| Parameter | Description |
|---|---|
| Tissue Panels | 37 human tissues tested, including liver, kidney, and lymphoid organs. |
| Antibody Concentrations | Low (e.g., 1 μg/mL) and high (e.g., 100 μg/mL) to assess affinity and saturation. |
| Controls | Negative and positive controls to differentiate specific vs. non-specific binding. |
| Analysis | Slide evaluation, peer review, and assessment of immune complex deposition. |
Early Risk Mitigation: Identifies off-target effects before clinical trials.
Regulatory Compliance: Required for IND/CTA submissions.
Mechanistic Insights: Reveals antibody binding patterns in diverse tissues.
CRL-2335 is a well-characterized cell line derived from African American patients, used to study basal-like breast carcinoma (BLBC). Key features include:
| Feature | Detail |
|---|---|
| ER/PR/HER2 Status | ER-negative, PR-negative, HER2-negative. |
| Biomarkers | EGFR and cytokeratin 5/6 (CK5/6) positive. |
| Applications | Validating therapeutic antibodies targeting BLBC-associated antigens. |
Protein vs. Transcriptional Analysis: qPCR for EGFR showed high transcriptional activity, but protein staining was absent, highlighting the need for protein-level validation in diagnostics.
Immunofluorescence Confirmation: ER negativity was confirmed via immunofluorescence, distinguishing it from ER-positive cell lines like CRL-2315.
While not branded as "CRL Antibodies," several mAbs developed or tested by CRL-affiliated partners are listed in global databases. Examples include:
| Antibody Name | Target | Application | Approval Status |
|---|---|---|---|
| Levilimab | IL-6R | COVID-19-related inflammation | Russia (2020) |
| RabiShield | Rabies virus G | Rabies exposure prophylaxis | India (2016) |
| Cipterbin | HER2 | HER2+ metastatic breast cancer | China (2020) |
Engineering: Levilimab includes mutations (E233P/L234V/L235A) to reduce Fc effector function.
Therapeutic Modalities: RabiShield is an unconjugated IgG1, while Cipterbin is humanized.
CRL contributed to SARS-CoV-2 detection methods, including saliva-based testing and serology.
| Method | Key Findings |
|---|---|
| Saliva vs. Nasal Swabs | Saliva detected 100% of positive cases vs. 55% for nasal swabs. |
| Viral Persistence | Detectable SARS-CoV-2 in saliva up to 4 weeks post-symptom onset. |
| Time-of-Day Variability | Higher viral loads in morning samples. |
IgM/IgG Discrimination: CRL used Roche’s Anti-SARS-CoV-2 test to differentiate active (IgM) vs. past (IgG) infections.
CRL’s TCR studies align with broader industry trends in antibody therapeutics:
Preclinical Validation: TCR studies ensure mAbs like Loncastuximab tesirine (Zynlonta) meet safety benchmarks.
Regulatory Support: Compliance with FDA/EMA guidelines for IND submissions.
Nanobodies: Extended CDR3 loops (e.g., camelid-derived antibodies) may enhance binding to complex epitopes.
Somatic Hypermutation: Critical for affinity maturation in therapeutic mAbs.
Off-Target Effects: CRL’s TCR studies highlight the need for rigorous preclinical testing.
Global Accessibility: Antibodies like Levilimab (Russia) and Cipterbin (China) underscore regional disparities in therapeutic access.
Biomarker Integration: Combining TCR data with serum antibody profiles (e.g., CRL’s COVID-19 workflow) could optimize patient stratification.
The following studies highlight the crucial role of the target protein in plant development and stress response:
Four main serological assay types have been validated for SARS-CoV-2 antibody detection, each with distinct methodological approaches. The enzyme-linked immunosorbent assays (ELISAs) typically target either the nucleocapsid (N) protein or the trimeric spike (S) protein ectodomain. The S-Flow assay recognizes the spike protein expressed at the cell surface using flow cytometry, while the luciferase immunoprecipitation system (LIPS) recognizes diverse viral antigens including the S1 domain and carboxyl-terminal domain of the nucleocapsid protein .
