CRS2A Antibody

Shipped with Ice Packs
In Stock

Description

Database and Literature Review

A systematic search across PubMed, PMC, clinical trial registries (ClinicalTrials.gov), and antibody-specific databases (e.g., The Antibody Society) yielded zero results for "CRS2A Antibody." Key observations include:

  • Structural antibody studies ( ) describe canonical antibody domains (e.g., CDRs, Fc regions) but do not reference CRS2A.

  • SARS-CoV-2 antibody research ( ) focuses on REGEN-COV (casirivimab + imdevimab), sotrovimab (S309), and other neutralizing antibodies, with no mention of CRS2A.

  • Antibody therapeutics databases ( ) list ~170 approved antibodies globally as of 2024, excluding CRS2A.

Nomenclature Issues

  • CRS2A may represent an internal project code, unpublished candidate, or typographical error (e.g., confusion with CR3022, a SARS-CoV-1 antibody).

  • Non-standard naming conventions could obscure visibility in public databases.

Therapeutic Context

Hypothetically, if CRS2A were an experimental antibody:

PropertyAssumed MechanismLikely Targets
StructureIgG1 or IgG4 subclassViral antigens or oncology
EngineeringHumanized or fully humanSpike protein, tumor antigens
StagePreclinical (unpublished)Infectious diseases or cancer

Recommendations for Further Inquiry

  1. Verify nomenclature with the originating institution or patent filings.

  2. Explore proprietary databases or industry pipelines for confidential candidates.

  3. Monitor preprint servers (e.g., bioRxiv) for emerging data.

Limitations

  • The analysis is constrained by publicly accessible data up to March 2025.

  • Proprietary or embargoed research may not be reflected.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CRS2A antibody; At5g38290 antibody; MSI17.10Chloroplastic group IIB intron splicing facilitator CRS2-A antibody; chloroplastic antibody; CRS2-like protein A antibody; Chloroplastic RNA splicing factor 2-A antibody
Target Names
CRS2A
Uniprot No.

Target Background

Function
CRS2A Antibody is essential for the splicing of group IIB introns in chloroplasts.
Database Links

KEGG: ath:AT5G38290

STRING: 3702.AT5G38290.2

UniGene: At.30420

Protein Families
PTH family, CRS2 subfamily
Subcellular Location
Plastid, chloroplast stroma.

Q&A

What is the relationship between antibodies and cytokine release syndrome (CRS)?

Antibody-mediated cytokine release syndrome represents a significant immunological phenomenon characterized by rapid and excessive release of inflammatory cytokines following antibody administration. CRS manifests as a systemic inflammatory response through dramatic increases in pro-inflammatory cytokines, particularly IL-6 (which can increase up to 146-fold within hours of antibody infusion), TNFα, IL-8, and IL-10 . This immune activation can lead to acute respiratory insufficiency and potentially progress to acute respiratory distress syndrome. The mechanism involves antibody-dependent enhancement (ADE), where antibodies paradoxically facilitate more severe manifestations of the disease they are designed to neutralize . When evaluating antibody therapies, researchers should monitor for CRS indicators including rapid cytokine elevation, respiratory parameters deterioration, and inflammatory marker fluctuations.

How can researchers distinguish between antibody-induced CRS and other causes of inflammatory responses?

Distinguishing antibody-induced CRS from other inflammatory responses requires methodical elimination of alternative triggers. Primary identification markers include: (1) temporal relationship between antibody administration and symptom onset (typically within hours), (2) characteristic cytokine profile showing rapid elevation, particularly IL-6, TNFα, IL-8 and IL-10, (3) absence of microbiological evidence for infectious triggers, and (4) normal procalcitonin levels despite elevated C-reactive protein .

Research methodologies should incorporate longitudinal sampling protocols to establish temporal patterns of cytokine elevation. Researchers should establish baseline inflammatory markers pre-antibody administration, then implement serial measurements post-administration (0, 2, 4, 6, 12, 24 hours) to capture the characteristic rapid-onset pattern distinct from bacterial infection-mediated inflammation (which typically shows slower progression and elevated procalcitonin) . Additionally, comprehensive microbiological assessment must be conducted to exclude infectious etiologies.

What physiochemical properties should researchers evaluate when characterizing antibodies for research applications?

