AL6 Antibody

Shipped with Ice Packs
In Stock

Description

Clinical Applications and Efficacy

Anti-IL6 antibodies are primarily used to treat autoimmune and inflammatory conditions. Key clinical applications include:

Rheumatoid Arthritis (RA) and Autoimmune Diseases

Tocilizumab (Actemra), the first approved anti-IL6R antibody, inhibits IL-6 binding to its receptor, reducing synovial inflammation and joint damage in RA . It is also used for polyarticular juvenile idiopathic arthritis, giant cell arteritis, and Castleman’s disease.

Transplant Rejection and Alloantibody Responses

Clazakizumab, an anti-IL6 antibody, has shown promise in late antibody-mediated rejection (ABMR) in kidney transplants. A phase 2 trial demonstrated:

  • Reduced donor-specific antibodies and slower eGFR decline compared to placebo .

  • Molecular ABMR score resolution in 38.9% of patients after prolonged treatment .

Oncology and Vaccine Development

While not directly targeting IL-6, racotumomab (an anti-idiotype vaccine) and Annexin A6 antibodies are used in cancer research. Racotumomab improved survival in non-small cell lung cancer (NSCLC) by eliciting anti-NeuGcGM3 ganglioside antibodies .

Mechanism of Action

Anti-IL6 antibodies disrupt IL-6 signaling through two primary pathways:

  1. IL-6 Receptor Blockade: Tocilizumab binds to membrane-bound and soluble IL-6R, preventing IL-6 from activating downstream pathways (e.g., STAT3) .

  2. IL-6 Neutralization: Clazakizumab directly binds IL-6, inhibiting its interaction with IL-6R .

Key Biological Effects:

  • Immune Cell Modulation: Reduces Th17 and T follicular helper (Tfh) cells while increasing regulatory T cells (Tregs) .

  • Inflammation Suppression: Lowers acute-phase proteins (e.g., CRP, fibrinogen) and hepcidin, addressing anemia .

  • Tissue Protection: Inhibits osteoclast differentiation (via RANKL suppression) and neovascularization (via VEGF) .

Preclinical Insights

  • Alloantibody Recall: Anti-IL6R therapy reduces IgG production in bone marrow and spleen, attenuating alloantibody responses in transplant models .

  • Cancer Immunology: AVIDa-hIL6, a large-scale dataset of VHH antibodies, enables computational prediction of IL-6 interactions, aiding therapeutic antibody design .

Clinical Trial Data

Tocilizumab in Rheumatoid Arthritis:

ParameterResult
Approval Year2008 (Japan), global adoption thereafter
Target PathwayIL-6R blockade
Primary IndicationsRA, JIA, giant cell arteritis

Clazakizumab in ABMR:

EndpointClazakizumab vs. Placebo
eGFR Decline (Part A)-0.96 vs. -2.43 mL/min/1.73m²/month
Molecular ABMR Resolution38.9% (post-treatment)

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
AL6 antibody; At2g02470 antibody; T16F16.26 antibody; PHD finger protein ALFIN-LIKE 6 antibody; Protein AL6 antibody
Target Names
AL6
Uniprot No.

Target Background

Function
This antibody recognizes the histone-binding component that specifically binds to H3 tails trimethylated on 'Lys-4' (H3K4me3). H3K4me3 marks the transcription start sites of virtually all active genes.
Gene References Into Functions
  1. AL6 contains a plant homeo domain finger that can bind to trimethylated lysine 4 of histone H3. PMID: 23531693
  2. Research suggests that AL PHD-PRC1 complexes (AL6 and AL7), formed around H3K4me3, may facilitate a transition from the H3K4me3-associated active to the H3K27me3-associated repressive transcription state of seed developmental genes during seed germination. PMID: 24465219
  3. AL6 plays a role in regulating the transcription of genes essential for root hair elongation under low phosphate conditions. PMID: 23432399
Database Links

KEGG: ath:AT2G02470

STRING: 3702.AT2G02470.1

UniGene: At.43901

Protein Families
Alfin family
Subcellular Location
Nucleus.
Tissue Specificity
Ubiquitously expressed.

Q&A

What are the different types of AL6 antibodies used in research?

