RSV Paired Antibody

Mouse Anti Human Respiratory Syncytial Virus
Shipped with Ice Packs
In Stock

Description

Definition and Mechanism of RSV Paired Antibodies

RSV paired antibodies involve two complementary mAbs that bind non-overlapping epitopes on viral proteins like the fusion (F) or nucleoprotein (NP). This dual-binding approach improves diagnostic accuracy by reducing cross-reactivity and increases therapeutic efficacy through synergistic neutralization . For example:

  • Diagnostic pairs: Capture and detection antibodies (e.g., Sino Biological’s Cat#: 11049-R338 and 11049-R302) bind RSV-F glycoprotein for antigen detection .

  • Therapeutic pairs: Bispecific antibodies like anti-idiotypic immunogens engage B-cell receptors (BCRs) encoding RSV-neutralizing antibodies .

Diagnostic Applications

Paired antibodies enable high-sensitivity RSV detection in lateral flow assays and ELISAs. Recent studies highlight:

Assay PlatformSensitivitySpecificityTarget ProteinSource
Quidel Sofia™78.6%93.9%F glycoprotein
BD Veritor™81.6%99.1%F glycoprotein
Rabbit phage-derived>95%N/ANucleoprotein

Sino Biological’s RSV-F antibody pairs achieved 100-fold enrichment in antigen detection, while novel rabbit-derived anti-NP pairs demonstrated >95% insertion rates in phage display screens .

Therapeutic Applications

Nirsevimab (Beyfortus®), a long-acting monoclonal antibody targeting RSV-F, reduced lower respiratory tract infections (LRTI) by 70–90% in adults and infants . Comparative data with palivizumab (Synagis®) show:

ParameterNirsevimab (Single Dose)Palivizumab (Monthly Doses)Source
Neutralizing antibody half-life~71 days~20 days
GMFR (Day 151)116 (103–130)11 (9–13)
Hospitalization reduction83%55%

Nirsevimab’s sustained neutralizing activity (17-fold above baseline at day 361) outperforms palivizumab, which requires monthly dosing .

Epitope-Specific Neutralization

Studies identify critical epitopes for paired antibody design:

  • Pre-fusion F (pre-F): Antibodies like D25 bind site Ø, achieving 2–3× higher neutralization than post-F antibodies .

  • Nucleoprotein (NP): Rabbit-derived mAbs (e.g., clones R338/R302) detect conserved RSV-A and RSV-B strains with 53 viable pairs identified .

  • G protein: Non-neutralizing mAbs targeting the central conserved domain (CCD) reduce viral dissemination in murine models .

Bispecific Antibody Engineering

Anti-idiotypic bispecific antibodies (ai-mAbs) activate BCRs encoding VH3-21/VL1-40 antibodies, which neutralize RSV without affinity maturation. These ai-mAbs selectively engage B cells expressing RSV-specific receptors, offering a vaccine-like immune boost .

Challenges and Future Directions

  • Durability: Nirsevimab’s protection wanes after 5 months in preterm infants, necessitating booster strategies .

  • Antigenic diversity: RSV-A and RSV-B subtypes require cross-reactive pairs, as seen in nucleoprotein-targeting mAbs .

  • Standardization: Multi-laboratory studies show variability in neutralization assays, urging harmonized protocols for clinical trials .

Product Specs

Description
This product contains both an RSV gold conjugation antibody and an RSV capture antibody, designed for use in the development of rapid tests for RSV. Please note: When ordering a quantity, such as 100µg, you will receive 50µg of each antibody (totaling 100µg).
Purity
Greater than 95%
Physical Appearance
This product consists of two vials containing a sterile, filtered, clear, and colorless solution.
Formulation
* RSV gold conjugation antibody in PBS, pH 7.4. * RSV capture antibody in PBS, pH 7.4.
Stability
For optimal storage, RSV Paired Antibody should be kept below -18°C. While it can remain stable at 4°C for up to one week, it is important to avoid repeated freeze-thaw cycles.
Applications
Lateral flow rapid tests.
Purification Method

Purified monoclonal IgG2a by protein A chromatography.

