*S. Typhi employs multiple surface antigens to evade immunity, but antibodies targeting these components are critical for defense.
Vi Polysaccharide: IgG1/IgG2 responses after Vi-TT prime-boost; mediates Fc-dependent functions (e.g., phagocytosis, complement deposition) .
O9:LPS: IgG titers correlate with delayed infection onset and reduced bacterial load .
Vi Polysaccharide: Higher IgA avidity predicts protection; enriched in phagocytosis-mediated responses .
O9:LPS/Flagellin: Elevated in acute infections but not protective .
t3090 (Membrane Protein): IgM responses identified as diagnostic markers in endemic settings .
Vi-TT + Vi-PS: Prime-boost regimens induce convergent antibodies with shared V genes and CDR3 regions; IgG1/IgG2 dominate, targeting subdominant epitopes .
Functional Synergy: Vi antibodies with high dissociation kinetics (avidity) enhance Fc effector functions (e.g., neutrophil phagocytosis) .
Multivariate models identify predictive antigen/antibody combinations:
Mechanism: Mediate complement-dependent lysis, reducing bacterial viability .
Clinical Impact: Higher titers delay disease onset, lower bacterial loads, and reduce cytokine storms (e.g., TNF-α, IL-6) .
Limitations: Cross-reactivity with non-typhoidal Salmonella limits specificity; no protective role observed .
This product features a pair of antibodies designed for the specific detection of Salmonella Typhi. The capture antibody binds to the Outer Membrane Protein of S. Typhi, while the conjugating antibody targets a polysaccharide unique to this bacterium. These antibodies are crucial components in various diagnostic assays.
This product serves a dual purpose, functioning as both a capture and conjugating antibody.
The purity level of this antibody exceeds 90%, indicating a high level of quality and reliability.
The antibody solution is sterile, ensuring its suitability for use in various applications. It is clear and colorless in appearance.
This product consists of S. Typhi capture and conjugate antibodies at a concentration of 2.7mg/1ml. It is formulated in a solution of 0.01M phosphate buffered saline with a pH of 7.2 and contains 0.1% sodium azide as a preservative.
To maintain optimal performance, the S. Typhi Paired antibody should be stored at a refrigerated temperature between 2-8 degrees Celsius.
Purified monoclonal IgG by protein A chromatography.
Paired antibody tests for S. typhi involve collecting two serum samples from the same individual at different time points to measure changes in antibody levels. The first sample is typically collected during the acute phase of infection or before vaccination, and the second during the convalescent phase or after immunization. This approach is significant in typhoid research because it enables scientists to distinguish between active infections and past exposures, as well as to evaluate vaccine-induced immune responses.
Methodologically, researchers analyze both samples using the same assay conditions to detect a fourfold or greater rise in antibody titers, which is considered indicative of recent infection or successful immunization. The tests typically measure antibodies against various S. typhi antigens, including the Vi polysaccharide, O antigens, and H antigens. This paired approach compensates for individual variations in baseline antibody levels that might otherwise confound single-sample testing .
Distinguishing between S. typhi and S. paratyphi antibody responses requires careful consideration of the distinct antigenic structures of these related pathogens. While both bacteria can cause enteric fever with similar clinical presentations, their surface antigens differ significantly, allowing for serological differentiation.
S. typhi possesses the Vi capsular polysaccharide, which is absent in S. paratyphi A. Additionally, S. paratyphi A contains the O2-antigen with paratose instead of the O9-antigen with tyvelose found in other Salmonella serovars. These differences enable researchers to develop antigen-specific assays targeting these distinctive structures. When testing paired serum samples, researchers typically use purified Vi polysaccharide to detect S. typhi-specific responses, while O-antigen preparations can help differentiate S. paratyphi A responses .
Cross-reactivity remains a challenge, particularly with common O-antigen epitopes shared among Salmonella serovars. Therefore, researchers often complement serological testing with molecular approaches such as PCR or bacterial culture when definitive differentiation is required.
The optimal time intervals for collecting paired serum samples depend on the specific research question being addressed. For acute infection diagnosis, the first sample should be collected as early as possible after symptom onset (acute phase), and the second sample 2-4 weeks later (convalescent phase).