AL6 antibodies in research primarily refer to antibodies targeting ALG6 (Alpha-1,3-Glucosyltransferase) or ALK-6 (BMPR-IB). These include polyclonal antibodies like the rabbit anti-ALG6 antibody, which has been validated for immunocytochemistry and immunofluorescence applications . For ALK-6, monoclonal antibodies such as the clone 477914 targeting human BMPR-IB/ALK-6 have been developed for applications including flow cytometry and cellular imaging . Additionally, humanized anti-IL-6 receptor antibodies represent another important category with significant clinical applications in autoimmune diseases .

How do researchers differentiate between the specificity of ALG6 and ALK-6 antibodies?

Researchers differentiate these antibodies through validation techniques specific to each target. For ALG6 antibodies, enhanced validation protocols confirm binding to the Alpha-1,3-Glucosyltransferase protein . In contrast, ALK-6 (BMPR-IB) antibodies are validated using flow cytometry with positive controls like PC-3 human prostate cancer cell lines and iPS cells differentiated to mesoderm, alongside appropriate isotype controls to confirm specificity . The validation process typically involves:

Validation MethodALG6 AntibodiesALK-6 Antibodies
Cell linesVarious human cell linesPC-3, iPS-derived mesoderm
ControlsIsotype controls, blocking peptidesMAB0041 (isotype control)
TechniquesICC-IF with enhanced validationFlow cytometry, fluorescent ICC
Target regionVaries by antibodyLys14-Arg126 region

What is the historical development pathway of humanized antibodies like anti-IL-6 receptor antibodies?

The development of humanized antibodies began with fundamental discoveries about B cell activation's role in autoimmunity in the late 1970s. By 1984, IL-6 was cloned as BSF-2 (B cell stimulatory factor 2) and recognized for its role in B cell differentiation and antibody production . The development pathway included:

  • Identification of IL-6's role in autoimmune disease pathogenesis

  • Development of mouse monoclonal antibodies against the IL-6 receptor

  • Humanization using CDR-grafting technology in collaboration with the Medical Research Council in 1991

  • Clinical trials demonstrating efficacy in inflammatory arthropathies

  • Regulatory approval (tocilizumab was launched in Japan in 2008)

This development illustrates the lengthy progression from basic research to clinical application, requiring approximately 30 years from initial concept to approved therapeutic .

How can researchers optimize antibody validation for ALK-6 detection in flow cytometry experiments?

Optimization of ALK-6 antibody validation in flow cytometry requires systematic protocol development. Based on successful applications, researchers should:

  • Select appropriate positive control cell lines known to express BMPR-IB/ALK-6, such as PC-3 human prostate cancer cells or mesoderm-differentiated iPS cells

  • Establish optimal antibody concentration through titration experiments (successful protocols have used concentrations around 10 μg/mL)

  • Implement rigorous controls including matched isotype controls (e.g., MAB0041) to establish background staining levels

  • Optimize secondary antibody selection; allophycocyanin-conjugated or PE-conjugated anti-mouse IgG antibodies have demonstrated effective detection

  • Establish standardized gating strategies that account for autofluorescence and non-specific binding

The validation process should include comparative analysis between treated and untreated samples to confirm specificity and sensitivity of the detection system .

What are the key considerations when performing immunocytochemistry with ALK-6 antibodies?

When performing immunocytochemistry with ALK-6 antibodies, researchers should consider several critical factors:

  • Fixation method: Immersion fixation has been successfully employed for PC-3 cells

  • Antibody concentration: 10 μg/mL has been effective for 3-hour room temperature incubations

  • Visualization strategy: Secondary antibody selection is critical; NorthernLights™ 557-conjugated Anti-Mouse IgG has proven effective for visualization

  • Counterstaining: DAPI provides effective nuclear counterstaining to contextualize ALK-6 staining patterns

  • Subcellular localization interpretation: Expected staining patterns include both cytoplasmic and cell surface localization, which is consistent with the biology of this receptor protein

Researchers should develop detailed protocols for coverslip preparation, fixation, permeabilization, blocking, and antibody incubation to ensure reproducible results.

How do site-specific conjugation methods improve antibody-drug conjugate (ADC) development compared to traditional approaches?

Site-specific conjugation methods represent a significant advancement over traditional stochastic conjugation approaches for ADC development. The key improvements include:

  • Homogeneous drug-antibody ratio (DAR): Site-specific methods produce ADCs with consistent DAR values, leading to more predictable pharmacokinetic and pharmacodynamic profiles. For example, ThioMab technology developed by Genentech can produce ADCs with DAR of 2 with over 92% homogeneity .

