Reovirus Antibody

Respiratory Enteric Orphan Virus, Mouse antibody
Shipped with Ice Packs
In Stock

Description

Structure and Functional Targets of Reovirus Antibodies

Reovirus antibodies primarily target structural proteins of the viral capsid, including:

ProteinRole in ReovirusAntibody Function
σ1 (Sigma 1)Host cell attachment (via JAM-A)Neutralizes viral entry
σ3 (Sigma 3)Outer capsid stabilityBlocks proteolytic uncoating
λ2 (Lambda 2)Core turret proteinInhibits transcription

Monoclonal antibodies (e.g., MAb 9BG5, 4F2) bind σ1 to prevent JAM-A-mediated viral entry into host cells , while polyclonal responses target multiple epitopes for broad neutralization .

Detection Methods and Seroprevalence

Reovirus antibodies are detected via enzyme-linked immunosorbent assays (ELISA), immunofluorescence, and hemagglutination inhibition . Seroprevalence studies reveal:

Age-Dependent Seropositivity in Children ( )

Age GroupSeroprevalence (%)Geometric Mean Titer (GMT)
0–3 months75.01:453
3–6 months11.1
6–12 months0
1–2 years8.21:508
5–6 years50.01:999

Maternal antibodies wane by 6–12 months, followed by natural infection-driven seroconversion .

Clinical Implications in Oncolytic Therapy

Reovirus antibodies influence the efficacy of reovirus-based cancer treatments:

Neutralizing Antireovirus Antibodies (NARA)

  • Impact on Therapy: High pre-existing NARA titers reduce viral replication and oncolysis in systemic therapies .

  • Dose Response: Intravenous reovirus administration triggers rapid NARA elevation (peak at 8–15 days post-infusion), limiting repeated dosing .

Antibody-Mediated Enhancement

  • Fc Receptor Uptake: In JAM-A-negative cancers, σ1-bound antibodies facilitate Fc receptor-mediated viral entry into tumor cells, enabling apoptosis .

  • Combination Therapies: Antibody-reovirus complexes enhance immune recruitment (e.g., NK cells, CD8+ T cells) in checkpoint blockade-resistant tumors .

Challenges and Research Frontiers

  1. Pre-existing Immunity: Over 50% of adults exhibit antireovirus antibodies, necessitating strategies like oral/intratumoral delivery to evade neutralization .

  2. Therapeutic Optimization:

    • Monoclonal Antibody Cocktails: Synergize with reovirus to target Ras-activated tumors .

    • Gut Microbiome Modulation: Oral reovirus (e.g., RC402) reshapes microbiota to enhance antitumor immunity independently of direct oncolysis .

Key Research Findings

  • Cancer Selectivity: Antibodies against σ1 block reovirus binding to JAM-A, but Ras-activated cells bypass this via PKR inhibition, enabling viral replication .

  • Immune Memory: Reovirus-treated mice show durable antitumor immunity despite neutralizing antibodies .

  • Safety Profile: Grade I/II toxicities (fever, fatigue) dominate; no dose-limiting toxicity observed in trials .

Product Specs

Introduction
Reovirus, a member of the Reoviridae family, is a non-enveloped RNA virus with a spheroidal shape. These viruses, approximately 70 nanometers in diameter, possess two icosahedral capsids enclosing a core of segmented, double-stranded RNA. Reoviruses are generally non-pathogenic and commonly infect humans, with nearly 100% of the population showing evidence of prior exposure. Infections typically manifest in the gastrointestinal or respiratory systems. Notably, reoviruses exhibit a predilection for cells with elevated Ras signaling activity, making them a potential tool for cancer therapy.
Formulation
Supplied as a 1 mg/ml solution in phosphate-buffered saline (PBS) upon reconstitution.
Shipping Conditions
The antibody is shipped in a lyophilized state at ambient temperature.
Storage Procedures
For long-term storage, maintain the lyophilized antibody at 4°C in a dry environment. Following reconstitution, aliquot and store at -20°C if not intended for use within one month.
Solubility
To reconstitute, add deionized water (H2O) to the vial. Mix gently by swirling, ensuring the solution reaches the sides of the vial. Allow 30-60 seconds for complete reconstitution before use.
Titer
For staining applications, use 10 microliters of antibody per 1,000,000 cells. For other applications, the optimal titer should be determined empirically.
Note
This antibody exhibits virus-neutralizing properties by preventing virus binding.
Purification Method
Ion exchange column.
Type
Mouse antibody Monoclonal.
Clone
YRreoV.
Immunogen
Peptide mix corresponding to T1L sigma 1 protein.
Ig Subclass
Mouse IgG2a.