For vaccine immunogenicity studies, baseline samples are typically collected immediately before vaccination, with follow-up samples collected at multiple time points: 2-4 weeks post-vaccination to capture peak antibody responses, and 6-12 months later to assess persistence of immunity. When evaluating booster responses, as described in the Vi-TT prime followed by Vi-PS boost studies, researchers should collect samples before the prime, before the boost (typically 6-12 months after prime), and 2-4 weeks after the boost to capture the amplification of the immune response .
For longitudinal studies examining waning immunity or breakthrough infections, longer intervals of 1-5 years may be appropriate. Regardless of the chosen intervals, standardization within a study is crucial for valid comparisons between subjects and experimental groups.
The Vi capsule of S. typhi significantly influences antibody binding and detection by functioning as a physical barrier that shields underlying antigens. Research demonstrates that the Vi capsule prevents binding of both IgG and IgM antibodies to O-antigen epitopes that would otherwise be accessible on the bacterial surface .
This shielding effect creates methodological challenges for researchers attempting to detect antibody responses to subcapsular antigens in Vi-positive strains. To overcome this limitation, researchers often employ multiple approaches:
Using Vi-negative mutant strains to expose underlying O-antigens for antibody binding studies
Performing analyses with both intact bacteria and purified antigens (Vi, O-antigen, etc.)
Employing antigen removal techniques to expose hidden epitopes
Characterization of convergent antibody responses after Vi polysaccharide vaccination involves sophisticated molecular and immunological approaches to identify similarities in the B cell repertoire across individuals. Convergent responses are identified by analyzing shared characteristics in the antibody sequences, particularly in the antigen-binding regions.
Methodologically, researchers isolate Vi-specific B cells through antigen-specific cell sorting, followed by single-cell RNA sequencing or paired heavy and light chain sequencing. Analysis focuses on identifying patterns in variable (V) gene usage, complementarity-determining region 3 (CDR3) sequences, and somatic hypermutation profiles. Evidence of convergence appears as restricted V gene usage and shared CDR3 characteristics across multiple individuals who received the same vaccination regimen .
For functional assessment of convergent antibodies, researchers express recombinant monoclonal antibodies from the identified sequences and characterize their binding kinetics using surface plasmon resonance or bio-layer interferometry. This reveals the range of affinities, from low (10 μM) to high (500 pM), that comprise the polyclonal response. Convergent antibodies can then be grouped based on their epitope specificity, such as those recognizing the O-acetylated Vi or the de-O-acetylated Vi backbone .
S. typhi employs multiple sophisticated mechanisms to evade serum antibodies, requiring specialized research methodologies to elucidate these strategies. The primary evasion mechanisms include:
Vi capsule expression: The Vi capsule serves as a physical barrier that masks underlying O-antigens from antibody recognition. To study this mechanism, researchers compare isogenic strains with and without the Vi capsule (controlled through tviB gene manipulation) for serum resistance, antibody binding, and complement deposition using flow cytometry and confocal microscopy .
O-antigen modifications: S. typhi lacks the very long O-antigen seen in S. paratyphi A due to a pseudogene in fepE. Researchers study this through complementation experiments, introducing functional fepE into S. typhi and observing changes in O-antigen patterns using silver-stained SDS-PAGE and Western blotting with specific antibodies .
Reduced complement activation: Vi-encapsulated S. typhi prevents complement activation and C3b deposition. Researchers quantify this by measuring C3a and C5a anaphylatoxins in serum after bacterial exposure using enzyme immunoassays, and by flow cytometric detection of surface-bound C3b .
To comprehensively study these evasion mechanisms, researchers often employ serum resistance assays to correlate molecular modifications with functional outcomes. These assays expose bacteria to human serum and measure survival over time, with isogenic mutants allowing attribution of serum resistance to specific bacterial factors .
To study these subclass distributions, researchers employ isotype-specific ELISA assays with standardized calibration curves for each IgG subclass. Single-cell analysis technologies can further reveal the proportion of plasmablasts secreting each subclass. The shift in subclass distribution has significant implications for protection, as different IgG subclasses engage different Fc receptors on effector cells:
IgG1 antibodies activate complement efficiently and bind with high affinity to FcγRIIa and FcγRIIIa on phagocytes, promoting phagocytosis
IgG2 antibodies are particularly effective against polysaccharide antigens and become more prominent in the mature response to carbohydrate antigens
For functional characterization, researchers employ assays that measure:
Complement deposition using flow cytometry with C3c-specific antibodies
Phagocytosis by monocytes or neutrophils using fluorescently labeled bacteria and flow cytometric quantification
Antibody-dependent cellular cytotoxicity using NK cells and appropriate target cells
These functional assays reveal that the Fc effector functions correlate more strongly with antibody dissociation kinetics than with association kinetics, suggesting that the stability of antibody-antigen complexes is critical for effective immune activation .
