Dengue Control Antibody

Monoclonal Mouse Anti Dengue Control for Lateral Flow Test
Shipped with Ice Packs
In Stock

Description

Definition and Targets of Dengue Control Antibodies

DCAs primarily target:

  • Envelope (E) protein domains: Domain III (DIII) for serotype-specific neutralization and domains I/II (DI/DII) for cross-reactive responses .

  • Non-structural protein 1 (NS1): A secreted glycoprotein linked to vascular leakage and severe dengue .

  • Quaternary epitopes: Structural interfaces between E protein dimers on the viral surface .

Neutralization Strategies

  • Domain III-specific antibodies (e.g., 3E31, SIgN-3C): Block viral attachment to host cells by binding conserved residues (e.g., Gln368, His317) .

  • Fusion loop-targeting antibodies (e.g., B3B9): Inhibit membrane fusion during viral entry but risk ADE at sub-neutralizing concentrations .

  • NS1-targeting antibodies (e.g., 2B7): Prevent endothelial barrier disruption by blocking NS1 interaction with host cells .

ADE Mitigation

  • Fc modifications: LALA mutations (Leu234Ala/Leu235Ala) or Asn297 substitutions in the antibody Fc region reduce FcγR binding, eliminating ADE .

  • Bispecific antibodies: Combine neutralizing and ADE-blocking epitopes .

Key Antibodies and Efficacy

AntibodyTargetNeutralization Potency (PRNT₅₀)ADE RiskSource
2B7NS1 proteinN/A (blocks pathogenesis)None
SIgN-3C-LALAQuaternary epitope0.001–3.08 nM (all serotypes)None
3E31DIII AB loop0.5–1.2 µg/mLLow
B3B9 (N297Q)Fusion loop0.1–0.5 µg/mLEliminated

Cross-Reactivity vs. Serotype Specificity

FeatureCross-Reactive AntibodiesSerotype-Specific Antibodies
Target EpitopesDI/II fusion loop, bc loop DIII, quaternary epitopes
NeutralizationWeak-to-moderate (broad)Potent (narrow)
ADE RiskHigh at sub-neutralizing doses Low with Fc engineering

Clinical and Preclinical Outcomes

  • 2B7: Reduced vascular leakage in mice by 90% and blocked DENV/Zika cross-reactivity .

  • SIgN-3C-LALA: Decreased viremia by 10⁴–10⁵ PFU/mL in mice for all serotypes .

  • TV003/TV005 vaccines: Stimulated tetravalent neutralizing antibodies in 76–90% of recipients .

Challenges and Innovations

  • ADE Limitations: Sub-neutralizing antibodies enhance infection via FcγR-mediated uptake .

  • Engineering Solutions:

    • scFv/Fab formats: Remove Fc regions to avoid ADE .

    • Epitope focusing: Target conserved regions (e.g., NS1 or quaternary epitopes) .

Future Directions

  • Universal antibody cocktails: Combine cross-reactive (e.g., 3E31) and serotype-specific antibodies .

  • Phase II trials: Evaluate Fc-modified antibodies in endemic regions .

Product Specs

Description
Monoclonal Mouse Anti Dengue Control for Lateral Flow Test. This antibody is used to prepare a control line for dengue IgG/IgM rapid tests. The recommended coating concentration is 0.4-0.5mg/ml, 1µl/cm.
Purity
90% purity as determined by SDS-PAGE and Coomassie staining.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
10mM phosphate buffered saline at pH 7.2.
Stability
For short-term storage (up to 2-4 weeks), store at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Applications
Immunoassays.
Purification Method

Purified monoclonal IgG by protein A chromatography.

Q&A

How do antibody responses to dengue virus differ from other viral infections?

Unlike infections such as measles that confer lifelong immunity, dengue presents a unique immunological challenge. When infected by one of the four dengue virus serotypes (DENV-1 through DENV-4), individuals develop serotype-specific (homotypic) immunity but remain susceptible to infection by the other three serotypes . More critically, secondary infection with a different serotype can lead to more severe disease manifestations through antibody-dependent enhancement (ADE), where pre-existing antibodies from the primary infection bind to the new serotype but fail to neutralize it effectively . This immunological peculiarity creates significant challenges for both vaccine development and antibody-based therapeutics, requiring careful consideration of cross-serotype effects and potential enhancement of disease severity.

What is the molecular basis for antibody-dependent enhancement in dengue infection?