Type
Mouse antibody Monoclonal.

Q&A

What are the primary antigenic targets for RSV neutralizing antibodies?

The RSV fusion (F) glycoprotein serves as the primary target for neutralizing antibodies, with at least six major antigenic sites (Ø, I, II, III, IV, V) identified in the literature . PreF-specific antibodies targeting the apex sites, particularly site Ø, are immunodominant and account for a large proportion of neutralizing activity in human sera . Recent studies indicate that site V is targeted by nearly half of the most potent neutralizing antibodies isolated from healthy adults . Understanding these targets is essential for developing effective vaccines and therapeutic antibodies against RSV.

Why is paired antibody analysis critical for RSV research compared to single-chain antibody studies?

Paired antibody data (containing both heavy and light chain information) provides a more comprehensive understanding of RSV binding mechanisms compared to single-chain analysis . While sequence identity searches using only the heavy chain variable domain (VH) identify antibodies binding to the same epitope approximately 25% of the time, including the light chain variable domain (VL) improves accuracy to around 75% . For RSV antibodies like those binding to the F protein, the paratope is often evenly distributed between heavy and light chains, with similar numbers of residues from each chain interacting with the antigen . This balanced contribution makes paired analysis essential for accurate epitope prediction and binding characterization.

How do antibodies isolated from mucosal tissues compare to those from peripheral blood in RSV research?

Analysis of 806 RSV F-specific antibodies isolated from paired adenoid and peripheral blood samples from young children revealed that adenoid-derived antibodies demonstrate higher binding affinities and neutralization potencies . Approximately 25% of neutralizing antibodies isolated from adenoids originate from a unique population of IgM+ and/or IgD+ memory B cells containing high somatic mutation loads but lacking classical memory B cell markers . This significant difference highlights the importance of studying mucosal immunity in RSV infection and vaccine development, as respiratory mucosal tissues may harbor distinct antibody populations with superior neutralizing capabilities.

What techniques effectively isolate RSV-specific antibodies from human samples?

Multiple complementary techniques are employed for isolating RSV-specific antibodies:

  • Phage display technology: Effective for developing antibodies against RSV nucleoprotein (NP), as demonstrated in studies where rabbits were immunized with recombinant NP and Fab libraries were constructed and sorted through successive panning rounds .

  • B cell sorting methods: Memory B cells from donors can be isolated via flow cytometry using RSV F antigen-specific single memory B cell sorting or memory B cell enrichment approaches .

  • Paired tissue sampling: Simultaneous collection from peripheral blood and mucosal tissues (e.g., adenoids) enables comparative analysis of tissue-specific antibody repertoires .

The selection of an appropriate isolation technique depends on research objectives, with each method offering unique advantages for specific applications in RSV antibody research.

What experimental designs best measure neutralizing activity of RSV antibodies?

Neutralizing activity evaluation requires systematic approaches:

  • Sandwich ELISA protocols: Typically involving coating antibodies (1 μg/w in PBS) overnight at 4°C, followed by blocking with 1% BSA-PBS and 10% sucrose, antigen incubation (dilution series ranging from 5 to 0.2 ng/w), and multiple wash steps .

  • Neutralization assays against multiple virus strains: Testing antibodies against both RSV-A and RSV-B strains to determine subtype-specific and cross-neutralizing activity .

  • Paired sample comparison: Evaluating neutralizing antibody titers (NATs) in matched samples, such as maternal blood and cord blood, to assess protective antibody transfer .

For comprehensive characterization, researchers should employ multiple complementary assays rather than relying on a single neutralization measure.

How does somatic hypermutation affect the binding and neutralization properties of RSV antibodies?

