HBV AG-1 antibody

Hepatitis B Virus (AD & AY Antigens) AG-1 for Capture ELISA, Mouse antibody
Shipped with Ice Packs
In Stock

Description

Key Mechanisms of Anti-HBsAg Antibodies

Antibodies targeting HBsAg employ multiple antiviral strategies:

  • Neutralization: Blocking viral entry into hepatocytes by binding to HBsAg on virions .

  • Immune Complex Clearance: Accelerating removal of viral particles via Fc-mediated phagocytosis .

  • Intracellular Blockade: Inhibiting HBsAg secretion from infected cells, reducing circulating antigen levels .

HuMAb006-11

  • Target: Conformational epitope on HBsAg (genotype-independent) .

  • Activity: Neutralizes HBV at nanomolar concentrations (>1,000x more potent than HBIG) .

  • Preclinical Data:

ParameterResult
Prophylactic efficacyHBV DNA undetectable in mice
Therapeutic reduction2.5–3.3 log10 HBV DNA decline

VIR-3434

  • Design: Engineered IgG1-vaccinal mAb with T-cell stimulation .

  • Phase 1 Results:

    • Single 18 mg dose reduced HBsAg by >1 log10 IU/mL within 4 weeks .

    • Combines neutralization with dendritic cell cross-presentation .

GIGA-2339

  • Composition: Recombinant polyclonal antibody with >1,000 anti-HBsAg clones .

  • Advantages:

    • Covers all HBV genotypes and escape mutants .

    • 2,000x higher potency vs plasma-derived HBIG in preclinical studies .

Clinical Trial Landscape

Recent Phase 1 trials highlight key developments:

AntibodySponsorPhaseKey OutcomeSource
GIGA-2339GigaGen1First patient dosed (Nov 2024)
VIR-3434Vir Bio1a4.6 log10 HBsAg reduction post-infusion

Biomarkers for Antibody Efficacy

Critical virological endpoints in antibody trials:

BiomarkerTarget ThresholdClinical Relevance
HBsAg<0.05 IU/mLFunctional cure criterion
HBV DNAUndetectable (LLOQ)Viral suppression
Anti-HBs>10 mIU/mLSeroprotection

Challenges and Future Directions

  • Antigen Load: High HBsAg levels (>1,000 IU/mL) reduce antibody efficacy .

  • Combination Therapy: Antibodies paired with siRNA (e.g., VIR-2218) show synergistic HBsAg reduction .

  • Durability: Transient antibody half-lives (7–21 days) necessitate repeated dosing or engineered Fc variants .

Product Specs

Introduction
Hepatitis B virus (HBV) is unique in that it is a non-retrovirus that utilizes reverse transcription during its replication cycle. While HIV, a retrovirus, also employs reverse transcription, it is not related to HBV. HBV infection begins with the virus binding to surface receptors on hepatocytes, followed by internalization. The viral core, containing partially double-stranded relaxed circular DNA (RC-DNA), migrates to the nucleus. Here, the RC-DNA is repaired to form covalently closed circular DNA (cccDNA), which serves as the template for viral RNA synthesis by the host cell's RNA polymerase II. One of the transcribed RNAs, the pregenomic RNA (pgRNA), is packaged into new capsids and reverse-transcribed into RC-DNA. These newly formed cores can either bud into the endoplasmic reticulum for envelope acquisition and export or return to the nucleus to replenish the cccDNA pool.
Formulation
Supplied at a concentration of 1 mg/ml in phosphate-buffered saline (PBS). The solution is ready to use after reconstitution.
Shipping Conditions
The antibody is shipped in lyophilized form at ambient temperature.
Storage Procedures
For long-term storage, keep the lyophilized antibody at 4°C in a dry environment. After reconstitution, use the antibody within a month. For extended storage, aliquot the reconstituted antibody and store at -20°C.
Solubility
Reconstitute the lyophilized antibody with sterile water (H2O). Mix gently by inverting the vial several times. Rinse the sides of the vial to ensure complete dissolution. Allow 30-60 seconds for the antibody to fully reconstitute before use.
Applications
This antibody is suitable for various applications, including: - Direct enzyme-linked immunosorbent assay (ELISA) - Western Blot - Immunoprecipitation - Immunohistochemistry
Titer
In a 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 an alkaline phosphatase-conjugated rabbit anti-mouse immunoglobulin G (IgG) secondary antibody (Jackson Laboratories).
Note
This antibody demonstrates reactivity against both the AD and AY antigens in direct ELISA assays and is suitable for use as a capture antibody in sandwich ELISAs when paired with an anti-hepatitis Ag-2 antibody.
Purification Method
Ion exchange column.
Type
Mouse antibody Monoclonal.
Clone
NYRHepB-1.
Immunogen
r.Hep B vaccine (BTG).
Ig Subclass
mouse IgG2b.