  • Preserved antibody function: By directing conjugation away from antigen-binding regions, site-specific methods maintain antibody affinity. Traditional lysine-based conjugation can result in modifications near the antibody-antigen recognition sites, potentially reducing target binding .

  • Improved stability: Site-specific conjugation can enhance the stability of the linker-payload attachment, reducing premature drug release in circulation.

  • Optimized therapeutic window: The combination of consistent DAR and preserved antibody function leads to improved efficacy and reduced off-target toxicity.

Conjugation ApproachTechnology ExampleAdvantagesLimitations
Engineered cysteineThioMabHigh homogeneity (>92% DAR of 2)Potential for wrong disulfide formation
Disulfide re-bridgingBissulfone reagents, NGMs, PDsPreserves structural integrityLower conjugation efficiency
Unnatural amino acidsN-acetyl-L-phenylalanine, azido lysineControllable, quantitativeProduction challenges, potential immunogenicity
Enzyme-assistedFGE, transglutaminaseSite-specific modificationPotential immunogenicity
Glycan remodelingN-glycan at N297 modificationMinimal impact on antigen bindingComplexity of glyco-engineering

What methodological approaches resolve the challenges of DAR heterogeneity in antibody-drug conjugates?

The challenges of DAR heterogeneity can be addressed through several methodological approaches:

  • ThioMab technology: By introducing engineered cysteine residues at specific positions (e.g., light chain V110A, heavy chain A114C), researchers can achieve highly controlled conjugation. This approach has demonstrated 92.1% homogeneity with DAR of 2 .

  • Disulfide re-bridging conjugation: This method employs cysteine-selective cross-linking reagents such as bissulfone reagents, next-generation maleimides (NGMs), and pyridazinediones (PDs). These bis-reactive reagents reconnect polypeptide chains while simultaneously conjugating payloads .

  • Unnatural amino acid incorporation: Introduction of amino acids with special functional groups (e.g., N-acetyl-L-phenylalanine, azido methyl-L-phenylalanine) enables site-specific conjugation through orthogonal chemistry, generating ADCs with homogeneous DAR .

  • pClick technology: This recent innovation uses proximity-activated crosslinkers that react with the closest lysine residues on the antibody. The azide groups introduced provide sites for click chemistry with bioorthogonal handle-modified payloads, improving yield and antibody stability without requiring extensive antibody engineering .

These approaches have demonstrated significantly improved homogeneity compared to first-generation ADCs, which relied on stochastic conjugation to lysine or cysteine residues.

How can researchers effectively distinguish between specific and non-specific binding when validating AL6 antibodies?

Effective distinction between specific and non-specific binding requires a multi-faceted validation approach:

  • Isotype control implementation: Use of appropriate isotype controls (e.g., MAB0041 for mouse monoclonal antibodies) establishes baseline non-specific binding under identical experimental conditions .

  • Cellular expression models: Validation across cell lines with differential target expression provides critical specificity information. For ALK-6 antibodies, comparison between PC-3 cells (positive expression) and known negative cell lines helps confirm specificity .

  • Blocking experiments: Pre-incubation of antibodies with purified target protein should abolish specific staining while leaving non-specific interactions unaffected.

  • Signal-to-noise ratio quantification: Quantitative analysis of staining intensity between positive and negative controls should demonstrate statistically significant differences.

  • Cross-validation with multiple detection methods: Confirmation of target detection using independent techniques (e.g., flow cytometry, immunocytochemistry, and Western blotting) strengthens validation evidence.

A comprehensive validation strategy incorporating these elements provides robust evidence of antibody specificity before application in research contexts.

What analytical methods are used to characterize drug-antibody ratios in ADCs?

Characterization of drug-antibody ratios requires sophisticated analytical techniques:

  • UV-Vis spectroscopy: Differential absorption spectra between the antibody and payload enable estimation of average DAR.

  • Mass spectrometry techniques:

    • Intact mass analysis provides information on the distribution of conjugated species

    • Peptide mapping after proteolytic digestion identifies specific conjugation sites

    • Native MS preserves non-covalent interactions and provides insights into the three-dimensional structure

  • Hydrophobic interaction chromatography (HIC): Separates ADC species based on hydrophobicity differences resulting from varying numbers of conjugated payloads.

  • Capillary electrophoresis (CE): Provides high-resolution separation of ADC species based on charge-to-mass ratio differences.

