    • Overexpression of GAA in certain cell systems can adversely affect M6P content

    • Controlled expression systems are essential to maintain optimal glycosylation

  • Paucimannose N-glycans:

    • GAA bearing predominantly M3 (mannose-3) glycan structures has shown advantages for mannose receptor (MR)-mediated cellular delivery

    • Plant-based expression systems, such as glycoengineered Arabidopsis cell cultures, can produce GAA with the desired M3 structure

    • This approach may benefit enzyme replacement therapies for multiple lysosomal storage disorders including Gaucher disease, Fabry disease, and Wolman disease

  • Antibody-Specific Glycosylation:

    • N-glycosylation of the Fc region of antibody portions in fusion proteins affects:

      • Serum half-life through interaction with FcRn receptors

      • Effector functions through binding to Fcγ receptors

      • Protein stability and solubility

      • Immunogenicity potential

  • Process-Dependent Variations:

    • Manufacturing process changes can significantly alter PTM profiles

    • Comparability studies are essential when process changes occur to ensure consistent quality attributes

    • PTM differences must be evaluated for potential impacts on safety and efficacy

Methodologically, researchers should implement comprehensive glycan analysis during development and manufacturing of GAA antibodies and fusion proteins. Techniques such as mass spectrometry, HPLC-based glycan profiling, and functional uptake assays are critical for establishing structure-function relationships between specific PTMs and therapeutic outcomes.

What are the optimal experimental designs for evaluating the efficacy of antibody:GAA fusion proteins in preclinical models?

Evaluating antibody:GAA fusion proteins in preclinical models requires carefully designed experiments to assess multiple aspects of efficacy, safety, and pharmacokinetics. Based on successful approaches in the literature, optimal experimental designs include:

  • Comparative Targeting Studies:

    • Direct comparison of antibody:GAA fusions with unconjugated GAA at equivalent doses

    • Inclusion of non-binding antibody:GAA controls to confirm antibody-mediated effects

    • Assessment in relevant cell lines with and without target receptor expression (e.g., HEK293 cells with/without CD63)

  • In Vivo Efficacy Assessment in Disease Models:

    • Utilization of established mouse models of Pompe disease (e.g., Gaa 6neo/6neo mice)

    • Measurement of glycogen clearance in multiple tissues (heart, skeletal muscles) as the primary efficacy endpoint

    • Time-course studies to determine onset and duration of therapeutic effect

    • Testing of multiple dose levels to establish dose-response relationships

  • Delivery Method Comparison:

    • When direct comparison to clinical ERT products is not feasible, alternative delivery methods such as hydrodynamic delivery (HDD) can be employed

    • This approach introduces plasmid DNA into hepatocytes for in vivo expression of the test proteins

    • Serum levels should be monitored to ensure comparable exposure between treatment groups

  • Processing and Trafficking Analysis:

    • Assessment of processing to the lysosomal GAA form in relevant tissues

    • Time-course analysis (e.g., evaluation at 6, 24, and 48 hours post-injection)

    • Subcellular fractionation to confirm lysosomal localization

  • Immune Response Monitoring:

    • Regular sampling for anti-rhGAA antibody quantification (e.g., every two weeks)

    • Utilization of ELISA to measure antibody titers

    • Inclusion of rechallenge phases after treatment interruption to assess memory response

  • Statistical Considerations:

    • Appropriate group sizes (n=10 per group has been successfully used)

    • Statistical analysis using two-tailed, equal variance student t-tests

    • Area under the curve analysis for cumulative antibody response

    • Significance threshold of p<0.05

These experimental approaches provide comprehensive evaluation of the pharmacological properties of antibody:GAA fusion proteins and enable evidence-based selection of candidates for further development.

What key parameters should be evaluated in comparability studies when process changes occur in GAA antibody production?

Process changes are inevitable throughout the lifecycle of recombinant monoclonal antibody therapeutics, including GAA antibodies. When such changes occur, comprehensive comparability studies are essential to ensure consistent product quality. Key parameters to evaluate include:

  • Primary Structure and Higher-Order Structure:

    • Amino acid sequence confirmation via peptide mapping

    • Disulfide bond patterns

    • Secondary and tertiary structure analysis using circular dichroism or FTIR

    • Thermal stability assessments

  • Post-Translational Modifications:

    • Complete glycosylation profile with special attention to mannose-6-phosphate content for GAA

    • Oxidation, deamidation, and other chemical modifications

    • C-terminal lysine variability

    • Glycation levels

  • Functional Characteristics:

    • Binding affinity to target antigens

    • For fusion proteins, enzymatic activity of the GAA component

    • Cellular uptake efficiency

    • Lysosomal targeting capability

  • Process-Related Impurities:

    • Host cell proteins

    • Host cell DNA

    • Leachables and extractables

    • Aggregates and particulates

  • Stability Indicators:

    • Real-time and accelerated stability profiles

    • Stress testing results (temperature, pH, agitation)

    • Freeze-thaw stability

The approach to comparability assessment should be risk-based, focusing most intensively on attributes that may affect safety and efficacy. Statistical tools should be employed to establish meaningful acceptance criteria based on historical data and process understanding. When differences are observed, their potential impact on clinical performance must be carefully evaluated using a combination of in vitro and in vivo functional assays .

How can researchers address data contradictions in GAA antibody research?

Data contradictions in GAA antibody research can arise from multiple sources, including differences in experimental models, analytical methods, and biological variability. A systematic approach to addressing these contradictions includes:

  • Methodological Standardization:

    • Implement consistent antibody characterization protocols across studies

    • Establish reference standards for key assays

    • Document detailed experimental conditions to enable proper comparison between studies

  • Cross-Validation of Results:

    • When contradictory findings emerge, reproduce experiments using multiple methodologies

    • For example, if cellular uptake differs between studies, compare different uptake quantification methods (flow cytometry, confocal microscopy, biochemical assays)

    • Validate findings across different cell lines or animal models

  • Addressing Mannose-6-Phosphorylation Variability:

    • Contradictions in uptake efficiency may stem from differences in M6P content

    • Conduct parallel glycan analysis alongside functional studies

    • Consider the impact of expression system on glycosylation patterns

  • Reconciling Immune Response Data:

    • Different mouse strains or immunological backgrounds can produce contradictory immune response data

    • When comparing immune tolerance strategies, include internal controls and standardized antibody measurement techniques

    • Consider genetic background effects and match control groups appropriately

  • Statistical Approaches:

    • Employ meta-analysis techniques when sufficient data are available

    • Use power calculations to ensure adequate sample sizes

    • Consider Bayesian approaches to integrate prior knowledge with new experimental data

  • Bridging Studies for Process Changes:

    • When process changes lead to seemingly contradictory results, design specific bridging studies

    • Focus on quality attributes with established relationships to clinical performance

    • Use orthogonal methods to confirm critical findings

By implementing these approaches, researchers can systematically address contradictions, identify their root causes, and develop a more coherent understanding of GAA antibody properties and performance.

What are the most promising glycoengineering strategies for optimizing GAA antibody therapeutics?

Glycoengineering represents a frontier in optimizing GAA antibody therapeutics, with several promising strategies emerging:

  • Targeted Mannose-6-Phosphate Enrichment:

    • Engineering expression systems with enhanced activity of N-acetylglucosaminyl-1-phosphotransferase and uncovering enzyme to increase M6P content

    • Developing in vitro enzymatic methods to modify glycans post-production

    • These approaches may enhance CI-MPR-mediated uptake while avoiding liver sequestration

  • Paucimannose Glycan Production:

    • Plant-based expression systems have demonstrated success in producing GAA with predominantly M3 glycan structures

    • The Arabidopsis alg3 cell culture system represents a promising platform for producing GAA with the desired M3 structure

    • This approach may benefit enzyme delivery through mannose receptor-mediated pathways

  • Hybrid Glycosylation Strategies:

    • Developing antibody:GAA fusion proteins with differential glycosylation of the antibody and enzyme components

    • Engineering the Fc portion for optimal serum half-life while independently optimizing GAA glycosylation for lysosomal targeting

  • Cell-Specific Glycoengineering:

    • Designing glycosylation patterns tailored to the receptors expressed by specific target tissues

    • For example, skeletal muscle-targeted formulations might employ different glycan strategies than cardiac-targeted formulations

  • Glycan Heterogeneity Control:

    • Implementing process controls to reduce batch-to-batch glycan variability

    • Using bioprocess parameters (temperature, pH, nutrient feeding strategies) to direct glycosylation patterns

    • This approach is critical for comparability during manufacturing scale-up

These glycoengineering strategies hold significant promise for creating next-generation GAA antibody therapeutics with enhanced targeting, reduced immunogenicity, and improved therapeutic efficacy. Implementation requires sophisticated analytical tools to characterize glycan structures and functional assays to correlate glycan patterns with therapeutic performance.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.