Somatic hypermutation (SHM) critically influences RSV antibody functionality. Analysis of 23 monoclonal antibodies with high RSV neutralization potency revealed average SHM rates of 11-52 nucleotides (median 25) in VH genes . This substantial mutation level correlates with improved binding affinity and neutralization capabilities. Interestingly, even unconventional memory B cell populations (IgM+ and/or IgD+) from adenoids show high SHM loads despite lacking classical memory markers . This finding suggests that extensive antigen-driven affinity maturation occurs in response to RSV exposure across multiple B cell lineages, contributing to diverse antibody repertoires with varied neutralization potentials.

How do structural variations in CDR regions influence epitope specificity and cross-reactivity?

Complementarity-determining regions (CDRs), particularly CDR-H3, exhibit considerable diversity in RSV antibodies, with lengths ranging from 8-21 amino acids (median 13) . This structural diversity significantly impacts epitope recognition and cross-reactivity patterns. For some RSV binding antibodies, specific CDR loop conformations are essential for recognizing particular epitopes on the RSV Fusion Glycoprotein . The three-dimensional configuration of CDR loops, rather than sequence identity alone, often determines binding specificity. This structural basis for binding also explains why antibodies targeting antigenic site IV may show cross-neutralizing activity against both RSV and human metapneumovirus (hMPV), while those targeting sites Ø, I, II, and V typically demonstrate mono-specific neutralization without cross-reactivity .

What factors influence maternal-infant RSV antibody transfer and how can these be measured?

Maternal-infant antibody transfer dynamics represent a critical research area with implications for passive protection and vaccination timing. Studies measuring neutralizing antibody titers (NATs) in 95 mother-child pairs revealed that cord blood (CB) contains slightly higher levels of RSV-specific antibodies than maternal blood (MB) . Strong positive correlations exist between maternal and cord blood for both NATs against RSV-A (r = 0.75) and epitope-specific antibody levels against sites Ø (r = 0.76) and IIa (r = 0.69) .

These correlations reflect efficient transplacental transfer of RSV-specific antibodies. Researchers should employ multiple measurement approaches to fully characterize this transfer:

  • Total neutralizing activity comparisons between maternal and cord blood

  • Epitope-specific antibody quantification targeting critical sites like Ø and IIa

  • Subtype-specific (RSV-A vs. RSV-B) antibody transfer assessment

How do prefusion (PreF) and postfusion (PostF) F protein conformations affect antibody binding and vaccine design?

The RSV F protein exists in two conformational states with significant implications for antibody recognition and vaccine development. The prefusion (PreF) conformation contains unique epitopes, particularly site Ø, that are absent in the postfusion (PostF) form . Studies suggest PreF elicits superior neutralizing antibody responses compared to PostF, making it a preferred vaccine antigen target .

The metastability of wild-type prefusion RSV F has prompted structure-based rational design approaches to engineer stabilized PreF conformations for subunit vaccine development . This strategy has yielded promising candidates like DS-Cav1, designed based on atomic-level understanding of RSV protein structure . In clinical trials, DS-Cav1 demonstrated ability to induce substantial increases in RSV-neutralizing antibodies that persisted for several months following a single dose . This structure-based vaccine design represents a significant advancement toward addressing the long-standing challenge of RSV prevention.

What strategies can overcome challenges in developing broadly neutralizing antibodies against diverse RSV strains?

Developing broadly neutralizing antibodies faces several obstacles due to RSV's genetic diversity. RSV has two antigenic subtypes (A and B) with approximately 50% variation in the G protein sequence between subtypes . Current research indicates antigenic site IV of the F protein as a promising target for cross-neutralizing antibodies active against both RSV and hMPV, while antibodies against sites Ø, I, II, and V typically show strain-specific neutralization .