Q&A

What are reovirus antibodies and how are they detected in experimental settings?

Reovirus antibodies are immunoglobulins produced by the host immune system in response to reovirus infection. In experimental settings, these antibodies are typically detected through various serological assays. The most common detection methods include:

Enzyme-linked immunosorbent assay (ELISA) is frequently used to measure binding antibodies, while neutralization assays assess functional antibody activity. For neutralization assays, serum samples are typically heat-inactivated (56°C for 30 minutes) before being subjected to dilution series (commonly starting at 1:5 for human samples and 1:25 or higher for murine samples). These diluted samples are mixed with standardized quantities of reovirus particles (e.g., 150 pfu/well) to allow antibody binding before measuring neutralizing capacity .

Different antibody isotypes may be relevant depending on the research question. For instance, studies on oral reovirus administration have shown increased IgA+ antibody-secreting cells in the lamina propria, highlighting the importance of mucosal immunity in certain contexts .

How do reovirus-specific antibody levels compare between healthy individuals and those with autoimmune conditions?

Research has revealed interesting differences in reovirus antibody profiles between healthy individuals and those with certain autoimmune conditions, particularly celiac disease. Studies have demonstrated that celiac disease patients tend to have higher anti-reovirus antibody titers compared to healthy controls, although these differences don't always reach statistical significance .

This relationship appears to be specific to reovirus rather than reflecting a general heightened antibody response. Celiac disease patients with high anti-reovirus antibody titers did not necessarily exhibit elevated antibodies against other viruses such as rotavirus or herpes simplex virus type 1 (HSV-1) . This specificity suggests a potential mechanistic link between reovirus exposure and celiac disease pathogenesis rather than a generalized immune hyperreactivity.

What is the relationship between reovirus antibody titers and IRF1 gene expression?

Interferon regulatory factor 1 (IRF1) is a transcriptional regulator that plays a key role in immune responses and has been implicated in the loss of oral tolerance to gluten. Research has demonstrated that celiac disease patients with high levels of reovirus antibodies also tend to have higher levels of IRF1 gene expression , suggesting a potential mechanistic link between reovirus infection and celiac disease development.

In experimental models, reovirus-induced loss of oral tolerance occurs in an IRF1-dependent manner . Mouse studies have shown that reovirus infection can significantly inhibit the conversion of antigen-specific CD4+ T cells into regulatory T cells and instead promote differentiation of inflammatory T-helper 1 (Th1) cells in an IRF1-dependent process .

Interestingly, the direct correlation between antibody titers and IRF1 expression is complex. Studies in humans have shown that while both anti-reovirus antibody titers and IRF1 expression are elevated in celiac patients compared to controls, there isn't always a direct correlation between these two measurements within individual patients . This suggests that while reovirus infection may trigger pathways leading to increased IRF1 expression and subsequent loss of oral tolerance, the persistence of high antibody titers may not be directly causative but rather a marker of previous exposure to the virus that initiated the pathogenic process.

How long do reovirus antibodies persist after infection?

The persistence of reovirus antibodies after infection represents an important aspect of immunity with implications for both disease pathogenesis and therapeutic applications. Evidence suggests that these antibodies can persist for extended periods and potentially leave a "permanent mark" on the immune system .

