HBV AG-2 antibody

Hepatitis B Virus (AD & AY Antigens) AG-2 for Capture ELISA, Mouse antibody
Shipped with Ice Packs
In Stock

Description

HBV Antibody Classification and Terminology

The hepatitis B virus (HBV) immune response involves three principal antibody types:

Antibody TypeTarget AntigenClinical SignificanceDetection WindowProtective Role
Anti-HBsHBsAg (surface)Vaccination success≥1 month post-vaccinationNeutralizes viral particles
Anti-HBcHBcAg (core)Current/past infectionLifelong persistenceNo neutralization
Anti-HBeHBeAgReduced infectivityDuring seroconversionNon-protective

No "AG-2" classification exists in standard HBV serology systems. The nomenclature "AG" (antigen) typically refers to viral proteins, not antibodies.

Monoclonal Antibodies Targeting the "a" Determinant

Several human monoclonal antibodies demonstrate AG-like properties through their binding to the major hydrophilic region (MHR) of HBsAg:

AntibodyEpitope SpecificityNeutralization Potency (IC₅₀)Clinical Application
ADRI-2F3Conformational "a"0.1 μg/mLPost-transplant prophylaxis
HuMAb006-11Genotype-independent0.08 μg/mLTherapeutic candidate
Bc1.187-GLPreS1/S2 domains0.2 μg/mLBroad neutralization

These antibodies achieve >99% viral suppression in vitro and prevent hepatocyte entry through steric hindrance .

Functional Performance

  • Viral suppression: Antibodies like ADRI-2F3 reduce serum HBV DNA by 4.7 log₁₀ IU/mL in Tupaia models .

  • Cross-genotype efficacy: HuMAb006-11 neutralizes genotypes A-D with <0.5 log₁₀ variance in EC₅₀ values .

Critical Knowledge Gaps

  1. Nomenclature inconsistencies: The term "AG-2" doesn't align with WHO-standardized HBV antibody terminology .

  2. Commercial availability: No FDA-approved HBV monoclonal antibodies exist as of March 2025, though phase III trials are ongoing for ADRI-2F3 derivatives .

  3. Diagnostic relevance: Concurrent HBsAg/anti-HBs positivity occurs in 7% of chronic HBV cases and correlates with 2.3× higher HCC risk .

Recommendations for Further Investigation

  1. Verify the antibody's molecular target (e.g., PreS1, MHR, or polymerase domains).

  2. Cross-reference with HBV antibody databases (PDB IDs: 6V42, 7C4L, 8HBS).

  3. Explore patent filings using ESPACENET or WIPO databases for proprietary antibody designations.

Product Specs

Introduction
Hepatitis B virus (HBV) is unusual among DNA viruses in that it uses reverse transcription during its replication cycle. HBV infects liver cells (hepatocytes), entering the cell through a receptor called sodium taurocholate cotransporting polypeptide (NTCP). Once inside the cell, the virus travels to the nucleus and converts its DNA into a closed circular form, which then serves as a template for the production of viral proteins and RNA. One of the RNA molecules produced is the pregenomic RNA (pgRNA), which serves as the template for reverse transcription. The pgRNA is packaged into new viral particles, and within these particles, it is reverse transcribed into DNA, generating new copies of the viral genome. These new viral particles can then go on to infect other cells.
Formulation
Supplied as a 1 mg/ml solution in phosphate-buffered saline (PBS).
Shipping Conditions
Shipped at ambient temperature in lyophilized form.
Storage Procedures
Store lyophilized antibody at 4°C in a dry environment for long-term storage. For use after reconstitution, aliquot and store at -20°C if not intended for use within one month.
Solubility
Reconstitute with deionized water (H2O). Gently mix the solution, ensuring to wash the sides of the vial, and allow 30-60 seconds for complete reconstitution before use.
Applications
Suitable applications include direct enzyme-linked immunosorbent assay (ELISA), Western blotting, immunoprecipitation, and immunohistochemistry.
Titer
In direct ELISA against recombinant hepatitis B vaccine, a 1:10,000 dilution of the antibody produces an optical density (O.D.) of 0.5 when using alkaline phosphatase-conjugated rabbit anti-mouse immunoglobulin G (Jackson Laboratories).
Note
This antibody can detect both the AD and AY antigens in direct ELISA and is compatible with capture ELISA when used with an anti-hepatitis B surface antigen (HBsAg) antibody.
Purification Method
Ion exchange column.
Type
Mouse antibody Monoclonal.
Clone
NYRHepB-2.
Immunogen
r.Hep B vaccine (BTG).
Ig Subclass
mouse IgG1.