S. paratyphi A contains three copies of the rfbV gene (also known as wbaV), which encodes an enzyme that links paratose to mannose in the O-antigen repeating unit. This gene duplication plays a crucial role in immune evasion by enhancing very long O-antigen production, which prevents complement deposition and immunoglobulin binding.
To experimentally demonstrate this role, researchers construct S. paratyphi A strains containing only a single copy of rfbV and compare them to wild-type strains with multiple copies. The following methodological approaches reveal the function of these multiple gene copies:
Serum resistance assays: S. paratyphi A with a single rfbV copy shows increased susceptibility to serum killing compared to wild-type strains. This can be quantified by measuring bacterial survival after exposure to human serum, with serum resistance fully restored by complementation with additional rfbV copies .
Complement deposition assays: Flow cytometry using fluorescently-labeled antibodies against C3b demonstrates increased complement deposition on S. paratyphi A strains with reduced rfbV copy number, establishing a direct correlation between rfbV copy number, very long O-antigen production, and complement evasion .
O-antigen analysis: Silver-stained SDS-PAGE gels followed by densitometry analysis show that S. paratyphi A with multiple rfbV copies produces more very long O-antigen than strains with a single rfbV copy. This provides direct evidence linking rfbV copy number to O-antigen production .
Genetic complementation studies: Constitutive expression of either rfbV alone or rfbV preceded by the hypothetical rfbU-rfbX fusion ORF in single-copy rfbV mutants restores very long O-antigen production and serum resistance, confirming the causal relationship .
These experimental approaches demonstrate that rfbV gene duplication is a key evolutionary adaptation that enhances S. paratyphi A immune evasion through increased production of very long O-antigen, preventing complement activation and antibody binding.
Differentiating between antibody responses to active S. typhi infection versus vaccination presents significant challenges that require careful methodological approaches. Researchers employ several strategies to make this distinction:
Antigen-specific responses: Vaccines like Vi-TT induce strong responses primarily to Vi polysaccharide, while natural infection elicits broader responses to multiple antigens, including flagellin (H antigen), lipopolysaccharide (O antigen), and outer membrane proteins. Researchers can test for antibodies against these diverse antigens to identify patterns consistent with natural infection versus vaccination .
IgG subclass distribution: Natural infection typically induces a mixed subclass response, while Vi polysaccharide conjugate vaccines induce predominantly IgG1 responses. Analysis of IgG subclass distribution using isotype-specific secondary antibodies in ELISA can help distinguish these responses .
Antibody avidity: Infection-induced antibodies often undergo more extensive affinity maturation than vaccine-induced antibodies, particularly those elicited by polysaccharide antigens. Researchers measure avidity using modified ELISA with chaotropic agents like ammonium thiocyanate or urea to disrupt low-avidity antibody binding .
Mucosal antibody detection: Natural infection stimulates mucosal immunity more effectively than parenteral vaccination. Detection of secretory IgA in saliva or other mucosal secretions can indicate natural infection rather than vaccination.
Paired serum kinetics: The kinetics of antibody rise and decline differ between natural infection and vaccination. With paired samples, researchers can analyze these patterns, with natural infection often showing more rapid initial increases followed by quicker declines than vaccine-induced responses.
These approaches should be used in combination rather than in isolation for more reliable differentiation.
Standardization of S. typhi antibody testing across research laboratories faces several methodological challenges that impact result comparability and reproducibility:
Antigen preparation variability: Different methods of preparing Vi polysaccharide and other S. typhi antigens result in variations in purity, O-acetylation levels, and molecular weight, affecting antibody binding characteristics. Standardized reference antigens with defined chemical properties are needed to overcome this challenge .
Assay platform differences: Various platforms (ELISA, multiplex immunoassays, flow cytometry-based methods) with different detection sensitivities and dynamic ranges are used across laboratories. Establishing platform-specific reference standards and providing conversion factors between methods would improve cross-laboratory comparability.