Q&A

What is the significance of HBsAg in HBV infection and research?

Hepatitis B surface antigen (HBsAg) is an antigenic protein found on the surface of HBV that serves as a critical marker for both acute and chronic infection. It indicates that a patient is infectious when detected in serum samples. HBsAg is particularly important in HBV research as it represents a primary target for neutralizing antibodies and is used extensively in diagnostic testing to identify infected individuals. The persistence of HBsAg in serum is associated with chronic infection, while its clearance and subsequent development of anti-HBs antibodies typically indicates recovery and immunity . The presence of elevated HBsAg levels contributes to the immunosuppressive environment in chronic HBV infection, making it a crucial target for therapeutic interventions aimed at functional cure .

What are the different types of serological markers used to monitor HBV infection status?

Several serological markers are used to assess HBV infection status, with each providing specific information about the stage of infection and immune response:

AbbreviationLab TestClinical Significance
HBsAgHepatitis B surface antigenIndicates active infection (acute or chronic); patient is infectious
Anti-HBsHepatitis B surface antibodyIndicates recovery/immunity from infection or successful vaccination
Anti-HBcTotal hepatitis B core antibodyDetects both IgM and IgG antibodies; indicates prior or ongoing infection
Anti-HBc IgMIgM antibody to hepatitis B coreIndicates recent or acute infection; not used for routine screening

These markers are crucial for determining infection status, monitoring treatment response, and assessing the risk of HBV reactivation in patients undergoing immunosuppressive therapy . Comprehensive serological testing, often followed by HBV DNA quantification when positive results are obtained, forms the cornerstone of clinical management strategies for HBV infection.

How do dual-targeting antibody-drug conjugates enhance therapeutic outcomes in HBV research?

Dual-targeting antibody-drug conjugates represent an innovative approach to HBV therapy by combining the specificity of HBV-neutralizing antibodies with immune-activating compounds. Recent research has developed immune-stimulating antibody conjugates (ISACs) consisting of Toll-like receptor 7/8 (TLR7/8) agonists linked to anti-HBsAg antibodies . For example, the 129G1-IMDQ conjugate combines the 129G1 antibody (which targets amino acids 137-151 of HBsAg) with the TLR7/8 agonist IMDQ via a non-cleavable linker .

This dual-targeting approach offers several advantages over traditional antibody therapy. When tested in AAV/HBV mouse models, 129G1-IMDQ demonstrated superior efficacy in reducing serum HBsAg levels compared to the antibody alone or a simple combination of antibody plus IMDQ . More importantly, the conjugate elicited strong and sustained anti-HBsAg antibody responses that persisted for at least 42 days after short-term treatment, suggesting potential for functional cure of chronic HBV infection . The mechanism involves both direct viral neutralization and enhanced immune activation, with the conjugate significantly upregulating activation markers CD80 and CD86 on dendritic cells and macrophages compared to antibody alone .

What novel mechanisms have been discovered regarding how antibodies block HBV infection?

