ADF8 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

ADF8 Antibody binds specifically to Actin-depolymerizing factor 8, a member of the ADF/cofilin protein family that modulates actin polymerization and depolymerization. Key features include:

  • Target species: Primarily validated in Oryza sativa subsp. japonica (rice) and cross-reactive with homologs in Arabidopsis thaliana .

  • Immunogen: A synthetic peptide with 100% sequence homology to ADF8 (UniProt ID: AT4G00680 in Arabidopsis) and 88% homology to ADF3 (AT5G59880) and ADF7 (AT4G25590) .

Research Applications

ADF8 Antibody is utilized in multiple experimental contexts:

  • Immunoblotting: Detects ADF8 expression in rice and other plant tissues .

  • Immunofluorescence: Localizes ADF8 in cytoskeletal networks .

  • Comparative studies: Assesses ADF8’s role in actin dynamics across plant species .

Role in Plant Cytoskeleton Regulation

ADF8 facilitates actin filament turnover, critical for processes like cell elongation and stress responses. Its antibody has been used to:

  • Track ADF8 expression during rice root development .

  • Study actin remodeling in Arabidopsis under abiotic stress .

Cross-Reactivity Insights

The antibody’s partial homology with other ADF isoforms necessitates careful validation to avoid off-target effects in species with conserved ADF sequences .

Comparative Analysis with Related Antibodies

ADF8 Antibody is part of a broader toolkit for studying actin dynamics. Contrasts with other ADF-targeting antibodies include:

AntibodyTarget SpeciesSpecificityApplications
ADF11 AntibodyArabidopsis thaliana81% homology to ADF6 and ADF8 Immunoprecipitation, blotting
ADF4 AntibodyOryza sativaQ84TB3 (Rice-specific) Cytoskeletal imaging

Technical Validation

  • Storage: Lyophilized at -20°C; stable for 12 months .

  • Quality Control: Validated via ELISA and Western blot using recombinant ADF8 protein .

Future Directions

Research leveraging ADF8 Antibody could explore:

  • Mechanistic studies of actin dynamics in crop plants under climate stress.

  • Development of CRISPR-edited ADF8 mutants to dissect its role in plant morphogenesis.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
ADF8 antibody; At4g00680 antibody; F6N23.12Actin-depolymerizing factor 8 antibody; ADF-8 antibody; AtADF8 antibody
Target Names
ADF8
Uniprot No.

Target Background

Function
ADF8 Antibody targets an actin-depolymerizing protein. This antibody severs actin filaments (F-actin) and binds to actin monomers.
Database Links

KEGG: ath:AT4G00680

STRING: 3702.AT4G00680.1

UniGene: At.34524

Protein Families
Actin-binding proteins ADF family
Subcellular Location
Cytoplasm, cytoskeleton.
Tissue Specificity
Expressed in the root trichoblast cells and developed root hairs.

Q&A

What are the key components of an Antibody-Drug Conjugate (ADC)?

Antibody-Drug Conjugates consist of three essential components:

  • Antibody: Usually a monoclonal antibody that targets specific antigens expressed predominantly on tumor cells. Most ADCs utilize immunoglobulin G (IgG) as the antibody isotype, with IgG1 being the most widely used platform due to its improved solubility, greater complement-fixation, low nonspecific immunity, and better immune effector cell receptor binding efficiencies .

  • Linker: The chemical bridge that connects the antibody to the cytotoxic payload. Linkers must maintain stability in circulation while facilitating drug release at the target site.

  • Cytotoxic Payload: A potent drug that exerts therapeutic effects on target cells once internalized. The payload is typically much smaller than the antibody component.

This structure allows ADCs to integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity .

How is the Drug-to-Antibody Ratio (DAR) determined and why is it important?

The Drug-to-Antibody Ratio (DAR) represents the average number of drug molecules attached to each antibody molecule and is a critical quality attribute of ADCs that impacts both cytotoxicity and pharmacokinetics.