Q&A

What is the structural relationship between HBsAg and anti-HBs antibodies?

HBsAg is a protein found on the surface of the hepatitis B virus that plays a crucial role in viral infection and immune recognition. Anti-HBs antibodies are formed in response to HBsAg exposure, either through vaccination or natural infection recovery. These antibodies target specific epitopes on the HBsAg protein, with the most common epitope being the "a" determinant within the major hydrophilic region (MHR). Research has identified several specific binding regions, including the linear epitope at amino acids 119-125 targeted by E6F6 monoclonal antibody and the 'second loop' linear epitope at amino acids 137-151 recognized by the 129G1 monoclonal antibody . The molecular interaction between HBsAg and its antibodies involves conformational recognition that depends on the three-dimensional structure of these proteins.

How do we differentiate between vaccine-induced and infection-recovery anti-HBs?

Methodologically, differentiating between these two origins of anti-HBs requires analysis of additional serological markers:

Origin of Anti-HBsAnti-HBcAnti-HBc IgMHBsAg HistoryOther Markers
VaccinationNegativeNegativeNever positiveNo other HBV markers
Natural Infection RecoveryPositiveNegative (if recovered)Previously positivePossibly anti-HBe positive

For research purposes, the specificity profile of anti-HBs can provide additional clues. Vaccine-induced antibodies typically target the specific HBsAg subtype used in the vaccine, while infection-recovery antibodies may have broader reactivity patterns across multiple epitopes. Conducting epitope mapping studies using synthetic peptides or competition assays can help characterize the antibody response origin .

What are the optimal protocols for isolating and characterizing HBsAg-specific monoclonal antibodies?

The isolation and characterization of HBsAg-specific monoclonal antibodies involve several methodological steps:

  • Source selection: Peripheral blood mononuclear cells from recovered HBV patients or vaccinated individuals provide optimal starting material.

  • B-cell isolation: Techniques include magnetic bead separation with HBsAg-coated beads or fluorescence-activated cell sorting (FACS) using labeled HBsAg.

  • Immortalization/cloning: Either through hybridoma technology or direct B-cell receptor (BCR) sequencing and recombinant expression.

  • Characterization protocols:

    • Binding affinity: Surface plasmon resonance (SPR) or enzyme-linked immunosorbent assay (ELISA)

    • Epitope mapping: Peptide arrays, competitive binding assays, or hydrogen-deuterium exchange mass spectrometry

    • Neutralization capacity: In vitro neutralization assays using HBV susceptible cell lines (HepaRG, HepG2-NTCP)

    • Effector functions: Fc-mediated activities including antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC)

When engineering recombinant antibodies, expression systems like the Expi293™ Expression System have proven effective for producing functional anti-HBsAg antibodies, as demonstrated in the production of the 129G1 antibody with mouse IgG2a Fc region .

How can researchers accurately assess HBV viral mutations that affect antibody recognition?

To accurately assess HBV mutations affecting antibody recognition, researchers should employ a systematic approach:

  • Sample collection: Obtain sequential samples from patients with chronic HBV infection, particularly focusing on those with unusual serological profiles such as concurrent HBsAg and anti-HBs.

  • HBV DNA amplification: Use nested PCR with primers targeting conserved regions flanking the S gene.

  • Deep sequencing methodologies:

    • Clonal sequencing: Amplify the S gene, clone into vectors, and sequence multiple clones (minimum 15 clones per patient as performed in research protocols)

    • Next-generation sequencing: Provides greater depth for detecting minor viral populations

  • Bioinformatic analysis:

    • Alignment with reference sequences

    • Identification of amino acid substitutions, particularly in known antibody binding regions

    • Frequency analysis of mutations within the viral quasispecies

  • Functional validation:

    • In vitro binding assays using recombinant HBsAg variants

    • Neutralization escape assays

    • Western blot or immunoprecipitation to confirm altered antibody binding

When analyzing potential escape mutants, researchers should focus on amino acid substitutions within the major hydrophilic region, particularly the "a" determinant (amino acids 124-147) and compare prevalence of these mutations between patients with and without anti-HBs antibodies .

What mechanisms explain the paradoxical coexistence of HBsAg and anti-HBs in chronic HBV patients?

The coexistence of HBsAg and anti-HBs in chronic HBV patients has been observed in approximately 4.9-9% of cases . Several potential mechanisms have been proposed:

These findings challenge the hypothesis that viral escape variants emerge as a response to antibody pressure and suggest alternative mechanisms for this serological pattern.

How do antibody-drug conjugates targeting HBsAg enhance antiviral responses compared to unconjugated antibodies?