Antibody-dependent enhancement occurs through a precise molecular mechanism involving imperfect antibody binding. After primary infection, individuals develop antibodies specific to that dengue serotype. When exposed to a different serotype, these antibodies recognize but don't perfectly bind to the new virus variant . The antibodies attach to the virus but cannot neutralize it effectively. Instead, they facilitate viral entry into immune cells like macrophages and monocytes through Fc receptor binding. Once inside these cells, the virus escapes neutralization and replicates more efficiently . This mechanism creates a paradoxical situation where the immune response actually enhances viral replication rather than controlling it. Research has identified specific structural determinants on the dengue envelope protein that contribute to this phenomenon, particularly in domain III, which serves as an important target for therapeutic antibody engineering aimed at overcoming ADE .

What distinguishes effective neutralizing antibodies from those that might enhance disease?

Effective neutralizing antibodies typically bind with high affinity to conserved epitopes across multiple dengue serotypes, completely blocking viral attachment or fusion mechanisms. These antibodies target functionally critical regions of the virus that cannot easily mutate without compromising viral fitness . In contrast, enhancing antibodies usually bind with moderate affinity to variable regions or exhibit serotype-specific binding patterns, which allows them to attach to the virus but not effectively neutralize it . Research shows that broadly neutralizing antibodies often target domain III of the envelope protein at epitopes that are structurally constrained across serotypes . The stoichiometry of binding also matters—neutralization typically requires a higher occupancy of binding sites than enhancement. Engineered antibodies like Ab513 demonstrate this principle by achieving high-affinity binding to conserved epitopes across all four serotypes, resulting in neutralization potency with EC50 values below 200 ng/ml for all serotypes .

How can structure-guided design overcome limitations of traditional antibody discovery approaches?

Structure-guided antibody design offers significant advantages over conventional B-cell panning methods for dengue control. Traditional approaches rely on isolating naturally occurring antibodies from infected individuals, which inherently limits discovery to immunodominant epitopes that may not be functionally optimal targets . By contrast, structure-guided design employs computational modeling of epitope-paratope interfaces to engineer antibodies directed at conserved, non-immunodominant epitopes with functional relevance. This approach was successfully demonstrated with Ab513, developed by analyzing atomic interactions between interacting amino acid pairs and introducing strategic mutations to enhance binding affinity . Using network theory computational frameworks, researchers identified key interaction points and engineered six affinity-enhancing point mutations and an affinity-enhancing deletion at position 26 (VH), resulting in up to 40-fold improved binding to diverse DENV strains . This method bypasses the limitations of natural immune responses and accesses epitopes that elicit little or no humoral response but may be crucial for broad neutralization.

What specific epitope targets show the most promise for broad-spectrum dengue neutralization?

Research indicates that domain III (EDIII) of the dengue envelope protein contains epitopes with exceptional promise for broad-spectrum neutralization. EDIII epitopes show greater conservation across serotypes compared to the more variable EDI/II hinge regions . Within EDIII, structural analysis has identified specific binding regions that are functionally constrained across all four serotypes, making them ideal targets for therapeutic antibodies. For example, the epitope targeted by antibody 4E11 and its engineered derivatives demonstrates significant cross-reactivity potential . Through painstaking structural mapping and sequence analysis, researchers have determined that targeting these conserved EDIII epitopes can yield antibodies like Ab513 with nanomolar binding affinities across diverse dengue strains . Importantly, these epitopes are often non-immunodominant in natural infection, explaining why they haven't emerged prominently in conventional antibody discovery approaches. The strategic focus on these regions has yielded therapeutic candidates capable of neutralizing all four serotypes with EC50 values below 200 ng/ml, representing a significant advance in broad-spectrum antibody development .

What methodological approaches can be used to optimize antibody binding to multiple dengue serotypes?

Optimization of antibody binding across dengue serotypes requires sophisticated methodological approaches. Researchers have successfully employed computational frameworks adapted from network theory to map and enhance epitope-paratope interfaces . This process begins with crystal structure analysis of antibody-antigen complexes to identify key interaction residues. Researchers then apply computational algorithms to calculate interaction energies between specific amino acid pairs, creating a detailed map of the binding interface . Strategic mutations can then be introduced at positions predicted to enhance binding energy or remove steric hindrances. This approach was exemplified in the development of Ab513, where researchers introduced seven key modifications (six point mutations plus a deletion) that dramatically improved binding to challenging serotypes like DENV-3 and DENV-4 while maintaining affinity to DENV-1 and DENV-2 . The process involves multiple rounds of directed evolution, including site-directed mutagenesis, expression of variant antibodies, and binding affinity measurements. Advanced techniques like surface plasmon resonance and neutralization assays with diverse viral isolates are essential for validating improvements across serotypes .