Experimental approaches to address these challenges include:

  • Structural mapping of conserved epitopes across RSV strains and related viruses

  • Deep mutational scanning to identify resistant viral variants and guide antibody optimization

  • Rational engineering of antibodies targeting multiple epitopes simultaneously

  • Isolation of naturally occurring broadly neutralizing antibodies from individuals with robust responses to multiple RSV strains

Understanding diverse binding modes and epitope recognition patterns remains crucial for developing broadly protective antibody-based interventions .

How can researchers optimize paired B-cell isolation protocols for low-frequency RSV-specific B cells?

The isolation of RSV-specific B cells, particularly paired heavy and light chains from rare populations, presents significant technical challenges. Based on current methodologies in the field, researchers should consider:

  • Antigen-specific enrichment steps prior to single-cell sorting to increase rare cell recovery

  • Optimizing flow cytometry parameters including using multiple fluorescently-labeled RSV antigens (PreF and PostF) simultaneously

  • Implementing index sorting to correlate cellular phenotypes with paired sequence data

  • Employing single-cell RNA sequencing with computational pairing algorithms for high-throughput analysis

For memory B cell isolation, researchers have successfully used flow cytometry with RSV F antigens for single memory B cell sorting and memory B cell enrichment approaches . These methods have yielded antibodies with varying degrees of RSV neutralization potency, binding specificity, and epitope targeting.

What analytical frameworks best correlate in vitro neutralization assays with in vivo protection?

Establishing correlations between laboratory neutralization measurements and clinical protection remains challenging. A comprehensive analytical framework should incorporate:

  • Multiple neutralization assay formats using different cell types and readouts

  • Fc-mediated effector function analysis beyond direct neutralization

  • Epitope-specific antibody quantification targeting functionally critical sites

  • Integration of antibody affinity, avidity, and neutralization potency metrics

  • Comparative analysis between serum neutralization and mucosal antibody measurements

Studies measuring both neutralizing antibody titers (NATs) and epitope-specific antibody levels against sites Ø and IIa have begun to establish these correlations , but more comprehensive frameworks integrating multiple parameters are needed to accurately predict in vivo protection from in vitro measurements.

How are computational approaches enhancing RSV antibody discovery and optimization?

Advanced computational tools are revolutionizing RSV antibody research. The Patent and Literature Antibody Database (PLAbDab) provides extensive paired antibody sequences from patents and literature sources , enabling sequence-based and structure-based searches for similar antibodies. For RSV antibodies, structure prediction tools like ABodyBuilder2 have demonstrated ability to accurately model antibody-antigen interactions .

These computational approaches support:

  • Identification of antibodies with similar binding properties but diverse sequences

  • Structural analysis of CDR conformations critical for RSV epitope recognition

  • Prediction of cross-reactivity based on paratope structural features

  • Virtual screening of antibody libraries prior to experimental validation

For example, structural analysis revealed that an RSV binding antibody required specific CDR loop conformations to bind the RSV Fusion Glycoprotein at a particular site, with 23 structurally similar antibodies targeting the same epitope despite sequence diversity .

What novel immunization strategies might enhance the development of mucosal RSV-specific antibodies?

Given the superior binding affinity and neutralization potency of adenoid-derived RSV antibodies compared to those from peripheral blood , novel immunization strategies targeting mucosal immunity represent a promising direction. Research should explore:

  • Mucosal delivery systems for RSV vaccine antigens to stimulate local immune responses

  • Prime-boost strategies combining systemic and mucosal immunization routes

  • Adjuvant formulations specifically designed to enhance mucosal antibody development

  • Vaccination approaches targeting the unique IgM+/IgD+ memory B cell population found in adenoids that produces potent neutralizing antibodies despite lacking classical memory markers

These strategies could potentially generate more robust and effective antibody responses at respiratory mucosal surfaces, the primary site of RSV infection.

How might maternal RSV vaccination impact infant antibody development and long-term immunity?