Determination Methods:

  • Hydrophobic Interaction Chromatography (HIC): Separates ADC species based on hydrophobicity differences caused by varying numbers of attached drug molecules

  • SE-HPLC: Size exclusion chromatography can assess changes in molecular weight

  • Mass Spectrometry: Provides precise determination of DAR with detailed structural information

Importance of DAR:

  • Typically, optimal DAR values range between 3.4 and 4.4, with 3.9 being an ideal target for many ADCs

  • Higher DAR values generally correlate with increased cytotoxicity but may compromise pharmacokinetic properties

  • Heterogeneity in DAR can compromise therapeutic efficacy and is a significant challenge in ADC development

Most ADC developers use DOE (Design of Experiments) approaches to control DAR during process development, establishing a "Design Space" and optimal setpoint to ensure consistent drug loading .

What analytical methods are commonly used for detection and quantification of ADCs?

Several complementary analytical methods are employed to characterize and quantify ADCs:

Ligand-Binding Assays (LBAs):

  • ELISA (Enzyme-Linked Immunosorbent Assay): The gold standard for large molecule analysis, offering high sensitivity without requiring sample extraction

  • Detection approaches: Utilizes generic anti-human antibody, protein A, G, or L, or more specific detection reagents such as anti-idiotype antibodies

Hybrid Approaches:

  • LB-LC-MS (Ligand-Binding Liquid Chromatography-Mass Spectrometry): Combines affinity enrichment with MS detection, becoming the gold standard in ADC bioanalysis

  • Workflow: Involves immobilized anti-human Fc antibody to isolate ADCs, followed by washing, proteolytic digestion, and MS analysis

Chromatographic Methods:

  • Size Exclusion Chromatography (SEC): Assesses purity and aggregation

  • Hydrophobic Interaction Chromatography (HIC): Characterizes DAR distribution

  • CE-SDS (Capillary Electrophoresis-Sodium Dodecyl Sulfate): Analyzes reduced and non-reduced ADCs

Each method offers unique advantages and the combination of multiple techniques provides comprehensive characterization of ADC quality attributes.

What strategies are being developed to overcome heterogeneity in ADC drug-to-antibody ratios?

Heterogeneity in drug-to-antibody ratios presents a significant challenge in ADC development. Several innovative approaches are being explored to achieve more homogeneous conjugation:

Site-Specific Conjugation Technologies:

  • Engineered Cysteine Residues: Introduction of cysteine at defined positions allows for controlled conjugation, reducing heterogeneity in drug loading

  • Unnatural Amino Acid (UAA) Incorporation: UAAs containing orthogonal side-chain functional groups can be introduced at specific positions in the antibody for stable conjugation. For example, HER2-targeted mAb incorporated with UAA pAF conjugated to tubulin inhibitor AS269 (ARX788) shows high serum stability and improved half-life

  • Enzymatic Approaches: Utilizing site-specific enzymatic conjugation to control conjugation sites

Antibody Engineering Strategies:

  • Genetic engineering: Modification of antibody sequences to introduce specific reaction sites

  • Chemoenzymatic modifications: Direct modification of antibody structures to facilitate controlled conjugation

  • Metabolic labeling: Incorporation of reactive groups during antibody production

These approaches aim to develop ADCs with accurate and consistent DARs to enhance therapeutic efficacy while maintaining optimal pharmacokinetic properties .

How do biparatopic and bispecific antibodies enhance ADC efficacy?

Biparatopic and bispecific antibodies represent advanced antibody engineering strategies that can significantly enhance ADC efficacy:

Biparatopic Antibodies:

  • Mechanism: Target two different epitopes on the same antigen

  • Benefits: Help cluster antigenic receptors, leading to rapid internalization of ADCs, which is crucial for payload delivery

  • Application: Particularly valuable for targets with limited expression or slow internalization kinetics

Bispecific Antibodies:

  • Mechanism: Simultaneously bind to two different antigens

  • Advantages:

    • Enhanced targeting by engaging two separate antigens simultaneously

    • Potential to overcome resistance mechanisms by targeting complementary pathways

    • Improved tumor penetration through dual-targeting strategies

Masked Antibody Approach:

  • Design: Antibody binding regions are masked by peptides selectively cleaved by proteases in the tumor microenvironment

  • Benefit: Activation of binding and therapeutic functions specifically within the tumor environment, minimizing off-target effects

These innovative antibody formats continue to advance the field of immunotherapy and targeted therapies with applications in cancer and other medical conditions, offering greater control and versatility while minimizing off-target effects .