These assays show similar results but differ in their sensitivity based on technique and target antigen. ELISA methods require recombinant antigens produced either in bacterial systems (for N protein) or human cells (for S protein), while flow-based methods like S-Flow can detect antibodies binding to native conformation antigens expressed on cell surfaces, potentially offering advantages for certain research applications .
In hospitalized COVID-19 patients, seroconversion typically occurs between 5 and 14 days after symptom onset. Studies have documented a median time of 5 to 12 days for anti-S IgM antibodies and 14 days for anti-S IgG and IgA antibodies. The kinetics of anti-N antibody responses are generally similar to anti-S responses, though some research suggests N responses might appear earlier .
The timing of antibody detection depends significantly on the assay methodology. Longitudinal sampling studies have shown that seropositivity may appear as early as 6-7 days after symptom onset with sensitive assays like S-Flow, while other methods may require 10-14 days to detect a positive response .
Research using CRL Rapid Response™ tests analyzing over 460,000 samples collected from July 2020 to January 2021 demonstrated that viral load in asymptomatic and symptomatic COVID-19 cases is remarkably similar. This finding has important methodological implications for studies measuring antibody responses, as it suggests that asymptomatic individuals may mount immune responses comparable to those with symptoms .
The similar viral loads also indicate that molecular tests using saliva samples are equally effective for detecting SARS-CoV-2 in both symptomatic and asymptomatic individuals, providing researchers flexibility in sample collection approaches .
Comprehensive antibody characterization for research applications must document four critical elements:
Confirmation that the antibody binds to the intended target protein
Validation that the antibody recognizes the target protein within complex protein mixtures (e.g., cell lysates or tissue sections)
Evidence that the antibody does not cross-react with proteins other than the intended target
Documentation that the antibody performs as expected under the specific experimental conditions used in the assay
The "antibody characterization crisis" has revealed that approximately 50% of commercial antibodies fail to meet even basic characterization standards, resulting in estimated financial losses of $0.4-1.8 billion annually in the United States alone. This underscores the importance of thorough characterization before conducting antibody-based research .
Studies examining concordance between major commercial antibody platforms demonstrate high agreement rates, though not perfect correlation. In a comprehensive evaluation of 24,079 participants, the percent agreement between Abbott and EuroImmun (EI) assays was 98.8% (95% CI: 98.7%-99.0%) .
| Assay Combination | Probability-weighted Agreement (%) | 95% Confidence Interval |
|---|---|---|
| Ortho and Roche | 98.4% | 97.9%-98.9% |
| EI and Ortho | 98.5% | 92.9%-99.9% |
| Abbott and Roche | 98.9% | 90.3%-100.0% |
| EI and Roche | 98.9% | 98.6%-100.0% |
| Abbott and Ortho | 98.4% | 91.2%-100.0% |
This high concordance suggests that researchers can generally expect consistent results across platforms when samples have robust antibody responses, though discordant results may occur in cases of low-titer antibodies or early seroconversion .
Several major international efforts have been launched to improve antibody characterization, particularly for human proteome research. These include:
The Protein Capture Reagents Program (PCRP) - Focused on generating and characterizing monoclonal antibodies, producing 1,406 antibodies targeting 737 human proteins
The Affinomics program - An EU-funded initiative growing from earlier projects (ProteomeBinders and AffinityProteome) focused on generating and validating protein binding reagents
The Research Resource Identifier (RRID) program - Addressing reagent traceability across research publications
The Developmental Studies Hybridoma Bank (DSHB) - A repository making antibodies available to researchers
These initiatives highlight the substantial resources required for proper antibody characterization, including generating high-quality antigens, developing appropriate recombinant antibodies, identifying high-affinity and highly specific reagents, and making validation data publicly accessible .
Researchers have multiple options for antibody quantification depending on their specific research questions. For detection of anti-SARS-CoV-2 antibodies, methods range from semi-quantitative ELISA assays to more specialized techniques:
ELISAs targeting N or S proteins - Provide semi-quantitative results based on optical density readings
S-Flow assay - Quantifies antibody binding using mean fluorescence intensity values from flow cytometry
LIPS assay - Offers relative quantification through luminescence readings from immunoprecipitated complexes
Neutralization assays - Measure functional antibody activity against live virus or pseudotyped particles, providing information on antibody potency
Within a study of 32 participants who tested positive by at least two immunoassays, 21 had quantifiable anti-SARS-CoV-2 antibody concentrations measured by specialized research assays, demonstrating the feasibility of precise quantification in addition to binary positive/negative results .