Comprehensive antibody characterization requires evaluation of multiple physiochemical properties through a structured workflow. Key parameters include:

  • Self-interaction propensity - Measured via self-interaction chromatography or concentration-dependent dynamic light scattering

  • Aggregation tendency - Assessed through accelerated stability studies and size-exclusion chromatography (SEC)

  • Thermal stability - Determined via differential scanning calorimetry or differential scanning fluorimetry

  • Colloidal stability - Evaluated through zeta potential measurements and salt/pH gradient stability studies

  • Hydrophobicity profile - Characterized through hydrophobic interaction chromatography

Methodologically, researchers should implement high-throughput screening approaches that enable analysis of hundreds of antibody variants with minimal material (<1mg per antibody). This allows efficient evaluation of critical developability parameters concurrent with binding affinity and biological function assessment . Data management systems should be implemented to track correlations between physicochemical properties and functional performance.

How should researchers design a size-exclusion chromatography protocol for antibody characterization?

Development of a robust SEC method for antibody characterization requires systematic optimization of multiple parameters. Researchers should follow this methodological approach:

  • Column selection: Evaluate multiple stationary phases, particularly comparing silica hybrid materials with hydrophilically modified hybrid surfaces for hydrophobic antibodies. Research demonstrates that highly hydrophobic antibodies interact strongly with some modern silica hybrid materials, significantly increasing elution time, while hydrophilically modified hybrid surfaces show reduced interactions .

  • Mobile phase optimization: Implement a design-of-experiments approach varying buffer composition, pH, ionic strength, and flow rate.

  • Representative test set: When developing SEC methods, utilize a diverse subset of antibodies representing the full range of physicochemical properties. Research shows that a subset of 12 antibodies representing diverse properties can effectively predict performance across larger antibody panels .

  • Validation parameters: Systematically evaluate linearity (R²>0.99 across concentration range), repeatability (RSD<2% for retention time), range (typically 0.05-2.5 mg/mL), and accuracy (recovery 95-105%) .

  • Performance metrics monitoring: Track peak symmetry and elution time distribution across diverse antibody types. For hydrophilic antibodies, anticipate potential asymmetric peaks while monitoring for elution time shifts with hydrophobic variants .

What methods provide the most reliable antibody detection in research samples, and how do their sensitivities compare?

Comparative analysis of antibody detection methodologies reveals distinct performance characteristics researchers should consider:

For immunoglobulin detection, ELISA remains the reference standard for quantitative antibody profiling. Implementation should include dual measurement of both IgM and IgG antibodies, with careful establishment of optical density (OD) thresholds based on pre-characterized negative samples . Research shows negative samples typically present with median OD values near zero (-0.0001 for IgM and -0.01 for IgG), while positive samples demonstrate significant elevation (median 0.18 for IgM and 3.0 for IgG) .

Lateral flow immunoassay (LFIA) devices offer rapid results but demonstrate variable performance. Rigorous validation protocols must include testing against pre-characterized samples with documented ELISA results, with careful photographic documentation of results using standardized lighting conditions .

What controls should be implemented when evaluating antibody-mediated effects in experimental systems?

Robust experimental design for antibody-mediated effects requires multiple control conditions:

  • Isotype controls: Include matched isotype antibodies (same Ig class and light chain type) lacking target specificity to distinguish target-specific effects from Fc-mediated phenomena.

  • Fab/F(ab')₂ controls: Compare intact antibodies with Fab/F(ab')₂ fragments to isolate Fc-dependent versus antigen-binding dependent effects, particularly important when studying potential antibody-dependent enhancement (ADE) .

  • Dose-response relationships: Establish clear dose-response curves across a wide concentration range (typically 0.01-100 μg/mL) rather than single-dose experiments.

  • Temporal controls: Implement time-course studies capturing both early (0-6 hours) and delayed (24-72 hours) responses, as antibody-mediated cytokine release typically presents rapidly while other effects may develop over longer timeframes .

  • Cell-specific controls: When studying immune activation, include experiments with specific immune cell populations (monocytes, macrophages, NK cells) alongside mixed cell populations to identify cellular mediators of observed effects.

  • Cytokine neutralization: Include experimental conditions with specific cytokine neutralizing antibodies to establish causality between individual cytokines and observed phenotypes .

How can researchers identify antibodies with optimal developability profiles during early-stage discovery?

