CYS2 Antibody

Shipped with Ice Packs
In Stock

Description

Cysteine-Linked Antibody-Drug Conjugates (ADCs)

Cysteine residues are often exploited for ADC development, where drugs are covalently linked to antibodies. Key findings include:

2.1. IgG2 Disulfide Isoforms and ADC Heterogeneity

IgG2 antibodies exhibit structural heterogeneity due to three disulfide isoforms (A, A/B, B), influencing ADC conjugation:

  • IgG2-A: Easier reduction of interchain disulfides, leading to higher drug-to-antibody ratios (DARs) and more homogeneous conjugation .

  • IgG2-B: Reduced accessibility of cysteines, resulting in lower DARs and greater heterogeneity .

IsoformDAR RangePrimary Conjugation SitesStability
IgG2-A0–12Hinge-region cysteinesModerate
IgG2-B0–12Hinge-region cysteinesHigh

2.2. Site-Specific Conjugation vs. Cysteine Conjugation

Comparative studies highlight advantages of site-specific methods over traditional cysteine conjugation:

  • Site-Specific ADCs: Use non-natural amino acids (e.g., para-acetylphenylalanine) for precise drug attachment, reducing off-target toxicity .

  • Cysteine ADCs: Heterogeneous drug loading (e.g., 3.8 drugs per antibody on average), increasing risks of aggregation .

3.1. COVID-19 and Cytokine Storm

  • Membrane Protein Epitopes: IgM-dominant responses to SARS-CoV-2 membrane proteins correlate with severe disease and long COVID, suggesting potential T-independent antigen targets .

  • Cytokine-Neutralizing mAbs: Clinical trials target IL-6, IL-1β, and TNF-α to mitigate cytokine storms in COVID-19 .

3.2. Antibody Engineering for Stability and Efficacy

  • Fc Modifications: Alterations in glycosylation (e.g., fucose removal) enhance effector functions like ADCC, improving therapeutic outcomes .

  • Hinge Optimization: Engineering the upper/lower hinge regions improves Fab-Fc flexibility and target binding .

Challenges and Future Directions

  • ADC Heterogeneity: Variability in DARs and conjugation sites remains a challenge for cysteine-linked ADCs, necessitating site-specific engineering .

  • Long COVID Biomarkers: Persistent IgM responses to conserved viral epitopes (e.g., membrane proteins) may inform diagnostics and vaccines .

Table 1: IgG2 Disulfide Isoforms and ADC Properties

ParameterIgG2-AIgG2-B
Reduction KineticsFaster disulfide reductionSlower disulfide reduction
DAR Range0–12 (higher average)0–12 (lower average)
Conjugation SitesHinge cysteinesHinge cysteines
StabilityModerateHigh

Table 2: Site-Specific vs. Cysteine ADCs

FeatureSite-Specific ADCsCysteine ADCs
Drug LoadingPrecise (2 drugs/antibody)Heterogeneous (0–12)
Toxicity ProfileLower off-target toxicityHigher risk of aggregation
StabilitySuperior serum stabilityModerate

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 weeks (Made-to-order)
Synonyms
CYS2 antibody; At2g31980 antibody; F22D22.27Cysteine proteinase inhibitor 2 antibody; AtCYS-2 antibody
Target Names
CYS2
Uniprot No.

Target Background

Function
This antibody targets a specific inhibitor of cysteine proteinases. It is likely involved in the regulation of endogenous processes and in plant defense mechanisms against pests and pathogens.
Gene References Into Functions
  • Studies have shown that high temperature and wounding stress increase the expression of both AtCYS1 and AtCYS2, although the timing and location of this induction differ. PMID: 20526604
Database Links

KEGG: ath:AT2G31980

STRING: 3702.AT2G31980.1

UniGene: At.13595

Protein Families
Cystatin family, Phytocystatin subfamily
Subcellular Location
Secreted.

Q&A

What is the structural and functional significance of cysteine residues in antibodies?

Cysteine residues are fundamental to antibody structure, forming critical disulfide bonds that maintain tertiary structure integrity. In IgG1 antibodies, approximately 50% of interchain disulfide bonds are involved in connecting heavy and light chains, compared to only 10% in IgG2 antibodies and 20% in IgG4 antibodies . These residues not only contribute to structural stability but also provide reactive thiol groups that serve as convenient chemical handles for site-specific conjugation of drugs, imaging agents, or other functional moieties.