Recent research has revealed previously unrecognized mechanisms by which antibodies can inhibit HBV infection. While antibodies were traditionally thought to function primarily by blocking viral entry and accelerating viral clearance from circulation, mathematical modeling combined with in vitro and in vivo studies has identified an additional mechanism: prolonged blocking of virion release from infected cells .

During clinical studies with HepeX-B (a combination of two human monoclonal anti-HBs antibodies, HBV-17 and HBV-19), researchers observed kinetic profiles of HBV DNA and HBsAg decline that suggested partial blocking of virion release from infected hepatocytes . This observation was subsequently validated in vitro using HBsAg-producing cells, which showed cellular uptake of antibodies and intracellular accumulation of viral particles when treated with anti-HBs antibodies . Notably, this blocking effect on HBsAg secretion continued even after antibodies were removed from the cell culture medium, indicating a prolonged therapeutic effect . This discovery has significant implications for designing new therapies for chronic HBV infection and potentially for approaches to other viral infections as well.

What techniques are employed for engineering and purifying HBV-targeting antibodies?

The engineering and purification of HBV-targeting antibodies involve several sophisticated techniques to ensure specificity, affinity, and functional activity. Based on current research methodologies:

  • Antibody generation: Hybridoma clones producing anti-HBV antibodies can be obtained through the iliac lymph node method, as demonstrated in the development of anti-preS1 antibodies . Alternatively, genes encoding specific antibodies (such as 129G1) can be inserted into recombinant mammalian expression vectors fused to appropriate Fc regions .

  • Expression systems: The Expi293™ Expression System has been successfully employed for producing recombinant HBV antibodies, offering high yield and proper protein folding .

  • Purification: Protein A chromatography is commonly used for purifying monoclonal antibodies targeting HBV components . This technique exploits the specific binding of most antibody Fc regions to Protein A.

  • Conjugation chemistry: For developing antibody-drug conjugates, specific crosslinkers such as SMCC (succinimidyl-4-[N-maleimidomethyl]cyclohexane-1-carboxylate) can be used to create amine-to-sulfhydryl conjugations . The process typically involves a two-step approach: first modifying antibody lysine residues using heterobifunctional cross-linkers like N-succinimidyl S-acetylthioacetate (SATA), followed by deprotection of the acetylated thiol with hydroxylamine to yield a reactive thiol for conjugation with maleimide-containing compounds .

  • Verification of conjugation: Hydrophobic interaction chromatography (HIC) can be employed to verify protein conjugation efficiency, with delayed elution peaks indicating successful conjugation .

How are animal models used to evaluate HBV antibody efficacy?

Animal models play a crucial role in evaluating the efficacy of HBV antibodies before clinical testing. The AAV/HBV mouse model has emerged as a valuable system for this purpose:

C57BL/6J mice (aged 4-6 weeks) can be infected with adeno-associated virus (AAV) harboring 1.3 copies of the HBV genome (genotype B, serotype adw) packaged in AAV serotype 8 capsids . Following tail vein injection of the AAV-HBV1.3 construct, mice require a minimum 5-week incubation period to establish stable HBV infection . This model allows researchers to:

  • Assess the dynamics of HBsAg levels and anti-HBsAg antibody titers by collecting serial blood samples from the retro-orbital sinus at specific time intervals.

  • Evaluate therapeutic interventions through systematically scheduled antibody administrations (e.g., on Days 1, 3, 5, and 7) followed by blood sampling at multiple time points (e.g., Days 7, 8, 11, 13, 15, 18, 21, 25, 32, and 42) .

  • Compare the efficacy of different antibody formulations (e.g., antibody alone, antibody plus adjuvant, or antibody-drug conjugates) in reducing viral antigen levels and stimulating host immune responses.

The AAV/HBV mouse model is particularly valuable for testing antibody therapies targeting high viral loads (~10,000 IU/ml), simulating conditions in chronic HBV carriers .