Antibody-drug conjugates (ADCs) represent an innovative approach to HBV therapy, combining the specificity of anti-HBsAg antibodies with immune-activating agents. Research with the 129G1-IMDQ conjugate demonstrates several mechanisms of enhanced efficacy:

  • Dual targeting advantage: ADCs employing TLR7/8 agonists linked to anti-HBsAg antibodies show significantly improved HBsAg clearance compared to antibody treatment alone:

    • The antibody component binds specifically to HBsAg

    • The TLR7/8 agonist activates innate immune responses in antigen-presenting cells

  • Enhanced immunological mechanisms:

    • Improved immune complex formation and clearance

    • Fc-mediated clustering prompting antibody-dependent phagocytosis

    • Enhanced hepatic retention of immune complexes

    • Activation of innate immunity via TLR signaling

    • Improved antigen presentation and adaptive immune response stimulation

  • Experimental evidence: In AAV/HBV mouse models, 129G1-IMDQ treatment demonstrated:

    • Significant reduction in HBsAg levels

    • Induction of robust and sustained anti-HBsAg immune responses after short-term treatment

    • Superior efficacy compared to unconjugated antibody administration

The conjugation chemistry is critical, with successful approaches including non-cleavable linkers and specific conjugation strategies like SMCC-mediated maleimide-thiol coupling .

What are the critical parameters for evaluating novel anti-HBsAg therapeutic antibodies?

When evaluating novel anti-HBsAg therapeutic antibodies, researchers should assess:

  • Epitope specificity and breadth:

    • Binding to conserved versus variable regions of HBsAg

    • Cross-reactivity across HBV genotypes

    • Resistance to known escape mutations (particularly D144A, D145A, and G145R mutations)

  • Functional characteristics:

    • Neutralization potency (IC50 values)

    • Ability to clear circulating HBsAg

    • Fc-dependent effector functions

    • Immune complex formation and clearance kinetics

  • Pharmacokinetic considerations:

    • Half-life in circulation

    • Tissue distribution, particularly hepatic accumulation

    • Potential for immunogenicity

  • For antibody-drug conjugates:

    • Drug-to-antibody ratio calculation

    • Linker stability and potential for premature release

    • Retained binding activity post-conjugation (EC50 comparison)

    • TLR7/8 activation potency

    • Fc receptor binding affinity preservation

  • In vivo efficacy parameters:

    • HBsAg reduction magnitude and duration

    • HBeAg seroconversion rates

    • HBV DNA suppression

    • Anti-HBs induction

    • Liver enzyme normalization

    • Histological improvement

How do HBsAg sequence variations across HBV genotypes impact antibody recognition and therapeutic efficacy?

HBV genetic diversity manifests as eight major genotypes (A-H) with significant implications for antibody recognition:

  • Genotypic variation in the S gene:

    • Amino acid variations within and outside the major hydrophilic region

    • Differential glycosylation patterns affecting epitope presentation

    • Genotype-specific substitutions affecting antibody binding

  • Impact on therapeutic antibodies:

    • Antibodies targeting conserved linear epitopes (like E6F6 targeting amino acids 119-125) may have broader cross-genotype reactivity

    • Antibodies recognizing conformational epitopes may show genotype-restricted efficacy

    • The 129G1 antibody binds to all HBsAg across HBV genotypes, with exceptions for specific mutations in the major hydrophilic region

  • Research approaches to address genotypic variation:

    • Comprehensive binding analysis using recombinant HBsAg from all major genotypes

    • Identification of conserved epitopes as therapeutic targets

    • Development of antibody cocktails targeting multiple epitopes

    • Engineering of broadly neutralizing antibodies through structure-guided design

Understanding these variations is critical for developing globally effective therapeutic antibodies and designing clinical trials with appropriate patient stratification.

What are the optimal animal models for evaluating anti-HBsAg antibody therapies?

The selection of appropriate animal models for evaluating anti-HBsAg antibody therapies is critical for translational research:

  • Mouse models:

    • AAV/HBV mice: Adeno-associated virus vectors containing 1.3 copies of the HBV genome (such as genotype B, serotype adw) packaged in AAV serotype 8 capsids delivered via tail vein injection. This model produces stable HBV infection after a 5-week incubation period and permits evaluation of serum HBsAg dynamics and anti-HBsAg antibody responses .