What animal models are most appropriate for evaluating therapeutic dengue antibodies?

Evaluating dengue antibody therapeutics requires specialized animal models that recapitulate key aspects of human dengue pathogenesis. Humanized mouse models have emerged as particularly valuable for assessing therapeutic efficacy . These models involve immunodeficient mice (typically NOD/SCID/IL2Rγnull) engrafted with human hematopoietic stem cells, creating chimeric animals with human immune components susceptible to dengue infection. These models can demonstrate critical clinical features including thrombocytopenia and vascular leakage . Additionally, maternal transfer models have been developed to specifically test antibody interventions against antibody-dependent enhancement scenarios, where maternal antibodies can enhance infection severity in offspring. These models allow researchers to assess whether therapeutic antibodies can overcome the enhancement effect . AG129 mice (lacking interferon α/β and γ receptors) represent another important model system, particularly useful for studying viral replication and antibody-mediated protection. When selecting models, researchers must consider the specific aspects of dengue pathogenesis they aim to study, such as viremia control, prevention of vascular leakage, or protection against lethal infection enhancement .

How can researchers effectively measure antibody-mediated protection against dengue clinical manifestations?

Comprehensive assessment of antibody-mediated protection requires multiparameter analysis targeting key clinical manifestations of dengue. Researchers should implement standardized methodologies to measure protection against thrombocytopenia, vascular leakage, viremia, and mortality in appropriate animal models . For thrombocytopenia assessment, serial complete blood counts with platelet enumeration are essential, comparing antibody-treated and control groups over the course of infection. Vascular leakage can be quantified through Evans blue dye extravasation assays, measuring dye concentrations in tissues as an indicator of capillary permeability . Viremia reduction represents a critical efficacy parameter, best measured through quantitative RT-PCR to determine viral RNA copies in serum, supplemented by plaque assays to confirm infectious virus reduction. Survival studies in lethal models provide the ultimate measure of protection, particularly in maternal antibody transfer models that recapitulate antibody-dependent enhancement . Additionally, researchers should monitor cytokine profiles and immune cell activation to understand mechanistic aspects of protection. Ab513 demonstrated efficacy across these parameters, reducing viremia to nearly undetectable levels, mitigating thrombocytopenia, resolving vascular leakage, and protecting mice in a maternal transfer model of lethal antibody-mediated enhancement .

What methodologies should be used to assess potential viral escape from therapeutic antibodies?

Assessing viral escape from therapeutic antibodies requires rigorous methodological approaches combining in vitro and in vivo techniques. Researchers should implement serial passage experiments where virus is cultured in sub-neutralizing antibody concentrations over multiple generations to select for potential escape variants . Each passage should undergo full genome sequencing to identify emerging mutations, with particular attention to the antibody binding epitope. Identified mutations should be introduced into reference strains through reverse genetics to confirm their role in escape. Additionally, researchers should perform neutralization assays against a panel of genetically diverse clinical isolates to assess pre-existing resistance patterns . For in vivo validation, monitoring viral populations during antibody therapy in animal models through deep sequencing is crucial. Understanding structural determinants of escape can be achieved through co-crystallization studies of antibody-antigen complexes. Importantly, in studies with Ab513, researchers observed that this therapeutic antibody targeting a functionally constrained EDIII epitope did not readily lead to resistance in either cell culture systems or animal models, suggesting high genetic barriers to escape . This resistance profile is likely due to targeting functionally critical, conserved epitopes where mutations would compromise viral fitness.

How should dengue serological status influence therapeutic antibody administration strategies?

Dengue serological status critically determines the optimal approach to therapeutic antibody administration. Individuals with no prior dengue exposure (seronegative) present different considerations than those with previous infection history (seropositive) . For seronegative patients, therapeutic antibodies must demonstrate complete neutralization across all serotypes to prevent potential enhancement of future infections. In contrast, seropositive individuals have existing immunity to at least one serotype, which modifies the risk-benefit calculation . Research indicates that serological testing should precede any antibody administration, with point-of-care rapid diagnostic tests offering critical information on infection history . Therapeutic strategies may differ based on results—for seronegative patients, antibodies targeting non-immunodominant epitopes like those in Ab513 may be preferable to avoid interference with natural immunity development . For seropositive individuals experiencing secondary infection, therapeutic antibodies can supplement existing immunity. The timing of administration is equally crucial, with maximum benefit likely achieved during early infection when viral loads peak . This personalized approach based on serological status reflects the complex immunological landscape of dengue and the need for precision in therapeutic interventions.