These analytical methods are crucial for characterizing ADC heterogeneity, particularly in comparing stochastic conjugation approaches (showing wide DAR distribution from 0-8) versus site-specific methods (showing narrow DAR distribution) .

How do researchers apply ALK-6 antibodies in stem cell differentiation studies?

ALK-6 (BMPR-IB) antibodies serve as valuable tools in stem cell differentiation research, particularly in studying mesoderm development. The methodological approach includes:

  • Differentiation protocol implementation: Human induced pluripotent stem (iPS) cells can be directed toward mesoderm lineage using standardized differentiation protocols (e.g., using protocols like SC030B) .

  • Temporal expression analysis: ALK-6 antibodies enable tracking of BMPR-IB expression during differentiation stages, providing insights into BMP signaling dynamics during lineage commitment.

  • Flow cytometry application: Single-cell analysis using ALK-6 antibodies (e.g., MAB5051) followed by PE-conjugated secondary antibodies allows quantification of BMPR-IB expression in differentiating populations .

  • Co-expression analysis: Combined detection of ALK-6 with other lineage markers helps identify specific progenitor populations and characterize differentiation hierarchies.

  • Functional validation: Correlation of ALK-6 expression with functional responses to BMP ligands provides insights into signaling competence during differentiation.

This approach allows researchers to investigate the role of BMP signaling in directing cell fate decisions, particularly in mesodermal lineage specification.

What are the methodological considerations when using anti-IL-6 receptor antibodies to study autoimmune disease mechanisms?

When applying anti-IL-6 receptor antibodies in autoimmune disease research, several methodological considerations are essential:

  • Model selection: Choose appropriate experimental models that recapitulate key aspects of human autoimmune pathology and IL-6 signaling. The development of tocilizumab was informed by studies of B cell activation in autoimmunity .

  • Blocking validation: Confirm effective blocking of IL-6 signaling through phosphorylation analysis of downstream effectors (e.g., STAT3) after antibody treatment.

  • Dose-response characterization: Establish dose-response relationships for IL-6 receptor blocking to understand threshold effects and maximal inhibition parameters.

  • Temporal considerations: Determine optimal timing for antibody administration relative to disease initiation or progression phases in model systems.

  • B cell functional assessment: Given the role of IL-6 in B cell differentiation and antibody production, assess effects on:

    • Plasma cell differentiation

    • Antibody class switching

    • Autoantibody production

    • B cell proliferation

  • Cross-species reactivity awareness: Recognize potential limitations in cross-species reactivity when translating between model systems and human applications .

These methodological approaches can provide valuable insights into IL-6's role in autoimmune pathogenesis and therapeutic intervention mechanisms.

How can researchers address non-specific binding issues when working with AL6 antibodies?

Non-specific binding problems can be systematically addressed through protocol optimization:

  • Blocking optimization:

    • Test different blocking agents (BSA, serum, commercial blocking reagents)

    • Evaluate concentration-dependent effects (typically 1-5% for protein blockers)

    • Consider extended blocking periods (1-2 hours at room temperature)

  • Antibody dilution optimization:

    • Perform serial dilution experiments to identify optimal concentration

    • For ALK-6 detection, 10 μg/mL has been effective, but this should be validated for each application

  • Buffer modification:

    • Add detergents (0.05-0.1% Tween-20) to reduce hydrophobic interactions

    • Adjust salt concentration to disrupt electrostatic interactions

    • Consider carrier proteins (0.1-0.5% BSA) to prevent non-specific adsorption

  • Incubation conditions:

    • Optimize temperature (4°C can reduce non-specific interactions)

    • Adjust incubation time (3 hours at room temperature has been effective for some applications)

  • Negative control implementation:

    • Include isotype controls (e.g., MAB0041 for mouse monoclonal antibodies)

    • Consider cell lines known to be negative for the target

These optimization strategies should be approached systematically, modifying one parameter at a time while maintaining detailed records of experimental conditions.

What factors affect the stability of humanized antibodies in experimental systems?