Studies investigating the relationship between reovirus and celiac disease have used antibody status as an indicator of previous reovirus infection , highlighting the long-term persistence of these antibodies. The durability of the antibody response may depend on several factors:

  • The strain of reovirus (different strains can induce varying levels of immune response)

  • The route of exposure (oral vs. systemic)

  • The host's genetic background and immune status

  • The age at which infection occurs (infections during critical developmental windows may have longer-lasting immune signatures)

In the context of celiac disease research, the finding that patients had higher anti-reovirus antibody titers suggests that these antibodies can persist long enough to be detected well after the initial infection event that might have triggered disease development . This persistence has implications for understanding the long-term consequences of reovirus infections, particularly in relation to autoimmune conditions.

What methods are most effective for quantifying neutralizing antibodies against reovirus in serum samples?

Neutralizing antibody (NAb) assays are considered the gold standard for quantifying functionally relevant antibodies against reovirus. Several methodological approaches exist, each with specific advantages and limitations:

NAb Quantification MethodPrincipleAdvantagesLimitationsTypical Readout
Plaque Reduction Neutralization TestReduction in viral plaque formationGold standard, direct measure of infectivityLabor-intensive, low throughputPRNT50 titer
Fluorescent Focus ReductionReduction in cells expressing viral antigensFaster than PRNT, quantifiable by flow cytometryRequires fluorescent antibodies or virusFF50 titer
Reporter Virus AssayReduction in reporter gene expressionHigh throughput, objective quantificationRequires genetically modified virusIC50 value
MicroneutralizationReduction in cytopathic effect in microwellsRequires less sample volumeLess sensitive than PRNTNT50 titer

For optimal results, serum samples should be heat-inactivated (56°C for 30 minutes) to eliminate complement activity that might interfere with the assay . Using standardized positive controls, such as pooled immunoglobulins from multiple donors (e.g., Nanogam), is essential for inter-assay consistency .

When working with murine samples, plasma is often used with higher initial dilutions (1:25, 1:50, or 1:100) compared to human samples (typically starting at 1:5) . These methodological details are critical for accurate quantification of neutralizing antibodies.

How can researchers distinguish between different strains of reovirus when analyzing antibody responses?

Distinguishing antibody responses to different reovirus strains is challenging but essential, as research shows that genetic differences between virus strains can dramatically alter their immunological effects. For example, studies have demonstrated that different reovirus strains can have distinct impacts on oral tolerance to gluten, despite both inducing protective immunity .

Several approaches can help researchers differentiate strain-specific antibody responses:

  • Strain-specific ELISA: Using purified proteins unique to each strain as coating antigens can help identify strain-specific antibodies. The sigma1 protein, which forms the viral attachment protein, shows substantial sequence variation between strains and is an excellent target for strain discrimination.

  • Cross-absorption studies: Serum can be pre-absorbed with one reovirus strain before testing reactivity against another strain to identify antibodies that bind uniquely to each strain.

  • Epitope mapping: Using peptide arrays or phage display libraries to identify the specific epitopes recognized by antibodies can reveal strain-specific recognition patterns.

  • Neutralization assays with strain specificity: Comparing the neutralizing capacity of serum against different reovirus strains can reveal strain-specific functional antibody responses. Research has shown that genetically different reovirus strains can induce distinct immune responses despite both generating protective immunity .

Understanding strain-specific antibody responses is particularly important when investigating reovirus's role in autoimmune conditions, as different strains may have variable capacities to trigger pathogenic immune responses .

What are the challenges in correlating reovirus antibody levels with clinical outcomes in autoimmune diseases?

Correlating reovirus antibody levels with clinical outcomes in autoimmune diseases faces several significant challenges that researchers must address through careful study design:

  • Temporal disconnect: The initial viral trigger may precede disease symptoms by months or years, making it difficult to establish causality. For example, in celiac disease, reovirus infection early in life might trigger immune processes that only manifest as clinical disease later, when other factors align .

  • Multifactorial disease etiology: Autoimmune diseases typically require multiple factors, including genetic predisposition and environmental triggers. Studies in transgenic mice expressing the celiac disease-predisposing HLA molecule DQ8 show that reovirus infection can trigger Th1 immune responses to dietary antigens , but similar complexity exists in human disease.