Reference sera availability: Limited availability of internationally standardized reference sera with defined antibody concentrations against S. typhi antigens hampers absolute quantification. WHO International Standards for anti-typhoid antibodies would facilitate standardization across laboratories.
Threshold determination: There is no consensus on antibody level thresholds that correlate with protection, complicating interpretation of results. Multi-center studies correlating antibody levels with protection in endemic settings are needed to establish clinically relevant thresholds.
Cross-reactivity considerations: Antibodies against other Salmonella serovars or enterobacteria may cross-react with S. typhi antigens. Researchers should include absorption steps with heterologous antigens or employ highly specific monoclonal antibody-based competitive assays to improve specificity.
To address these challenges, international collaborative efforts are needed to develop standardized protocols, reference materials, and quality assurance programs specifically for S. typhi antibody testing in research contexts.
The functional characteristics of anti-Vi antibodies that correlate with protection against S. typhi infection extend beyond simple binding to include various effector functions. Current research indicates that antibody functionality, rather than mere concentration, may better predict protective immunity.
The primary functional characteristics that researchers analyze include:
These functional characteristics provide a more comprehensive assessment of protective immunity than antibody titers alone and may explain the variable protection observed in typhoid challenge models despite similar antibody concentrations.
The differential O-antigen production between Salmonella serovars involves several molecular mechanisms that significantly impact their interactions with the host immune system. These differences represent key evolutionary adaptations for distinct ecological niches.
For S. paratyphi A, several molecular mechanisms contribute to its distinct O-antigen pattern:
WzzB functionality: S. paratyphi A contains an Arg98-to-Cys mutation in WzzB that reduces its activity, resulting in diminished long O-antigen production. This hypofunctional WzzB diverts O-antigen precursors toward FepE-dependent very long O-antigen production. Researchers can demonstrate this mechanism through complementation studies with functional WzzB from other serovars and site-directed mutagenesis to restore Arg98 .
Multiple rfbV copies: S. paratyphi A contains three copies of rfbV (wbaV), compared to the single copy in S. typhi and S. typhimurium. This gene encodes an enzyme that links paratose to mannose in the O-antigen repeating unit. The increased gene dosage enhances very long O-antigen production, which can be experimentally verified through genetic manipulation of rfbV copy number and O-antigen analysis by SDS-PAGE .
RfbU-RfbX fusion protein: S. paratyphi A contains a hypothetical open reading frame composed of the C-terminus of RfbX and the N-terminus of RfbU that precedes the extra rfbV copies. This putative fusion protein appears to retain enzymatic activity affecting O-antigen production and is required for tolerating increased RfbV levels .
For S. typhi, key mechanisms include:
fepE pseudogene: S. typhi contains a pseudogene of fepE, which is required for very long O-antigen production. This genetic loss explains the absence of very long O-antigen in S. typhi .
Vi capsule acquisition: S. typhi has acquired the SPI-7 pathogenicity island encoding the Vi capsule, which functionally compensates for the loss of very long O-antigen by providing an alternative barrier to complement and antibody binding .
These molecular differences result in distinct immune evasion strategies: S. typhimurium allows complement activation and inflammatory responses, which it exploits, while S. typhi and S. paratyphi A prevent complement and antibody binding through the Vi capsule and very long O-antigen, respectively, allowing them to evade innate immunity and disseminate via trafficking phagocytes .
Designing experiments to identify correlates of protection for new S. typhi vaccine candidates requires a multifaceted approach that integrates immunological assessments with clinical protection data. The following methodological framework addresses this complex challenge:
Controlled Human Infection Model (CHIM) studies:
Vaccinate cohorts with new candidate vaccines and include control groups
Challenge participants with standardized doses of S. typhi under carefully monitored conditions
Collect comprehensive pre-challenge immune parameters (antibodies, cellular responses)
Track protection outcomes (prevention of bacteremia, symptomatic disease)
Apply statistical methods to correlate immune parameters with protection
Comprehensive antibody profiling:
Measure not only antibody concentrations but also functional characteristics
Assess antibody avidity using surface plasmon resonance or modified ELISA with chaotropic agents
Determine IgG subclass distribution using isotype-specific detection reagents
Map epitope specificity using modified antigens (e.g., differentially O-acetylated Vi)
Quantify Fc-mediated functions including complement activation, phagocytosis, and ADCC
Systems immunology approach:
Perform transcriptomic analysis of peripheral blood at key timepoints post-vaccination
Identify gene signatures that correlate with subsequent protection
Conduct multi-parameter flow cytometry to characterize B and T cell responses
Apply machine learning algorithms to identify immune signature combinations that predict protection
Field efficacy studies with nested case-control analysis:
Conduct vaccine efficacy trials in endemic settings
Collect and biobank samples from all participants prior to typhoid exposure
Identify typhoid cases through enhanced surveillance
Match cases with protected controls from the same population
Compare pre-exposure immune parameters between cases and controls
Passive transfer experiments:
Isolate IgG from vaccinated human subjects
Transfer purified antibodies to animal models
Challenge with mouse-adapted S. typhi strains
Determine the antibody levels and characteristics required for passive protection
This integrated approach enables identification of both simple correlates (single immune parameters associated with protection) and mechanistic correlates (immune responses that directly mediate protection), guiding rational vaccine improvement strategies.