The strategic positioning of cysteines significantly impacts antibody functionality. Modifications at these positions must be carefully designed to preserve antigen-binding capability and, in the case of therapeutic antibodies, maintain effector functions. The solvent accessibility of cysteine residues directly influences their susceptibility to chemical modifications and exchange reactions in biological environments, making their spatial arrangement a critical consideration in antibody engineering .

How can researchers assess the stability of cysteine-modified antibodies in experimental systems?

Stability assessment of cysteine-modified antibodies requires a multi-faceted approach combining biophysical characterization, accelerated degradation studies, and functional assays. Solution behavior analysis through concentration experiments and diffusion interaction parameter measurements helps evaluate how cysteine modifications affect antibody properties and aggregation propensity . These measurements are particularly important as drug conjugation can alter solution properties differently depending on the antibody isotype.

For in vivo stability assessment, pharmacokinetic (PK) studies using sensitive LC-MS-based bioanalytical methods are essential to monitor conjugate integrity over time . Research has demonstrated that antibodies with highly solvent-accessible cysteine sites can rapidly lose conjugated thiol-reactive linkers in plasma due to maleimide exchange with reactive thiols in albumin, free cysteine, or glutathione . In contrast, partially accessible sites with positively charged environments promote succinimide ring hydrolysis in linkers, preventing exchange reactions and improving stability.

What analytical techniques are most effective for characterizing cysteine-modified antibodies?

Liquid chromatography-mass spectrometry (LC-MS) stands as the gold standard for characterizing cysteine modifications in antibodies. This technique enables precise drug conjugation profiling, revealing the extent of disulfide bond disruption and modification patterns across different antibody domains . For assessing binding functionality, ELISA-based inhibitor screening assays provide quantitative data on whether antibody modifications affect antigen recognition, as demonstrated in studies with SARS-CoV-2 spike protein RBD antibodies .

Bead-based immunoassays, such as Luminex technology, offer multiplexed analysis of antibody specificity against panels of related antigens, allowing researchers to confirm that cysteine modifications don't compromise target selectivity . Additionally, neutralization assays provide critical functional data for therapeutic antibody candidates, ensuring that engineering modifications preserve or enhance the desired biological activity. For instance, ELISA-based SARS-CoV-2 inhibitor screening assays can demonstrate that modified antibodies maintain their ability to block viral protein interactions with cellular receptors .

What are common experimental challenges when working with cysteine-engineered antibodies?

A primary challenge with cysteine-engineered antibodies is achieving homogeneous conjugation products with defined stoichiometry. Traditional approaches often result in heterogeneous mixtures with variable drug-to-antibody ratios (0-8 drugs/antibody) and low yields (10-30%) . This heterogeneity complicates characterization and can introduce variability in experimental results.

Maintaining the balance between modification and functional integrity presents another significant challenge. Researchers must ensure that cysteine modifications don't disrupt critical disulfide bonds or interfere with antigen binding. Studies have shown that drug conjugation can affect different antibody isotypes differently, with some modifications increasing aggregation propensity .

The stability of cysteine-based conjugates in biological matrices also presents difficulties, as demonstrated by pharmacokinetic studies showing varying stability profiles across antibody isotypes . Researchers must carefully design experiments to monitor conjugate integrity throughout their studies, particularly for in vivo applications where plasma component interactions can lead to premature payload release.

How do computational design approaches enhance the development of cysteine-engineered antibodies?

Computational design has revolutionized cysteine-engineered antibody development by enabling rational epitope targeting with optimized complementarity-determining regions (CDRs). Recent fragment-based computational methods allow researchers to design antibody CDR loops that precisely target specific epitopes when structural information is available . This approach involves generating CDR-like fragments and strategically assembling them to create antibodies with multiple CDR candidates that recognize specific epitopes with high affinity and specificity.

The computational pipeline has been successfully applied to design antibodies against challenging targets, including the receptor-binding domain (RBD) of SARS-CoV-2, demonstrating efficacy even when using lower-resolution structural models as input . Remarkably, analysis of AlphaFold2 models from the CASP14 competition revealed that when using computational models as input for antibody design pipelines, typically about 75% of the generated CDRs would be identical to those obtained from corresponding crystal structures . This suggests that in silico approaches can reliably predict optimal configurations for cysteine-engineered antibodies, accelerating development timelines.