  • Heterogeneity in antibody responses: The search for correlations is complicated by variations in antibody isotypes, subclasses, affinity, and functionality. Research has shown that celiac disease patients have higher anti-reovirus antibody titers, but without direct correlation to IRF1 expression levels that are also elevated in these patients .

  • Strain-specific effects: Different reovirus strains can induce distinct immunological outcomes , meaning that general "anti-reovirus" antibody measurements may not capture the relevant pathogenic exposure.

  • Confounding by disease state: The autoimmune disease itself might alter antibody production or persistence, making it difficult to determine whether elevated antibody levels are cause or consequence.

These challenges require sophisticated study designs, including prospective cohorts starting in early childhood, strain-specific antibody assessments, and integration of genetic, immunological, and clinical data to establish meaningful correlations.

How do different immunoassays for reovirus antibodies compare in sensitivity and specificity?

Various immunoassays for detecting reovirus antibodies offer different advantages in terms of sensitivity, specificity, and application:

Immunoassay MethodSensitivitySpecificityThroughputTechnical ComplexityFunctional Relevance
ELISAHighModerateHighLowModerate
Neutralization AssayModerateHighLowHighHigh
Western BlotModerateHighLowHighLow
ImmunofluorescenceModerateModerateLowModerateLow
Multiplex AssayModerateModerateHighModerateModerate

Enzyme-linked immunosorbent assay (ELISA) offers high throughput and relative simplicity but may detect both relevant and non-relevant binding antibodies. Neutralization assays provide the highest functional relevance as they measure antibodies that can prevent viral infection and are considered the gold standard for specificity, though they are more labor-intensive and have lower throughput than ELISA.

Western blotting allows identification of antibodies against specific viral proteins, making it useful for differentiating responses to different viral components, but has lower throughput and can have sensitivity issues for conformational epitopes. Immunofluorescence assays are useful for determining subcellular localization of antibody binding but offer relatively low throughput and subjective quantification.

When selecting an assay, researchers must consider the specific research question. Neutralization assays are essential when studying the impact of antibodies on oncolytic efficacy , while ELISAs might be sufficient for epidemiological studies of exposure.

How might pre-existing reovirus antibodies affect the efficacy of reovirus-based oncolytic therapy?

Pre-existing reovirus antibodies, particularly neutralizing antibodies (NAbs), can significantly impact the efficacy of reovirus-based oncolytic therapy through several mechanisms:

  • Impaired direct oncolysis: Research has demonstrated that reovirus-specific NAbs can hamper the oncolytic function of reovirus . These antibodies can neutralize viral particles before they reach tumor sites, reducing direct tumor cell infection and lysis.

  • Differential effects on immune stimulation: Interestingly, while NAbs impair direct oncolytic effects, they do not necessarily inhibit the T cell-attracting capacity of reovirus . This suggests that even neutralized virus may retain some immunotherapeutic properties.

  • Route of administration considerations: The impact of pre-existing antibodies varies depending on administration route. Intravenous delivery may be more susceptible to neutralization by circulating antibodies compared to intratumoral injection. Oral administration, which has shown efficacy in preclinical models , may interact differently with mucosal antibodies.

  • Potential for enhanced delivery: Some research suggests that antibody-bound virus may be more efficiently delivered to tumors through antibody-dependent cellular cytotoxicity or complement-dependent mechanisms, potentially offering an advantage in certain contexts.

These complexities highlight the importance of assessing pre-existing immunity in patients receiving reovirus therapy and potentially developing strategies to mitigate NAb effects, such as shielding viral particles, modifying dosing schedules, or combining with immunomodulatory agents.

What is the evidence linking reovirus antibodies to the pathogenesis of celiac disease?

The evidence linking reovirus antibodies to celiac disease pathogenesis comes from both animal models and human studies, suggesting a potential role for reovirus infection in triggering this autoimmune condition:

  • Elevated antibody titers in patients: Studies have found that celiac disease patients tend to have higher anti-reovirus antibody titers compared to healthy controls, suggesting greater exposure or response to the virus . Importantly, this elevation appears specific to reovirus rather than reflecting general hyper-reactivity to viruses .