The CDC recommends a sequential testing approach when the first test yields a positive result, particularly in contexts where the prevalence of SARS-CoV-2 is low. This approach increases specificity and reduces false-positive results, which is especially important for seroprevalence studies .
Previous comparisons of serological assays with different target antigens have shown substantial variability in performance characteristics even when using the same positive control specimens and pre-pandemic negative controls. A sequential testing strategy helps mitigate these assay-specific limitations and validates positive results .
Understanding differences in antibody responses between asymptomatic and symptomatic cases requires careful study design. Research has shown that anti-SARS-CoV-2 antibody titers correlate with disease severity, likely reflecting higher viral replication rates and immune activation in severe cases .
For studying asymptomatic cases, researchers should consider:
Paired sampling of asymptomatic and symptomatic individuals with similar viral loads
Employing multiple assay methodologies to detect potentially lower antibody titers
Extending follow-up sampling timepoints, as some studies have detected seropositivity in only 32% of mildly symptomatic individuals within 15 days of symptom onset
Including neutralization assays to determine whether lower antibody titers in asymptomatic cases still confer functional protection
Whether asymptomatic SARS-CoV-2 infections lead to protective immunity and whether this immunity is mediated by neutralizing antibodies remain crucial questions requiring careful methodological approaches .
Research using the CRL Rapid Response™ test has demonstrated that molecular tests using saliva samples are equally effective for detecting viral load in both symptomatic and asymptomatic individuals. This finding has several methodological advantages for research studies :
Non-invasive sample collection, potentially increasing participation rates
Reduced requirements for personal protective equipment compared to nasopharyngeal swabbing
Potential for self-collection, enabling remote or community-based study designs
Comparable analytical sensitivity to traditional sampling methods
Applicability across participant cohorts regardless of symptom status
These advantages make saliva-based testing particularly valuable for large-scale seroprevalence studies or longitudinal monitoring of antibody development where repeated sampling is necessary .
When researchers encounter discordant results between different assay platforms, several analytical approaches can be employed:
Consider the target antigens - Discordances may reflect genuine differences in antibody populations (e.g., anti-N vs. anti-S antibodies)
Evaluate timing - Early in seroconversion, more sensitive assays may detect antibodies while others remain negative
Examine quantitative results - "Borderline" results near assay cutoffs are more likely to be discordant across platforms
Employ a third method - Testing with an additional platform can help resolve ambiguous results
In a comprehensive study of 24,079 participants, researchers identified only 277 discordant results between Abbott and EuroImmun assays, demonstrating relatively rare disagreement between well-validated platforms .
Despite significant progress, several challenges remain in standardizing antibody characterization:
The market for antibodies has grown exponentially from ~10,000 commercially available antibodies 15 years ago to more than six million today, outpacing characterization efforts
Many antibodies target the same proteins, creating redundancy without improved quality
Insufficient training for researchers in selecting and validating antibodies for specific applications
Limited incentives for commercial vendors to provide comprehensive characterization data
Lack of standardized reporting formats for antibody validation across publications
Addressing these challenges requires coordinated efforts across stakeholders including researchers, universities, journals, antibody vendors, scientific societies, and funding agencies .
Neutralizing antibodies represent a critical functional measure of protective immunity. Several approaches are used to quantify these responses:
Plaque neutralization assays - The gold standard using live SARS-CoV-2 virus (requires BSL-3 facilities)
Microneutralization assays - Measure inhibition of viral cytopathic effects in cell culture
Pseudotype virus neutralization - Uses lentiviral particles carrying the SARS-CoV-2 S protein (can be performed in BSL-2 facilities)
Surrogate neutralization assays - Measure inhibition of receptor binding without live virus
Research has shown that high anti-SARS-CoV-2 antibody titers are associated with neutralization activity. Potent neutralizing monoclonal antibodies specifically targeting the receptor-binding domain (RBD) of the S protein have been cloned from individuals infected with SARS-CoV-2, providing important reagents for research and potential therapeutic applications .