Implementing an effective high-throughput developability workflow during early antibody discovery requires integration of multiple analytical approaches. Research shows this methodology significantly increases success rates in CMC phases:

The optimal screening cascade employs multiparametric analysis combining:

  • Thermal stability assessment: Implement differential scanning fluorimetry to determine melting temperatures (Tm) with a minimum acceptable threshold of 65°C for lead candidates .

  • Colloidal stability evaluation: Utilize dynamic light scattering to measure diffusion interaction parameter (kD) and static light scattering for second virial coefficient (B22) determination. These parameters strongly correlate with downstream process behavior .

  • Sequence liability analysis: Employ in silico tools to identify post-translational modification sites (deamidation, oxidation, glycosylation), unpaired cysteine residues, and hydrophobic patches that may impact stability.

  • Expression level screening: Quantify transient expression yields as early indicators of manufacturability, with correlation to final process yields .

  • Accelerated stability studies: Conduct forced degradation studies under thermal and mechanical stress conditions to identify aggregation-prone candidates.

This workflow should be implemented iteratively, with circular analytics following each engineering cycle (e.g., mutagenesis to remove PTM sites or disrupt hydrophobic patches) . Data integration systems should correlate biophysical properties with downstream process parameters to enable rational candidate selection.

What factors contribute to antibody-dependent enhancement (ADE), and how can researchers assess this risk in experimental systems?

Assessing antibody-dependent enhancement risk requires sophisticated experimental approaches targeting multiple potential mechanisms:

ADE manifests through two primary mechanisms: enhanced viral replication and immune complex-mediated inflammation . Researchers should implement comprehensive evaluation protocols including:

  • Fc receptor-dependent enhancement assays: Conduct infection experiments in the presence of antibodies across multiple cell types expressing different Fc receptor classes (FcγRI, FcγRIIa, FcγRIIb, FcγRIII). Compare infection rates between wild-type cells and Fc receptor-blocked or knockout systems .

  • Complement-dependent enhancement assessment: Evaluate viral infection and immune activation in the presence and absence of complement, using heat-inactivated serum controls to distinguish complement-dependent effects.

  • Cytokine profiling: Implement multiplex cytokine arrays to identify specific inflammatory signatures associated with antibody-mediated enhancement. Key indicators include rapid elevation of IL-6, TNFα, IL-8, and IL-10 within hours of antibody exposure .

  • Tissue-specific models: Assess enhancement effects across multiple relevant tissue models, particularly focusing on systems with distinct Fc receptor expression profiles.

  • Antibody concentration ranges: Evaluate ADE potential across sub-neutralizing to super-neutralizing concentrations, as enhancement effects often demonstrate bell-shaped curves with peak enhancement at sub-neutralizing concentrations .

Importantly, research indicates that hematological malignancies may predispose to enhanced ADE risk, necessitating specialized assessment in this context .

How should researchers approach engineering antibodies to mitigate cytokine release syndrome risk?

Engineering antibodies to reduce CRS risk requires systematic modification of multiple structural elements:

  • Fc engineering approaches: Implement strategic modifications to reduce Fc receptor engagement, including:

    • L234A/L235A mutations to reduce FcγR binding

    • N297A/Q mutations to eliminate glycosylation and reduce Fc effector functions

    • IgG4 backbone implementation with S228P stabilization to minimize FcγR engagement

  • Binding kinetics optimization: Engineer antibody variable regions to favor moderate affinity (KD ~10⁻⁸-10⁻⁹ M) and faster dissociation rates (koff >10⁻³ s⁻¹) which demonstrate lower inflammatory potential compared to ultra-high affinity variants .

  • Epitope selection: Target epitopes that minimize crosslinking of cellular receptors, as receptor clustering often triggers inflammatory cascades.

  • Valency modification: Consider monovalent formats (Fab, scFv) or asymmetric bispecific approaches that maintain target binding while reducing receptor crosslinking.

  • Validation cascade: Implement a hierarchical testing strategy progressing from in vitro systems (human PBMC cytokine release assays) to humanized mouse models and NHP studies with comprehensive cytokine profiling .

Research indicates that the immunological landscape of patients significantly influences CRS risk, with hematological malignancies creating particularly complex scenarios requiring additional preclinical evaluation .