The stability of humanized antibodies like anti-IL-6 receptor antibodies is influenced by multiple factors that researchers must control:

  • Storage conditions:

    • Temperature (typically -20°C to -80°C for long-term storage)

    • Freeze-thaw cycles (minimize through aliquoting)

    • Buffer composition (pH, ionic strength, presence of stabilizers)

  • Aggregation factors:

    • Protein concentration (higher concentrations increase aggregation risk)

    • Mechanical stress during handling (minimize vortexing or vigorous pipetting)

    • Interface exposure (air-liquid, liquid-solid)

  • Chemical modifications:

    • Oxidation of methionine residues in CDR regions can reduce binding affinity

    • Deamidation of asparagine residues affects stability and function

    • Isomerization of aspartic acid residues may alter conformation

  • Formulation considerations:

    • Addition of stabilizers (e.g., sucrose, trehalose)

    • Surfactants to prevent adsorption and aggregation

    • Antioxidants to prevent oxidation

  • Experimental design factors:

    • Incubation time and temperature

    • Matrix effects from biological samples

    • Compatibility with fixation procedures

Controlling these factors is crucial for maintaining antibody activity across experimental applications and ensuring reproducible results, particularly important for antibodies like tocilizumab that underwent extensive development for clinical applications .

How might emerging ADC technologies improve targeting specificity and therapeutic index in next-generation antibody applications?

Emerging ADC technologies are poised to enhance targeting specificity and therapeutic index through several innovative approaches:

  • Advanced site-specific conjugation:

    • Further refinement of ThioMab technology to eliminate wrong disulfide bond formation

    • Development of improved bioorthogonal chemistry approaches with higher efficiency

    • Integration of pClick technology with engineered antibodies for optimal conjugation sites

  • Novel linker technologies:

    • Development of tumor microenvironment-responsive linkers sensitive to multiple stimuli

    • Engineering linkers with improved plasma stability but efficient intracellular release

    • Creation of linkers capable of controlled drug release kinetics for optimized payload delivery

  • Payload innovations:

    • Development of hydrophilic payloads that maintain potency while reducing ADC aggregation

    • Exploration of immune-modulating payloads beyond traditional cytotoxics

    • Engineering of dual-action payloads with complementary mechanisms

  • Antibody engineering:

    • Fc engineering to enhance or reduce immune effector functions as needed

    • Development of bispecific ADCs targeting two tumor antigens simultaneously

    • Engineering of pH-dependent binding to enhance tumor-specific drug delivery

The integration of these approaches aims to overcome limitations of first and second-generation ADCs, particularly addressing the challenges of heterogeneous DAR, rapid clearance, and off-target toxicity that have limited therapeutic windows in current applications .

What are the emerging applications of quantitative imaging techniques in understanding AL6 antibody binding kinetics and cellular distribution?

Advanced quantitative imaging techniques are expanding our understanding of antibody-target interactions:

  • Super-resolution microscopy applications:

    • Visualization of nanoscale distribution of targets like ALK-6 on cell surfaces

    • Resolution of receptor clustering and co-localization with signaling partners

    • Tracking of internalization pathways with unprecedented spatial precision

  • Live-cell imaging innovations:

    • Real-time tracking of antibody binding and internalization kinetics

    • Visualization of receptor dynamics following antibody engagement

    • Monitoring intracellular trafficking and payload release for ADCs

  • Correlated light and electron microscopy (CLEM):

    • Correlation of fluorescence signals with ultrastructural context

    • Precise localization of antibody binding sites within cellular compartments

    • Validation of internalization pathways identified by fluorescence techniques

  • Quantitative analysis approaches:

    • Machine learning algorithms for automated quantification of binding patterns

    • Mathematical modeling of binding kinetics from imaging data

    • Spatial statistics to characterize clustering and co-localization phenomena

These techniques extend beyond traditional applications of ALK-6 antibodies in flow cytometry and static immunocytochemistry , enabling dynamic understanding of receptor biology and antibody-target interactions with potential implications for both basic research and therapeutic development.

How do the characteristics of first, second, and third-generation ADCs compare in research applications?

The evolution of ADC technology across three generations provides important insights for research applications:

CharacteristicFirst GenerationSecond GenerationThird Generation
Antibody typeMurine or humanized IgG4Humanized IgG1Engineered humanized IgG1
Linker stabilityAcid-labile, prone to hydrolysisImproved stability, cleavableSite-specific, customized stability
Conjugation methodStochastic (lysine/cysteine)Improved stochasticSite-specific
DAR distributionHighly heterogeneous (0-8)Moderately heterogeneousHomogeneous
Payload potencyModerateHighUltra-high
Payload hydrophobicityHighModerateEngineered hydrophilicity
ExampleGemtuzumab ozogamicinBrentuximab vedotinSite-specific ADCs
Research applicationsLimited by heterogeneityImproved reliabilityPrecise mechanistic studies
Key limitationsImmunogenicity, aggregationOff-target toxicity, rapid clearanceComplexity of production

This evolution demonstrates how technological advancements have addressed key limitations of earlier generations, enabling more precise and reliable research applications . Third-generation ADCs with homogeneous DAR and optimized pharmacokinetic properties provide superior tools for mechanistic studies and translational research.