What factors determine the dosing and timing of therapeutic antibody administration for dengue infections?

Optimal dosing and timing for dengue therapeutic antibodies depend on multiple clinical and virological factors. Viremia kinetics play a crucial role—dengue typically demonstrates a brief but intense viremic period, with peak viral loads occurring early (2-3 days post-symptom onset) before declining rapidly . This narrow window necessitates early intervention, ideally before the critical phase of illness when vascular leakage and other severe manifestations emerge. Dosing considerations include the antibody's pharmacokinetic properties, neutralization potency, and biodistribution to key replication sites. Antibodies like VIS513 and Ab513 with high neutralization potency (EC50 <200 ng/ml) may require lower doses than less potent alternatives . The patient's weight, kidney function, and disease severity also influence dosing calculations. In animal models, single doses of therapeutic antibodies have demonstrated efficacy when administered early in infection, reducing viremia to nearly undetectable levels . The presence of pre-existing enhancing antibodies may necessitate higher therapeutic doses to overcome enhancement effects. Finally, different clinical objectives—whether prevention of progression to severe disease in mild cases or treatment of already severe cases—may require distinct dosing and timing strategies.

How can researchers address the challenges of antibody interference with natural immunity development?

Addressing antibody interference with natural immunity development represents a significant research challenge. Therapeutic antibodies may potentially mask important epitopes, preventing the immune system from developing a comprehensive response to the infection . Researchers can mitigate this through strategic epitope targeting—focusing on non-immunodominant epitopes allows therapeutic neutralization while leaving immunodominant regions available for natural immune processing . Another approach involves engineering antibodies with modified Fc regions that maintain neutralization capacity but minimize interference with antigen presentation and processing. Careful timing of antibody administration is also crucial; administering after the initial immune priming has occurred (typically 2-3 days post-infection) may reduce interference while still providing therapeutic benefit . Combination approaches that pair antibody therapy with immune-stimulating agents could potentially enhance rather than inhibit natural immunity development. Researchers should implement comprehensive immunological monitoring in clinical trials, including measurements of endogenous antibody development across multiple epitopes and T cell responses. Longitudinal follow-up studies are essential to determine whether therapeutic antibody recipients develop protective immunity against subsequent infections . These considerations are particularly important given dengue's unique immunology, where incomplete immunity can potentially enhance future infections.

How can mathematical modeling improve prediction of antibody effects across diverse dengue strains?

Mathematical modeling offers powerful tools for predicting antibody effects across genetically diverse dengue strains. Advanced statistical frameworks have been developed to analyze changes in antibody levels after successive infections with different dengue serotypes, enabling identification of individuals at enhanced risk for severe disease . These models integrate multiple parameters including antibody binding affinities to different serotypes, neutralization potencies, Fc-receptor interactions, and viral genetic diversity. By modeling the epitope-paratope interface as a network of atomic interactions, researchers can predict how specific mutations in either the antibody or virus might affect binding energetics . This approach successfully guided the development of Ab513, where computational analysis identified key interaction residues for optimization. Population-level models incorporating antibody kinetics can predict the impact of therapeutic interventions on transmission dynamics and disease severity distributions . Additionally, structural modeling of antibody-virus complexes helps identify conserved epitopes that minimize escape potential. These mathematical approaches are particularly valuable for dengue given its complex immunology, where the relationship between binding, neutralization, and enhancement is non-linear and context-dependent . Implementation requires integration of diverse datasets including cohort studies, experimental binding measurements, and genetic surveillance of circulating strains.

What novel approaches can overcome antibody-dependent enhancement while maintaining therapeutic efficacy?