How should antibody studies be designed in the context of complex immune backgrounds such as hematological malignancies?

Researching antibody effects in complex immune backgrounds requires specialized methodological approaches:

Patients with hematological malignancies represent particularly challenging research contexts due to their altered immune landscapes. When designing studies for these populations, researchers should implement:

  • Patient-derived systems: Utilize primary cells from patients with the specific hematological malignancy under study rather than relying solely on healthy donor cells or established cell lines.

  • Disease-state stratification: Segment experimental analyses based on disease subtype, treatment status, and leukocyte subset distribution, as these factors significantly impact antibody responses .

  • Differential immune profiling: Characterize CD4/CD8 T cell ratios and myeloid cell distributions before antibody exposure, as these parameters correlate with differential cytokine release patterns. Research demonstrates that shifting CD4/CD8 ratios can alter inflammatory response profiles .

  • Therapy-interaction assessment: Evaluate antibodies in the context of concurrent therapies used in hematological malignancies (e.g., ATRA/ATO for acute promyelocytic leukemia), as these can promote differentiation of leukemic blasts and amplify cytokine responses .

  • Cytokine cascade modeling: Implement mathematical modeling approaches to predict cytokine cascade amplification based on baseline immune parameters.

Research documents cases where patients with acute promyelocytic leukemia experienced severe cytokine release syndrome following anti-SARS-CoV-2 antibody administration, with temporal relationships suggesting antibody-triggered immune activation .

What are the optimal approaches for analyzing antibody hydrophobicity and its impact on chromatographic behavior?

Advanced characterization of antibody hydrophobicity requires systematic implementation of complementary analytical techniques:

  • Stationary phase comparative analysis: Evaluate antibody behavior across multiple stationary phases, particularly comparing silica hybrid materials with hydrophilically modified hybrid surfaces. Research demonstrates that highly hydrophobic antibodies interact strongly with some modern silica hybrid materials (significantly increasing elution time), while hydrophilically modified hybrid surfaces show reduced interactions .

  • Peak symmetry assessment: Implement quantitative peak symmetry analysis using USP tailing factor or asymmetry factor calculations. Research shows hydrophilic antibodies exhibit asymmetric peaks to varying degrees across different stationary phases, even when elution time remains unaffected .

  • Reference panel implementation: Establish a diverse reference panel representing the full spectrum of antibody hydrophobicity. Studies demonstrate that a carefully selected subset of 12 antibodies can effectively represent the complete range of properties found across 138 clinical-stage antibodies .

  • Multi-parameter correlation analysis: Analyze correlations between hydrophobicity metrics and downstream behavior. Implement statistical approaches to correlate surface property measurements with chromatographic behaviors .

  • Mobile phase modulation: Systematically evaluate the impact of mobile phase parameters (ionic strength, pH, organic modifier concentration) on retention behavior to further characterize interaction mechanisms .

How can researchers establish antibody kinetic profiles to optimize detection timing in clinical and research applications?

Establishing robust antibody kinetic profiles requires systematic implementation of longitudinal sampling protocols:

  • Time-series experimental design: Implement consistent sampling intervals (baseline, days 3-5, 7-10, 14-17, 21-28, and 35-45) to capture the complete kinetic profile. Research demonstrates IgG antibody titers typically rise during the first three weeks post-exposure, with the lower bound of the 95% confidence interval crossing detection thresholds between days 6-7 .

  • Dual isotype monitoring: Simultaneously track both IgM and IgG development, recognizing their distinct kinetic patterns. Research indicates IgG demonstrates clear temporal association with time since symptom onset, while IgM shows no consistent temporal pattern .

  • Quantitative threshold determination: Establish optical density (OD) thresholds based on pre-characterized negative cohorts. Research shows negative samples typically present with median OD values near zero (-0.0001 for IgM and -0.01 for IgG), creating clear separation from positive samples (median 0.18 for IgM and 3.0 for IgG) .

  • Multivariate regression modeling: Implement statistical approaches to model antibody development rates accounting for variables including age, disease severity, and hospitalization status. Research suggests these factors do not significantly impact antibody titers in multivariable models .

  • Sensitivity timing optimization: Recognize that despite detection thresholds being crossed around day 7, optimal test performance typically occurs several days later due to sampling variation and individual immune response differences .

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.