  • Mechanistic studies in mouse models: Experimental evidence shows that reovirus infection can disrupt intestinal immune homeostasis and cause loss of oral tolerance to food antigens . In mice, reovirus promoted differentiation of food antigen-specific CD4+ T cells toward inflammatory Th1 responses rather than regulatory T cells , mirroring key immunological features of celiac disease.

  • HLA-associated effects: In transgenic mice expressing the celiac disease-predisposing HLA molecule DQ8, reovirus infection upregulated Th1 cytokine production in mucosal dendritic cells and induced transglutaminase 2 activation , both relevant to celiac pathogenesis.

  • IRF1 expression correlation: Celiac patients with high anti-reovirus antibody titers also had higher expression levels of IRF1 , a transcriptional regulator implicated in loss of oral tolerance to gluten.

  • Timing of infection hypothesis: The research suggests that reovirus infection during a critical window in early life, coinciding with first gluten exposure and immune system maturation, may be particularly important for triggering celiac disease in genetically susceptible individuals .

While these findings suggest a link between reovirus and celiac disease, a direct causative relationship has not been definitively established in humans . The relationship is likely complex and multifactorial, with reovirus potentially acting as one of several environmental triggers in genetically predisposed individuals.

Can reovirus antibody profiles predict susceptibility to autoimmune conditions?

The potential of reovirus antibody profiles to predict susceptibility to autoimmune conditions, particularly celiac disease, is an emerging area of research with both promising findings and significant limitations:

  • Association with celiac disease: Research has identified that celiac disease patients tend to have higher anti-reovirus antibody titers , suggesting that antibody profiles might serve as biomarkers for disease risk or progression.

  • Strain-specific considerations: Different reovirus strains can induce distinct immunological effects . One strain triggered inflammatory immune responses and loss of oral tolerance to gluten in mouse models, while a closely related but genetically different strain did not . This suggests that strain-specific antibody profiling may be more informative than general anti-reovirus antibody measurement.

  • Temporal challenges: The timing of reovirus infection relative to first gluten exposure appears critical . Antibody profiles measured in adulthood may not reflect the relevant childhood exposure that coincided with first gluten introduction, limiting predictive value.

  • Integration with genetic risk factors: Predictive models would likely need to integrate antibody profiles with known genetic risk factors (e.g., HLA-DQ2/DQ8) to achieve meaningful predictive power. Studies in transgenic mice expressing celiac disease-predisposing HLA molecules show that genetic background influences how reovirus infection affects tolerance to dietary antigens .

Currently, reovirus antibody profiles alone cannot reliably predict autoimmune disease susceptibility, but they may contribute valuable information when combined with genetic, environmental, and other immunological markers in comprehensive risk assessment models.

How do reovirus antibodies interact with the gut microbiome in experimental models?

The interaction between reovirus antibodies and the gut microbiome represents a complex and bidirectional relationship that can influence both antiviral immunity and broader immune homeostasis:

  • Microbiome reshaping after reovirus exposure: Research has demonstrated that oral reovirus administration can reshape the gut microbiome . This viral-induced alteration may contribute to the immunomodulatory effects observed with reovirus therapy.

  • IgA production and microbial targeting: Oral reovirus increases IgA+ antibody-secreting cells in the lamina propria . These IgA antibodies likely target both the virus and commensal bacteria, potentially altering microbial community composition and function.

  • Microbiome requirement for antitumor immunity: Notably, the gut microbiome appears essential for orally administered reovirus-induced antitumor immunity, unlike with intratumoral reovirus injection . This suggests that microbiome-antibody interactions may be critical for certain therapeutic applications.

  • MAdCAM-1+ blood vessel involvement: Reovirus interaction with the gut immune system, particularly in Peyer's patches of the terminal ileum, increases IgA+ antibody-secreting cells through MAdCAM-1+ blood vessels . This specific anatomical interaction may facilitate crosstalk between viral immunity, antibody production, and the microbiome.