What protocols are recommended for epitope mapping of anti-HBV antibodies?

Epitope mapping is essential for characterizing the binding sites of anti-HBV antibodies, which directly influences their neutralizing potential and therapeutic applications. For anti-HBsAg antibodies, both linear and conformational epitopes have been identified, requiring different mapping approaches.

For antibodies targeting linear epitopes, such as E6F6 (which binds to amino acids 119-125 of HBsAg) and 129G1 (which targets amino acids 137-151 in the 'second loop'), peptide-based approaches can be effective . These typically involve synthesizing overlapping peptides spanning the target antigen sequence and testing antibody binding to each fragment.

For antibodies recognizing conformational epitopes, such as HBV-17 (one component of HepeX-B), more complex approaches are needed that preserve the three-dimensional structure of the antigen . These may include:

  • Competition binding assays with antibodies of known epitope specificity

  • Hydrogen-deuterium exchange mass spectrometry

  • X-ray crystallography of antibody-antigen complexes

  • Mutagenesis studies where specific amino acids are altered to assess their impact on antibody binding

Importantly, epitope mapping should include assessment across different HBV genotypes and common mutant variants. For example, the 129G1 antibody binds to all HBsAg across HBV genotypes, except for certain major hydrophilic region mutations (D144A, D145A, and G145R) . This information is crucial for predicting therapeutic coverage and potential escape mechanisms.

How can antibody-mediated activation of immune cells be assessed in HBV research?

Evaluating the immunomodulatory effects of anti-HBV antibodies is critical for understanding their therapeutic potential beyond direct viral neutralization. Several methodological approaches have been employed:

  • Flow cytometry analysis of phagocytosis: HBsAg can be labeled with a fluorescent marker (e.g., FITC) and incubated with antibodies before addition to immune cells such as dendritic cells (DCs) and macrophages. Flow cytometry can then quantify the percentage of cells that have phagocytosed the immune complexes . In one study, 129G1-IMDQ and 129G1 mediated phagocytosis in approximately 20% of splenic DCs and 50% of macrophages, while cells exhibited minimal phagocytosis (2% and 10%, respectively) in the absence of antibodies .

  • Assessment of activation markers: Following exposure to antibody-antigen complexes, immune cells can be analyzed for the expression of activation markers such as CD80 and CD86 on DCs and macrophages . Upregulation of these markers indicates successful immune cell activation, which is essential for initiating effective adaptive immune responses.

  • Cytokine profiling: Measurement of cytokine production (e.g., IL-12, TNF-α, IFN-γ) by activated immune cells provides insight into the quality and type of immune response being generated.

  • In vivo immune response assessment: The ultimate test of immune activation is the generation of specific anti-HBsAg antibodies in animal models. Serial blood sampling can be used to monitor antibody titers over time, with sustained high titers indicating successful breaking of immune tolerance .

When comparing different antibody formulations (e.g., 129G1-IMDQ vs. 129G1 alone), these methods can reveal the added value of immunostimulatory components like TLR7/8 agonists in enhancing immune cell activation and subsequent adaptive responses .

What methods are used for developing and evaluating antibody-drug conjugates for HBV therapy?

The development and evaluation of antibody-drug conjugates for HBV therapy involve several specialized techniques:

  • Conjugation chemistry and verification:

    • Selection of appropriate conjugatable small molecules (e.g., IMDQ) and non-cleavable linkers (e.g., SMCC)

    • Two-step conjugation process: (a) modification of antibody lysine residues with SATA, followed by (b) deprotection with hydroxylamine to yield reactive thiols for reaction with maleimide-containing compounds

    • Verification of conjugation efficiency using hydrophobic interaction chromatography (HIC), with successfully conjugated proteins showing delayed elution peaks

    • Assessment of binding efficacy post-conjugation using ELISA to ensure the EC50 value remains comparable to the unconjugated antibody

  • Functional testing:

    • Evaluation of HBsAg clearance through phagocytosis assays using fluorescently labeled antigens

    • Assessment of immune cell activation by measuring expression of activation markers (CD80, CD86) on dendritic cells and macrophages

    • Comparison of conjugated antibodies with unconjugated counterparts and simple mixtures of antibody plus small molecule to demonstrate the advantages of chemical conjugation

  • In vivo efficacy evaluation:

    • Establishment of AAV/HBV mouse models with stable HBV infection (minimum 5-week incubation period)

    • Administration of treatment according to defined schedules (e.g., intraperitoneal injections every other day for a total of four doses)

    • Serial blood sampling to monitor HBsAg levels and anti-HBsAg antibody titers

    • Long-term follow-up (e.g., 42 days) to assess the durability of therapeutic effects, which is crucial for determining potential for functional cure

Research has shown that properly designed ISACs like 129G1-IMDQ can significantly reduce serum HBsAg levels and elicit robust, lasting anti-HBsAg immune responses after short-term treatment, offering advantages over both antibody monotherapy and simple combinations of antibody plus immunostimulatory molecule .

How can preS1 measurement be utilized as a biomarker in chronic HBV infection?

The preS1 region plays an essential role in HBV infection, and recent research has developed methods to measure serum preS1 levels as a potential biomarker in chronic HBV-infected patients. A specific antibody (3-55) that binds to amino acids 38-47 of preS1 with high affinity has been developed using the iliac lymph node method to generate hybridoma clones . This antibody has been incorporated into an enzyme-linked immunosorbent assay (ELISA) system capable of measuring serum preS1 levels in clinical samples .

Clinical studies have shown that serum preS1 levels correlate with other established markers of HBV infection, including HBsAg, HBV core-related antigen (HBcrAg), and HBV DNA levels . Importantly, among HBeAg-negative patients who don't meet conventional criteria for antiviral therapy (HBV DNA <3.3 log IU/mL or alanine aminotransferase ≤30 U/L), preS1 levels were significantly higher in individuals who subsequently progressed to requiring antiviral therapy compared to those who maintained their status over a three-year period (p<0.01) . This suggests that preS1 measurement could serve as a novel predictive tool for identifying patients who might benefit from earlier therapeutic intervention, potentially allowing for more personalized management strategies in chronic HBV infection.

What are the considerations for antibody-based therapies in preventing HBV reactivation?

HBV reactivation (HBVr) is a significant concern in patients undergoing immunosuppressive therapy, and antibody-based approaches could potentially play a role in prevention strategies. Current screening recommendations involve testing for HBsAg and anti-HBc, followed by HBV DNA quantification if either is positive . Prophylactic antiviral therapy is currently the standard approach for preventing HBVr in high-risk patients.

Therapeutic antibodies targeting HBV could potentially complement existing prophylactic strategies by:

  • Directly neutralizing circulating virus, preventing infection of new hepatocytes

  • Blocking the release of virions from infected cells, as demonstrated with HepeX-B antibody therapy

  • Enhancing immune-mediated clearance of infected cells when used in conjunction with immune-stimulating agents

For patients at highest risk (e.g., HBsAg-positive individuals receiving B-cell depleting therapies like rituximab), the duration of monitoring after cessation of prophylaxis is typically 6-12 months, followed by occasional long-term monitoring . Novel antibody-based approaches could potentially influence this monitoring paradigm by providing additional protective mechanisms beyond those offered by conventional nucleos(t)ide analogs.

Future research should evaluate whether dual-targeting antibody conjugates like 129G1-IMDQ, which have demonstrated sustained anti-HBsAg responses in animal models , could play a role in preventing HBVr in immunocompromised hosts, potentially offering advantages in terms of dosing frequency and durability of protection.

How might antibody engineering approaches advance HBV therapeutic development?