S. typhi Vi antibody responses show marked differences between endemic and non-endemic populations, which has significant implications for vaccine efficacy studies. These differences stem from prior exposure patterns, cross-reactive immunity, and genetic factors influencing immune responses.
Key differences in Vi antibody responses include:
Baseline antibody levels: Individuals in endemic regions often have detectable pre-vaccination Vi antibody levels due to previous subclinical exposures or cross-reactive antigens. Researchers should measure baseline antibodies and consider stratified analysis based on pre-existing immunity when interpreting vaccine responses .
Magnitude of response: Endemic populations typically show lower fold-increases in antibody titers following vaccination compared to non-endemic populations, despite potentially reaching similar final concentrations. This "ceiling effect" necessitates careful consideration of appropriate immunogenicity endpoints.
Antibody persistence: Antibody longevity may differ between populations, with some studies suggesting more rapid waning in endemic settings due to immune regulation mechanisms. Longitudinal sampling (6, 12, 24 months post-vaccination) should be incorporated into study designs to capture these differences.
Functional characteristics: The quality of antibodies may differ substantially, with endemic populations potentially developing antibodies targeting a broader range of epitopes, including subdominant ones like the de-O-acetylated Vi backbone .
Memory B cell responses: Differences in germinal center reactions and memory B cell formation between populations affect recall responses to vaccination or natural exposure. Researchers should include memory B cell ELISpot assays to quantify these differences.
These differences have important implications for vaccine efficacy studies:
Study design: Vaccine studies in non-endemic populations may overestimate immunogenicity compared to target populations in endemic regions.
Sample size calculations: Higher variability in endemic populations requires larger sample sizes to achieve similar statistical power.
Correlates of protection: Correlates established in non-endemic populations may not translate directly to endemic settings, necessitating population-specific validation.
Dosing and scheduling: Optimal vaccination schedules may differ between populations, requiring tailored approaches rather than universal recommendations.
Interpretation of licensure data: Regulatory authorities should consider population-specific immunogenicity data when evaluating vaccines for global use.
Methodologically, researchers should implement identical protocols across sites, include both population types in early-phase trials when possible, and consider innovative trial designs such as cluster randomization or step-wedge approaches to account for herd immunity effects in endemic settings.
Isolation and characterization of S. typhi-specific memory B cells require sophisticated techniques that go beyond traditional serological assays. These advanced approaches enable detailed understanding of the B cell repertoire and can inform vaccine design strategies:
Antigen-specific B cell sorting:
Label Vi polysaccharide or other S. typhi antigens with fluorophores or biotin
Perform multiparametric flow cytometry to identify and sort antigen-binding B cells
Include phenotypic markers (CD19, CD27, IgG/IgM/IgA) to distinguish naive, memory, and plasmablast populations
Use dual-color antigen labeling strategies to increase specificity and reduce false positives
Single-cell B cell receptor (BCR) sequencing:
Recombinant monoclonal antibody expression:
B cell receptor repertoire analysis:
Apply next-generation sequencing to analyze the diversity of the B cell repertoire
Identify convergent sequences across multiple individuals receiving the same vaccine
Map public clonotypes that may represent optimal solutions to Vi antigen recognition
Track clonal expansion patterns following vaccination or infection
B cell functional assays:
Establish B cell-derived immortalized cell lines through Epstein-Barr virus transformation
Analyze antibody secretion patterns using enzyme-linked immunospot (ELISpot) assays
Perform limiting dilution assays to quantify antigen-specific memory B cell frequencies
Assess cytokine production profiles of memory B cells using intracellular cytokine staining
Spatial analysis of lymphoid tissues:
When tissue samples are available (e.g., from animal models), perform multiplex immunofluorescence imaging
Localize S. typhi-specific B cells within germinal centers and other lymphoid structures
Characterize spatial relationships between antigen-specific B cells and T follicular helper cells
These technologies, when applied systematically, provide unprecedented insights into the development and maintenance of S. typhi-specific B cell immunity, enabling rational approaches to vaccine design.