How does the microenvironment surrounding cysteine residues influence conjugate stability?

The local microenvironment surrounding cysteine residues profoundly impacts both conjugation chemistry efficiency and long-term conjugate stability. Research has established that solvent accessibility is a critical determinant of stability, with highly solvent-accessible cysteine sites prone to rapid loss of conjugated thiol-reactive linkers in plasma due to maleimide exchange reactions . Conversely, partially accessible sites offer improved conjugate retention.

The electrostatic environment surrounding the cysteine residue further modulates stability. Sites with positively charged environments promote hydrolysis of the succinimide ring in maleimide linkers, a beneficial reaction that prevents subsequent exchange with plasma thiols . This hydrolysis effectively "locks" the conjugate, enhancing its stability under physiological conditions. In mouse tumor models, the therapeutic activity of antibody conjugates correlated positively with succinimide ring hydrolysis and negatively with maleimide exchange reactions .

These findings demonstrate that the chemical and structural dynamics of the conjugation site can significantly influence antibody conjugate performance by modulating the stability of the antibody-linker interface. Understanding these microenvironment effects enables rational design of optimal conjugation sites based on specific application requirements.

What advanced strategies exist for achieving site-specific conjugation to antibody cysteine residues?

Several sophisticated strategies have emerged for site-specific conjugation to antibody cysteine residues. One approach involves engineering interchain disulfide bonds by replacing solvent-accessible cysteines with serines to reduce potential conjugation sites from eight down to four or two . This method has achieved near-quantitative yields (89-96%) with defined stoichiometries, producing engineered antibody-drug conjugates with comparable antigen-binding affinities and in vitro cytotoxic activities to their parental counterparts.

Selenocysteine interface technology represents another advanced approach, involving a minor modification at the C-terminus that doesn't interfere with disulfide bridges . This technique doesn't require activation steps and generates unique 1:1 stoichiometries of biological and chemical components, resulting in highly defined antibody conjugates.

Microenvironment engineering presents a third strategy, where cysteines are introduced at sites with specifically designed solvent accessibility and charge characteristics to control conjugation stability . By strategically selecting sites based on their local environment properties, researchers can create conjugates with optimal stability profiles for specific applications, balancing conjugation efficiency with in vivo performance.

How can high-throughput screening technologies accelerate the development of cysteine-engineered antibodies?

Cutting-edge high-throughput screening technologies have transformed the discovery process for cysteine-engineered antibodies. A particularly innovative approach combines microfluidic encapsulation of single antibody-secreting cells (ASCs) into antibody capture hydrogels with antigen bait sorting by conventional flow cytometry . This method enables screening of millions of cells at rates up to 10^7 cells per hour, dramatically accelerating the identification of promising candidates.

The workflow begins with droplet microfluidics to encapsulate single cells into hydrogel beads, creating a stable capture matrix that concentrates secreted antibodies . Flow cytometry then facilitates multiplexed detection and high-throughput sorting of antigen-specific ASCs for subsequent single-cell sequencing and recombinant antibody expression. This approach has successfully yielded monoclonal antibodies against SARS-CoV-2 with sub-nanomolar affinities (<1 pM) and potent neutralizing capacity (<100 ng/ml) .

The modular nature of these screening platforms enables their adaptation to various secreted molecules through simple replacement of capture and detection reagents . By facilitating access to the underexplored ASC compartment, these technologies enable efficient antibody discovery and detailed immunological studies into the generation of protective antibodies, with the ability to deliver pathogen-specific antibodies within just two weeks.

What experimental design principles should guide site selection for cysteine engineering in antibodies?

Site selection for cysteine engineering should follow a structured experimental design approach based on solvent accessibility, local charge environment, and functional considerations. Researchers should first analyze the three-dimensional antibody structure to identify potential modification sites that won't disrupt critical structural elements or antigen binding interfaces. Experimental evidence suggests engineering cysteines into sites with partial solvent accessibility rather than highly accessible positions, as the latter rapidly lose conjugated thiol-reactive linkers in plasma .