These findings suggest that the therapeutic efficacy of reovirus, particularly when administered orally, may depend partly on antibody-mediated interactions with the gut microbiome. This represents an important consideration for both basic research and clinical applications of reovirus-based therapies.

What control measures should be implemented when studying reovirus antibodies in animal models?

Rigorous control measures are essential when studying reovirus antibodies in animal models to ensure reliable and interpretable results:

  • Strain and genetic background controls:

    • Use genetically defined animal strains with known susceptibility to reovirus

    • Include transgenic models expressing human disease-relevant genes (e.g., HLA-DQ8 for celiac disease studies)

    • Maintain age and sex-matched controls for all experimental groups

  • Viral controls:

    • Compare multiple reovirus strains with known genetic differences

    • Include UV-inactivated virus controls to distinguish between effects requiring viral replication and those triggered by viral particles alone

    • Quantify and standardize viral doses across experiments using plaque assays

  • Environmental controls:

    • Maintain animals in specific pathogen-free conditions to prevent confounding infections

    • For microbiome studies, control for housing conditions, diet, and antibiotic exposure

    • Consider co-housing or microbiome normalization procedures to reduce cage effects

  • Immunological readout controls:

    • Include isotype controls for all antibodies used in detection assays

    • Use serum from virus-naïve animals as negative controls

    • Include hyperimmune serum or monoclonal antibodies as positive controls

    • For neutralization assays, include non-reovirus viruses (e.g., VSV) to confirm specificity

  • Route of administration considerations:

    • Compare different administration routes (oral, intravenous, intraperitoneal) which may elicit different antibody responses

    • Use vehicle-only administration as a procedural control

Implementing these controls helps isolate reovirus-specific effects from confounding variables and increases the translational relevance of findings to human disease and therapeutic applications.

How should researchers account for cross-reactivity with other viral antibodies when studying reovirus-specific responses?

Accounting for antibody cross-reactivity is crucial for accurate characterization of reovirus-specific immune responses, particularly given the ubiquity of exposure to multiple viruses in both humans and research animals:

  • Pre-absorption studies:

    • Pre-absorb serum samples with related viruses (e.g., rotavirus, other reoviruses) to remove cross-reactive antibodies

    • Compare antibody titers before and after absorption to quantify cross-reactivity

  • Competitive binding assays:

    • Perform competitive ELISA or other binding assays using purified proteins from related viruses

    • Measure the ability of heterologous viral antigens to inhibit binding to reovirus antigens

  • Epitope-specific assays:

    • Use peptide arrays or recombinant proteins representing unique regions of reovirus proteins

    • Focus on viral proteins with higher sequence divergence from related viruses

    • The sigma1 protein, which shows substantial variation between viral strains, is particularly useful for strain-specific detection

  • Multiple virus testing:

    • Test antibody responses against a panel of viruses simultaneously

    • Compare patterns across viruses to identify reovirus-specific responses

    • Research shows that celiac disease patients with high anti-reovirus antibodies did not necessarily have high antibodies against rotavirus or herpes simplex virus

  • Functional assays:

    • Neutralization assays typically offer higher specificity than binding assays

    • Include control viruses (e.g., VSV) in neutralization assays to confirm specificity

Researchers studying the relationship between reovirus and celiac disease have demonstrated the importance of accounting for cross-reactivity by showing that elevated antibody responses were specific to reovirus rather than representing general antiviral hyperreactivity .

What are the key considerations for designing longitudinal studies of reovirus antibody persistence?

Designing effective longitudinal studies to investigate reovirus antibody persistence requires careful attention to several methodological considerations:

  • Sampling frequency and timing:

    • Establish baseline measurements before known exposure when possible

    • Include frequent early sampling to capture peak antibody responses (typically 2-4 weeks post-exposure)

    • Schedule longer-term follow-up at strategic intervals (3, 6, 12 months, and beyond)

    • Consider developmental windows when studying pediatric populations, as age at infection may influence antibody persistence

  • Comprehensive antibody profiling:

    • Measure multiple antibody isotypes (IgG, IgA, IgM) and IgG subclasses

    • Include both binding and neutralizing antibody assays

    • Monitor antibody affinity maturation over time using techniques like avidity assays