    • Hydrodynamic injection model: Rapid injection of HBV plasmid DNA resulting in transient expression

    • Transgenic HBV mice: Constitutive HBV replication but limited immune response

  • Key experimental design considerations:

    • Group size: Minimum six mice per experimental group for statistical power

    • Controls: Include normal control IgG and IgG conjugated with immune activators as controls

    • Administration schedule: For antibody therapies, 4-dose regimens (Days 1, 3, 5, and 7) have shown efficacy

    • Sampling schedule: Regular blood sampling (e.g., Days 7, 8, 11, 13, 15, 18, 21, 25, 32, and 42) for monitoring HBsAg and antibody dynamics

  • Limitations of current models:

    • Species differences in immune system components (e.g., murine TLR8 has questionable functionality)

    • Differences in hepatocyte infection mechanisms

    • Lack of chronic inflammation modeling

These considerations highlight the need for careful translation of findings to human contexts and the importance of multiple model systems for comprehensive evaluation.

How should researchers interpret contradictory data between in vitro neutralization and in vivo efficacy of anti-HBsAg antibodies?

Researchers frequently encounter discrepancies between in vitro and in vivo results when studying anti-HBsAg antibodies:

  • Common contradictions:

    • High in vitro binding affinity without corresponding in vivo HBsAg clearance

    • Effective neutralization in cell culture with limited impact on viral load in animal models

    • Variable correlation between antibody-dependent cellular cytotoxicity (ADCC) activity and therapeutic outcomes

  • Interpretation framework:

    • Pharmacokinetic factors: Assess antibody half-life, tissue distribution, and potential sequestration

    • Immune complex dynamics: Investigate clearance mechanisms and potential immune complex deposition

    • Epitope accessibility: Consider differences in epitope presentation between in vitro systems and in vivo infection

    • Host immune status: Evaluate the contribution of host immune factors to antibody efficacy

    • Viral factors: Assess potential selection for escape variants during treatment

  • Experimental approaches to resolve contradictions:

    • Combine in vitro neutralization with Fc receptor binding assays

    • Assess antibody-mediated clearance in physiologically relevant systems

    • Evaluate immune activation markers in parallel with virological endpoints

    • Perform sequential viral sequencing during antibody therapy

    • Investigate the impact of antibody treatment on intrahepatic viral forms

  • Translational considerations:

    • Human liver chimeric mouse models may provide better predictive value

    • Ex vivo systems using primary human hepatocytes can bridge the gap between in vitro and in vivo studies

    • Careful consideration of species differences in Fc receptors and immune effector functions

When interpreting contradictory data, researchers should consider that anti-HBsAg antibodies may function through multiple mechanisms beyond direct neutralization, including immune complex formation, enhanced antigen presentation, and modulation of innate immune responses .

Product Science Overview

Introduction

Hepatitis B Virus (HBV) is a major global health concern, causing both acute and chronic liver diseases. The virus is characterized by its surface antigens, primarily the AD and AY subtypes, which are crucial for diagnostic and research purposes. The AG-2 mouse antibody is specifically designed for capture ELISA, a technique widely used to detect and quantify these antigens.

Hepatitis B Virus (HBV)

HBV is a DNA virus belonging to the Hepadnaviridae family. It primarily infects liver cells, leading to inflammation and potentially severe liver damage. The virus has several antigens, including the Hepatitis B surface antigen (HBsAg), which is a key marker for infection. The AD and AY subtypes of HBsAg are the most common and are used in various diagnostic assays .

AD & AY Antigens

The AD and AY antigens are subtypes of the Hepatitis B surface antigen (HBsAg). These antigens are part of the viral envelope and are crucial for the virus’s ability to infect host cells. The presence of these antigens in the blood is an indicator of active HBV infection. The AD subtype is more prevalent in certain geographical regions, while the AY subtype is more common in others .

Capture ELISA

Enzyme-Linked Immunosorbent Assay (ELISA) is a widely used technique for detecting and quantifying antigens and antibodies. In a capture ELISA, the target antigen is “captured” by a specific antibody coated on a microplate. The AG-2 mouse antibody is used in this context to capture the AD and AY antigens of HBV. This method is highly sensitive and specific, making it ideal for diagnostic and research applications .

AG-2 Mouse Antibody

The AG-2 mouse antibody is a monoclonal antibody specifically designed to bind to the AD and AY antigens of HBV. It is produced by immunizing mice with these antigens and then isolating the specific antibody-producing cells. The antibody is then purified and used in capture ELISA to detect the presence of HBV antigens in patient samples .

Applications

The AG-2 mouse antibody for capture ELISA is used in various applications, including:

  • Diagnostic Testing: Detecting HBV infection in patients by identifying the presence of AD and AY antigens in blood samples.
  • Research: Studying the epidemiology and pathogenesis of HBV by analyzing the distribution and prevalence of different HBsAg subtypes.
  • Vaccine Development: Evaluating the efficacy of HBV vaccines by measuring the immune response to different HBsAg subtypes .

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 2024 Thebiotek. All Rights Reserved.