What methodological approaches can researchers use to compare the efficacy of different AL6 antibody clones in experimental systems?

Systematic comparison of AL6 antibody clones requires comprehensive methodological approaches:

  • Binding affinity characterization:

    • Surface plasmon resonance (SPR) to determine kon, koff, and KD values

    • Enzyme-linked immunosorbent assay (ELISA) for comparative binding under standardized conditions

    • Flow cytometry with quantitative beads to establish binding site numbers

  • Epitope mapping:

    • Peptide array analysis to identify linear epitopes

    • Hydrogen-deuterium exchange mass spectrometry for conformational epitopes

    • Competition assays to determine epitope overlap between clones

  • Functional assessments:

    • Signaling pathway activation or inhibition (e.g., SMAD signaling for ALK-6 antibodies)

    • Receptor internalization rates quantified by flow cytometry or imaging

    • Downstream functional effects in relevant cellular models

  • Cross-reactivity profiling:

    • Testing against related family members (e.g., other ALK receptors)

    • Species cross-reactivity evaluation for translational applications

    • Assessment of binding to splice variants or post-translationally modified forms

  • Application-specific performance:

    • Side-by-side comparison in flow cytometry with standardized protocols

    • Parallel immunocytochemistry using identical samples and conditions

    • Comparative Western blotting with standardized sample preparation

These methodological approaches provide comprehensive characterization that enables researchers to select optimal antibody clones for specific applications or experimental questions.

What are the most promising research directions for improving antibody specificity and reducing off-target effects?

Several promising research directions are emerging to enhance antibody specificity:

  • Computational design approaches:

    • Machine learning algorithms to predict cross-reactivity risks

    • Structure-based optimization of CDR regions for enhanced specificity

    • In silico screening against human proteome to identify potential off-targets

  • Affinity maturation innovations:

    • Directed evolution with negative selection against off-targets

    • Yeast display technologies with multi-parameter sorting

    • Rational design of CDR residues based on structural insights

  • Conditional activation strategies:

    • Development of antibodies that activate only in specific microenvironments

    • Masking technologies that reveal binding sites selectively in target tissues

    • Split-antibody approaches requiring reconstitution by tumor-specific factors

  • Multi-specific targeting:

    • Bispecific or multispecific formats requiring engagement of multiple targets

    • AND-gate logic in antibody design to improve specificity

    • Avidity-based targeting of unique epitope combinations

These approaches aim to address the specificity challenges encountered with current antibodies, potentially leading to research tools and therapeutics with improved target engagement and reduced off-target effects .

How might advances in antibody engineering influence the future development of research tools for studying ALG6 and ALK-6 functions?

Antibody engineering advances are poised to transform research tools for studying ALG6 and ALK-6:

  • Intrabody development:

    • Engineering antibody fragments capable of intracellular expression

    • Creation of domain-specific inhibitors for precise functional dissection

    • Development of sensors for real-time monitoring of ALG6 or ALK-6 activity

  • Spatiotemporal control systems:

    • Optogenetic antibody systems allowing light-controlled binding

    • Chemically inducible antibody fragments for temporal control

    • Subcellular targeting motifs for compartment-specific inhibition

  • Functional antibody formats:

    • Proximity-inducing antibodies to study protein-protein interactions

    • Degrader antibodies to induce target protein degradation

    • Conformation-specific antibodies to distinguish active vs. inactive states

  • Multiplexed detection systems:

    • DNA-barcoded antibody technologies for single-cell multi-parameter analysis

    • Mass cytometry-compatible antibodies for highly multiplexed phenotyping

    • Imaging mass cytometry applications for spatial analysis of receptor distributions

  • Enhanced validation approaches:

    • Integration of CRISPR knockout controls in antibody validation

    • Single-molecule tracking capabilities for dynamic studies

    • Correlative multi-omics approaches linking antibody detection to functional outcomes

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.