The local charge environment represents another critical parameter, with positively charged environments promoting beneficial succinimide ring hydrolysis that prevents subsequent exchange reactions . When designing experiments, researchers should include multiple candidate sites with varying microenvironment characteristics and perform comparative stability analyses under physiologically relevant conditions.

Functional testing must be integrated into the experimental design, assessing whether engineered antibodies retain their antigen binding capability and effector functions. Research has demonstrated that well-designed cysteine modifications can preserve these properties while gaining conjugation functionality . Finally, the experimental approach should include thorough characterization of conjugate homogeneity and stoichiometry, as these parameters significantly impact reproducibility and performance.

How can researchers optimize cysteine-engineered antibodies for maximum in vivo stability?

Optimizing cysteine-engineered antibodies for in vivo stability requires a systematic approach combining structural design considerations with empirical testing. The strategic selection of conjugation sites based on their microenvironment represents the first optimization step. Research has established that partially accessible cysteine sites with positively charged surroundings promote linker stabilization through succinimide ring hydrolysis .

Antibody isotype selection provides another optimization avenue, as pharmacokinetic studies have revealed isotype-dependent stability profiles. For instance, IgG1-based antibody-drug conjugates have shown poorer peripheral linker-payload stability compared to IgG2- and IgG4-based conjugates in rat models . When optimizing for in vivo applications, researchers should conduct comparative pharmacokinetic studies with sensitive LC-MS-based bioanalytical methods across multiple candidate designs.

Linker chemistry optimization further enhances stability, with self-hydrolyzing maleimides or alternative conjugation chemistries offering improved performance in physiological environments. The optimization process should include stability assessment in relevant biological matrices (serum, plasma) and appropriate animal models that reflect the intended application environment . This comprehensive approach balances conjugation efficiency with in vivo stability to produce antibody conjugates with optimal performance characteristics.

What methodological approaches best characterize the impact of cysteine modifications on antibody function?

A multi-faceted methodological approach is essential for comprehensively characterizing how cysteine modifications affect antibody function. Binding assays using technologies like surface plasmon resonance or bio-layer interferometry provide quantitative measurements of affinity and binding kinetics, allowing direct comparison between modified and unmodified antibodies. For therapeutic antibodies targeting viral pathogens, neutralization assays offer critical functional data, as demonstrated in studies with SARS-CoV-2 antibodies .

Competition assays represent another valuable methodology, assessing whether modified antibodies maintain their ability to block specific molecular interactions. For example, ELISA-based inhibitor screening assays can demonstrate that SARS-CoV-2 antibodies retain their capacity to inhibit spike protein binding to the ACE2 receptor despite modification . This approach directly links structural modifications to functional outcomes.

For antibodies intended for therapeutic applications, cell-based assays evaluating effector functions (ADCC, CDC, ADCP) provide essential functional characterization. Pharmacokinetic studies using sensitive bioanalytical methods further characterize the in vivo behavior of modified antibodies, revealing circulation half-life and tissue distribution patterns . Together, these methodologies create a comprehensive functional profile that guides optimization efforts and application-specific tailoring.

How can knowledge from serology studies inform the design of cysteine-engineered antibodies against viral targets?

Serology studies provide invaluable insights that can directly inform the design of cysteine-engineered antibodies against viral targets. Comprehensive analyses of antibody responses against pathogens like SARS-CoV-2 reveal which epitopes elicit neutralizing antibodies, guiding target selection for engineered constructs. For instance, antibodies binding to regions overlapping with the ACE2 receptor binding site on the SARS-CoV-2 spike protein are known to yield neutralizing candidates by sterically hindering virus binding to human cell receptors .

Isotype-specific response data from serology studies further inform antibody design. Research examining IgM, IgG, and IgA responses against various SARS-CoV-2 antigens (RBD, S protein, N protein) has documented significantly higher antibody levels against these antigens in acute and convalescent COVID-19 samples compared to healthy individuals . These findings can guide the selection of optimal antibody frameworks and isotypes for engineering efforts.

Studying antibody-secreting cells (ASCs) from convalescent individuals provides another methodological avenue, identifying naturally occurring antibodies with therapeutic potential that can serve as templates for cysteine engineering . By starting with antibodies already proven effective against viral targets, researchers increase the likelihood of developing successful cysteine-engineered therapeutic candidates with potent neutralizing capacity.

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.