Antibody engineering technologies offer numerous opportunities to enhance the therapeutic potential of anti-HBV antibodies. Future research directions may include:

  • Bispecific antibodies: Developing antibodies that simultaneously target different epitopes on HBsAg or multiple HBV antigens (e.g., HBsAg and HBcAg) could improve neutralization breadth and potency while reducing the potential for viral escape.

  • Fc engineering: Modifying the Fc region of anti-HBV antibodies could enhance effector functions such as antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), potentially improving the clearance of infected hepatocytes.

  • Novel conjugation approaches: Building on the success of TLR7/8 agonist conjugation , researchers could explore conjugation of anti-HBV antibodies to other immunomodulatory molecules or targeted delivery systems to improve therapeutic efficacy while minimizing systemic toxicity.

  • Half-life extension technologies: Incorporating half-life extension strategies (e.g., Fc mutations, albumin fusion) could reduce dosing frequency while maintaining therapeutic antibody levels, potentially improving patient compliance and reducing treatment costs.

  • Combination approaches: Investigating synergistic effects between antibody therapies and other HBV treatment modalities, such as nucleos(t)ide analogs, RNA interference technologies, or therapeutic vaccines, could lead to more effective functional cure strategies.

The developing field of antibody-based therapies for chronic HBV infection holds significant promise, particularly as our understanding of antibody mechanisms expands beyond simple virus neutralization to include blocking of virion release and immune system modulation through targeted delivery of immunostimulatory compounds .

Product Science Overview

Introduction

Hepatitis B Virus (HBV) is a significant global health concern, affecting millions of people worldwide. The virus can cause both acute and chronic liver infections, leading to severe liver diseases such as cirrhosis and hepatocellular carcinoma. The detection and quantification of HBV antigens are crucial for diagnosing and monitoring the infection. One of the methods used for this purpose is the Enzyme-Linked Immunosorbent Assay (ELISA), specifically designed to capture HBV antigens using antibodies.

Hepatitis B Virus (HBV)

HBV is a DNA virus belonging to the Hepadnaviridae family. It has a partially double-stranded DNA genome and replicates through an RNA intermediate. The virus primarily infects liver cells (hepatocytes) and can integrate into the host genome, leading to chronic infection. HBV has several antigens, including the surface antigen (HBsAg), core antigen (HBcAg), and e antigen (HBeAg), which are used as markers for infection and immune response.

AD & AY Antigens

The AD and AY antigens refer to specific subtypes of the HBV surface antigen (HBsAg). These subtypes are determined by variations in the amino acid sequences of the HBsAg protein. The AD subtype is more prevalent in certain geographical regions, while the AY subtype is found in others. These variations can affect the antigenicity and immunogenicity of the virus, making it essential to detect and differentiate between these subtypes for accurate diagnosis and treatment.

Capture ELISA

Capture ELISA is a widely used technique for detecting and quantifying antigens in various samples, including blood, serum, and plasma. The assay involves capturing the target antigen using a specific antibody immobilized on a solid surface, followed by detection using a secondary antibody conjugated to an enzyme. The enzyme catalyzes a colorimetric reaction, producing a measurable signal proportional to the amount of antigen present.

Mouse Antibody AG-1

The AG-1 mouse antibody is a monoclonal antibody specifically designed to capture the AD and AY antigens of HBV in a capture ELISA. Monoclonal antibodies are produced by a single clone of B cells and are highly specific to a particular epitope on the antigen. The AG-1 antibody binds to the AD and AY subtypes of HBsAg with high affinity, ensuring accurate and sensitive detection of these antigens in clinical samples.

Applications

The Hepatitis B Virus (AD & AY Antigens) AG-1 for Capture ELISA is used in various applications, including:

  • Diagnosis: Detecting HBV infection in patients by identifying the presence of AD and AY antigens in blood samples.
  • Monitoring: Assessing the viral load and progression of HBV infection in chronic carriers.
  • Research: Studying the epidemiology and molecular biology of HBV, including the distribution and prevalence of different 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.