Computational modeling offers powerful approaches for understanding the evolutionary adaptations of S. typhi and S. paratyphi for immune evasion. These in silico methods complement traditional experimental techniques and provide unique insights into pathogen evolution and host-pathogen interactions.
Comparative genomics and phylogenetic analysis:
Whole genome sequence comparison across Salmonella serovars to identify lineage-specific genetic changes
Reconstruction of phylogenetic relationships between typhoidal and non-typhoidal Salmonella
Identification of convergent evolution patterns, such as the independent acquisition of immune evasion mechanisms in S. typhi (Vi capsule) and S. paratyphi A (very long O-antigen)
Analysis of selection pressures on genes involved in O-antigen synthesis and modification
Structural biology and molecular dynamics:
Homology modeling of proteins involved in O-antigen synthesis and modification (e.g., WzzB, FepE, RfbV)
Simulation of the effects of mutations (e.g., the Arg98-to-Cys mutation in S. paratyphi A WzzB) on protein structure and function
Modeling of polysaccharide structures to predict accessibility to antibodies and complement
Virtual screening approaches to identify small molecules that could inhibit immune evasion mechanisms
Systems biology of host-pathogen interactions:
Network analysis of human immune responses to different Salmonella serovars
Identification of key host pathways targeted by bacterial immune evasion mechanisms
Prediction of immune evasion consequences on disease progression and transmission
Integration of transcriptomic, proteomic, and metabolomic data into comprehensive models
Population genetics and epidemiological modeling:
Simulation of the spread of immune evasion traits in bacterial populations under different selection pressures
Analysis of the fitness costs and benefits of specific immune evasion mechanisms
Modeling of the impact of vaccination on pathogen evolution and immune escape variant emergence
Prediction of geographical patterns in strain distribution based on local immune selection pressures
Machine learning approaches:
Development of predictive models for identifying potential new immune evasion mechanisms
Classification of bacterial strains based on their immunomodulatory potential
Feature extraction from complex datasets to identify patterns associated with successful immune evasion
Deep learning applications for predicting antibody-antigen interactions and epitope accessibility
These computational approaches can generate testable hypotheses about the evolutionary pathways that led to the distinctive immune evasion strategies of typhoidal Salmonella, guiding experimental validation and potentially informing the development of countermeasures that target these evasion mechanisms.
Salmonella Typhi is a Gram-negative bacterium responsible for causing typhoid fever, a severe and potentially life-threatening illness. Typhoid fever affects millions of people worldwide, particularly in regions with poor sanitation and limited access to clean water . The development of effective diagnostic tools and treatments is crucial in managing and controlling the spread of this disease.
Monoclonal antibodies (mAbs) are laboratory-produced molecules engineered to serve as substitute antibodies that can restore, enhance, or mimic the immune system’s attack on cells . These antibodies are designed to bind to specific antigens, in this case, the antigens present on the surface of Salmonella Typhi bacteria . Monoclonal antibodies are highly specific, targeting a single epitope on the antigen, which makes them valuable tools in both diagnostic and therapeutic applications .
The Mouse Anti-Salmonella Typhi Paired Antibody is a monoclonal antibody specifically developed to target Salmonella Typhi. This antibody is used in various laboratory research applications, including the detection and quantification of Salmonella Typhi in biological samples . The paired antibody system typically involves two antibodies: a capture antibody and a detection (or conjugate) antibody. The capture antibody binds to a specific antigen on the Salmonella Typhi bacterium, while the detection antibody binds to another epitope on the same antigen, allowing for precise identification and measurement .
The production of monoclonal antibodies involves the fusion of myeloma cells with spleen cells from a mouse immunized with the target antigen. This fusion creates hybridoma cells that can produce large quantities of the desired antibody . The antibodies are then purified using techniques such as protein A chromatography to ensure high purity and specificity .