Innovative strategies to overcome antibody-dependent enhancement while preserving therapeutic efficacy are at the forefront of dengue research. One promising approach involves structure-guided antibody engineering targeting non-immunodominant epitopes that are functionally critical for the virus . Antibodies like Ab513, engineered to target domain III of the envelope protein with high affinity across all serotypes, demonstrate this concept by achieving broad neutralization without enhancing effects . Another strategy involves Fc engineering to modify antibody effector functions—by introducing specific mutations in the Fc region, researchers can eliminate Fc receptor binding while maintaining neutralization capacity, effectively preventing the cellular entry mechanism that drives enhancement. Bispecific antibodies that simultaneously target multiple epitopes represent another innovative approach, creating high-avidity binding that favors neutralization over enhancement . Some researchers are exploring antibody-antiviral combination therapies, where the antibody provides immediate viral control while the antiviral prevents enhancement of any remaining virus. Novel delivery systems using nanoparticles or sustained-release formulations may achieve more consistent neutralizing concentrations, preventing the sub-neutralizing levels associated with enhancement. These approaches demonstrate how mechanistic understanding of dengue immunopathology can drive innovative therapeutic solutions that maintain efficacy while addressing the unique challenges of antibody-dependent enhancement.

How can high-throughput screening methods be optimized for identifying novel dengue neutralizing antibodies?

Optimizing high-throughput screening for novel dengue neutralizing antibodies requires sophisticated methodological approaches extending beyond traditional techniques. Researchers should implement multi-parameter screening cascades that simultaneously assess binding to all four serotypes using multiplexed bead-based assays with recombinant envelope proteins . These initial screens should be followed by functional neutralization assays using reporter virus particles expressing structural proteins from diverse dengue strains. Critical to effective screening is the incorporation of enhancement assays using Fc receptor-bearing cells to identify and eliminate enhancing candidates early . Advanced structural biology approaches, including epitope binning through competition assays, help classify antibodies by target region and identify those binding to conserved, functionally critical epitopes. Deep sequencing of antibody libraries before and after selection provides valuable information on enrichment patterns and structural features associated with broad neutralization. Computational approaches can further refine screening by predicting cross-reactivity based on epitope conservation analysis . Additionally, single B-cell sorting technologies combined with next-generation sequencing enable rapid identification of naturally occurring neutralizing antibodies from dengue-exposed individuals. These comprehensive screening approaches have already yielded promising candidates like VIS513 and Ab513, demonstrating their value in identifying therapeutic antibodies with the desired profile of broad neutralization without enhancement .

Product Science Overview

Introduction

Dengue fever, caused by the dengue virus (DENV), is a mosquito-borne viral infection that has become a significant public health concern in tropical and subtropical regions. The rapid and accurate diagnosis of dengue infection is crucial for effective disease management and control. One of the diagnostic tools used for this purpose is the lateral flow test, which employs monoclonal antibodies to detect specific antigens or antibodies in patient samples.

Monoclonal Antibodies

Monoclonal antibodies (mAbs) are antibodies that are produced by identical immune cells, cloned from a single parent cell. These antibodies are highly specific to a particular antigen, making them valuable tools in diagnostic assays. In the context of dengue diagnosis, monoclonal mouse anti-dengue antibodies are used to detect dengue virus antigens or antibodies in patient samples.

Lateral Flow Test

The lateral flow test is a simple, rapid, and cost-effective diagnostic tool that can be used at the point of care. It consists of a test strip with a sample pad, a conjugate pad, a nitrocellulose membrane with test and control lines, and an absorbent pad. When a sample is applied to the sample pad, it migrates along the test strip by capillary action. If the target antigen or antibody is present in the sample, it binds to the labeled antibodies on the conjugate pad, forming a complex that is captured by the test line on the nitrocellulose membrane. The appearance of a colored line at the test line indicates a positive result, while the control line confirms the validity of the test.

Monoclonal Mouse Anti Dengue Control

The monoclonal mouse anti-dengue control antibody is used to prepare the control line in the lateral flow test for dengue diagnosis. This control line ensures that the test is functioning correctly and provides a reference for interpreting the test results. The antibody is typically purified using protein A chromatography and formulated in a phosphate-buffered saline solution. It is essential to store the antibody under appropriate conditions to maintain its stability and functionality .

Applications and Benefits

The use of monoclonal mouse anti-dengue control antibodies in lateral flow tests offers several advantages:

  • High specificity and sensitivity: Monoclonal antibodies are highly specific to the dengue virus, reducing the likelihood of cross-reactivity with other pathogens.
  • Rapid results: Lateral flow tests provide results within minutes, enabling timely clinical decision-making.
  • Ease of use: The tests are simple to perform and do not require specialized laboratory equipment, making them suitable for use in resource-limited settings.
  • Cost-effective: Lateral flow tests are relatively inexpensive compared to other diagnostic methods, making them accessible to a broader population.

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.