What factors influence antibody distribution and penetration in solid tumors?

A significant challenge in ADC development is achieving optimal antibody distribution within solid tumors:

Key Limitations:

  • Heterogeneous distribution of antibodies when administered systemically is a long-recognized challenge

  • Antibody internalization and clearance obstruct uptake in solid tumors

  • Tumor vascular permeability and diffusion barriers limit antibody penetration

Factors Affecting Penetration:

  • Antibody Size: The large size of antibodies (∼150 kDa) can impede penetration into solid tumors

  • Binding Affinity: For solid tumors, optimal binding affinity depends on the level of target expression

  • Transport and Clearance Mechanisms: The balance between antibody transport and clearance controls tumor penetration and efficacy

Analytical Approaches:

  • Mathematical analysis using scaling relationships helps understand tumor physiology and antibody penetration

  • Fundamental understanding of transport mechanisms and time scales is essential for predicting penetration distance through tumor tissue

These insights guide the development of next-generation ADCs with improved tumor penetration properties, potentially including smaller antibody formats or optimized binding kinetics.

How does Design of Experiments (DOE) methodology contribute to process development for ADCs?

Design of Experiments (DOE) is a critical methodology for developing robust ADC manufacturing processes:

Key Applications of DOE in ADC Development:

  • Analytical Method Development:

    • Facilitates the development of complex assays considering all three elements: antibody, payload, and conjugate

    • Helps develop methods for measuring critical quality attributes including SEC, DAR distribution, icIEF, residual free drug, and CE-SDS

  • Process Optimization:

    • Identifies critical process parameters that influence product quality

    • Establishes a robust Design Space to ensure consistent quality during scale-up

    • Enables faster and more reliable process development

DOE Implementation Approach:

  • Statistical Design Selection: Factorial designs (full or fractional) are typically used for early-phase development

  • Parameter Selection: Based on scientific understanding of the process and product

  • Scale-Down Model Development: Essential to avoid introducing undesired variability during execution

  • Specification Setting: Defines the "sweet spot" or Design Space and is used in robust setpoint calculations

A well-executed DOE strategy helps meet the goals of developing scientifically sound analytical methods, establishing process conditions to meet key quality attributes, understanding process robustness for safe scale-up, and establishing an effective control strategy .

What are the current challenges and future directions in ADC development?

Despite significant advances, several challenges remain in ADC development:

Current Challenges:

  • Heterogeneity: Variable drug loading and distribution affect product consistency and efficacy

  • Drug Resistance: Has impeded broader application of ADCs in clinical settings

  • Limited Tumor Penetration: Particularly problematic for solid tumors with poor vascularization

  • Immunogenicity: Early ADCs using murine antibodies elicited robust immune responses with some patients producing anti-human antibodies

Future Directions:

  • Multi-specific Drugs with Accurate DAR: Development of multi-specific ADCs with homogeneous drug loading to combat drug resistance

  • Next-Generation Antibody Engineering:

    • Fully humanized mAbs to reduce immunogenic responses

    • Biparatopic and bispecific formats to enhance target binding and internalization

  • Novel Conjugation Strategies: Site-specific conjugation technologies for homogeneous ADCs with precise drug loading

  • Expansion Beyond Oncology: Efforts to expand ADC applications for non-oncological indications and in combination therapies

These advancements aim to overcome current limitations, potentially expanding the therapeutic applications of ADCs and improving patient outcomes across multiple disease areas.

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.