    • Assess antibodies against multiple viral proteins to track epitope spreading

  • Biological specimen considerations:

    • Collect and properly store multiple specimen types (serum, plasma, mucosal secretions)

    • Standardize collection procedures and minimize freeze-thaw cycles

    • Consider biobanking additional samples for future novel assays

  • Relevant covariates and confounders:

    • Record intercurrent illnesses and vaccinations

    • Document exposure to related viruses

    • Collect genetic information relevant to immune responses

    • For studies of autoimmune conditions, monitor disease-specific biomarkers alongside antibody titers

  • Statistical and analytical planning:

    • Use mixed-effects models to account for repeated measures

    • Plan for missing data and participant attrition

    • Consider mathematical modeling of antibody decay kinetics

    • Calculate minimum sample sizes needed to detect clinically meaningful changes

The persistent nature of reovirus antibodies and their potential role in long-term immune modulation, as suggested by research on celiac disease , makes well-designed longitudinal studies particularly valuable for understanding the clinical implications of reovirus exposure.

How can researchers effectively neutralize pre-existing reovirus antibodies in experimental settings?

Neutralizing pre-existing reovirus antibodies is particularly important for oncolytic virus research and therapeutic applications where antibodies might interfere with treatment efficacy. Several strategies can be employed:

  • Virus shielding approaches:

    • Encapsulate reovirus in liposomes or nanoparticles to physically shield from antibody binding

    • Use cell carriers (e.g., dendritic cells, T cells) that can transport virus while protecting it from neutralizing antibodies

    • Develop polymer coatings that temporarily mask viral epitopes recognized by antibodies

  • Antibody depletion techniques:

    • Plasmapheresis to physically remove antibodies (applicable in clinical settings)

    • Immunoadsorption columns specific for anti-reovirus antibodies

    • Protein A/G columns for general IgG depletion in experimental samples

  • Competitive binding strategies:

    • Pre-treatment with high doses of non-replicating (UV-inactivated) virus to saturate antibodies

    • Use of recombinant viral proteins as decoys to absorb neutralizing antibodies

    • Development of peptide mimetics that bind to neutralizing antibodies

  • Route of administration optimization:

    • Direct intratumoral injection to bypass systemic antibodies

    • Oral administration, which has shown efficacy in preclinical models despite systemic antibodies

    • Regional delivery methods (e.g., intraperitoneal for peritoneal metastases, intra-arterial for liver tumors)

Each approach has advantages and limitations, and the optimal strategy may depend on the specific research question or clinical application. For instance, research has shown that while neutralizing antibodies impair the direct oncolytic function of reovirus, they may not inhibit its T cell-attracting capacity , suggesting that complete neutralization may not be necessary for all therapeutic applications.

Product Science Overview

Mouse Antibody

Introduction

Mouse antibodies, also known as murine antibodies, are antibodies derived from mice. These antibodies are widely used in research and therapeutic applications due to their ability to target specific antigens .

Types and Production

There are five antibody isotypes in mice: IgA, IgD, IgE, IgG, and IgM, each with a different heavy chain . Monoclonal antibodies (MAbs) are produced by immunizing mice with a target antigen and then harvesting the antibody-producing cells . These cells are fused with myeloma cells to create hybridomas, which can produce large quantities of the desired antibody .

Human Anti-Mouse Antibody (HAMA) Response

One challenge with using mouse antibodies in humans is the Human Anti-Mouse Antibody (HAMA) response . The human immune system can recognize mouse antibodies as foreign and produce antibodies against them, leading to allergic reactions and reduced efficacy of the treatment . Advances in antibody engineering have led to the development of chimeric and humanized antibodies, which are less likely to trigger the HAMA response .

Applications

Mouse antibodies are used in various diagnostic and therapeutic applications. They are employed in immunoassays, imaging studies, and as treatments for diseases such as cancer and autoimmune disorders . Fully human monoclonal antibodies, produced using transgenic mice or in vitro techniques, are becoming increasingly common due to their reduced immunogenicity .

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.