Maternal vaccination represents a promising strategy for protecting infants against RSV, with pregnant women now being offered RSV vaccines in several countries . Research questions regarding this approach include:

  • The duration of protection conferred by maternally-transferred antibodies

  • Potential interference of maternal antibodies with infant immune responses to natural RSV exposure

  • Impact on the development of infant B cell repertoires and memory formation

  • Optimal timing of maternal vaccination to maximize antibody transfer

Studies showing positive correlations between maternal and cord blood antibody levels (r = 0.75 for neutralizing antibodies, r = 0.76 and r = 0.69 for epitope-specific antibodies against sites Ø and IIa) provide encouraging evidence for efficient transfer, but long-term impacts on infant immunity require further investigation.

Table 1: Characteristics of RSV F Protein Antigenic Sites and Corresponding Antibodies

Antigenic SiteLocation on F ProteinConformation SpecificityCross-Reactivity PotentialNeutralization Potency
Site ØApex of PreFPreF-specificMono-specificHigh
Site IPreF and PostFSharedMono-specificModerate
Site IIPreF and PostFSharedMono-specificModerate to High
Site IIIPreF and PostFSharedLimitedVariable
Site IVPreF and PostFSharedCross-neutralizing (RSV/hMPV)Moderate
Site VPreF-specificPreF-specificMono-specificHigh

Table compiled based on data from sources and

Table 2: Comparison of Antibody Properties from Different Anatomical Sites

PropertyAdenoid-Derived AntibodiesPeripheral Blood Antibodies
Binding AffinityHigherLower
Neutralization PotencyHigherLower
Somatic HypermutationHigh (even in IgM+/IgD+)Variable
Unique B Cell Populations25% from IgM+/IgD+ memory B cellsConventional memory B cells
Classical Memory MarkersSome populations lackingTypically present

Table compiled based on data from source

Table 3: Maternal-Infant Antibody Transfer Correlations

Antibody MeasureCorrelation Coefficient (r)
NATs for RSV subtype A0.75
ESA levels against site Ø0.76
ESA levels against site IIa0.69

Table compiled based on data from source

Product Science Overview

Introduction

Human Respiratory Syncytial Virus (hRSV) is a negative-sense single-stranded RNA virus belonging to the genus Pneumovirus. It is a major cause of respiratory infections, particularly in infants, young children, and the elderly. The virus can lead to severe bronchiolitis and pneumonia, making it a significant public health concern.

Mouse Models in hRSV Research

Mouse models have been extensively used in hRSV research due to their genetic similarities to humans and the availability of various immunological tools. These models help in understanding the pathogenesis of hRSV and evaluating potential therapeutics.

  1. Rag2 Knockout Mice: One notable model involves Rag2 gene knockout mice, which are deficient in T and B cells. These mice can sustain high viral loads upon intranasal inoculation with hRSV, making them useful for studying long-term infection and pathogenesis . The use of CRISPR/Cas9 technology has enabled the creation of these models, which exhibit severe bronchopneumonia and elevated cytokine levels.

  2. Immunological Studies: The laboratory mouse is the species of choice for most immunological studies, ranging from simple vaccine testing to intricate dissection of immunopathogenic mechanisms . Although not fully adapted to mice, some strains of hRSV can replicate in the murine respiratory tract and induce specific T and B cell responses. Passive transfer of neutralizing antibodies has shown to be protective and assists in viral clearance.

Development of Monoclonal Antibodies

Monoclonal antibodies targeting hRSV have shown promise in both prophylactic and therapeutic settings. These antibodies can neutralize the virus and prevent its spread within the host. In mouse models, humanized monoclonal antibodies have demonstrated strong antiviral activity, suggesting their potential for clinical use .

Challenges and Opportunities

Despite the advancements, there are challenges in using mouse models for hRSV research. The semi-permissiveness and short duration of infection in some models limit their application. However, the development of more sophisticated models, such as the Rag2 knockout mice, offers new opportunities for studying hRSV and evaluating novel therapeutics .

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.