The search included:
None of these sources mention "AAE18 Antibody" or similar nomenclature.
The name "AAE18" does not align with standard antibody nomenclature (e.g., "AAE" is not a recognized prefix for antibodies in the International Nonproprietary Name [INN] system).
Possible typographical errors (e.g., "AAV8" , "EY6A" , or "C002" are documented antibodies).
If "AAE18" is a proprietary or experimental candidate, it may not yet be published in open-access databases or may be under confidential development.
To resolve this discrepancy:
Verify the compound name for accuracy (e.g., cross-check patent filings or internal documentation).
Expand the search to proprietary databases (e.g., Cortellis, ClinicalTrials.gov) or direct collaboration with academic/industry researchers.
Provide additional context, such as:
Target antigen or disease indication
Developmental stage (preclinical/clinical)
Institution or company associated with the antibody
While "AAE18" is unconfirmed, the search revealed structurally and functionally characterized antibodies that may serve as analogs:
When conducting research with AAV vectors, researchers must consider two main types of antibodies: binding antibodies (BAbs) and neutralizing antibodies (NAbs). Binding antibodies recognize and attach to AAV capsid proteins but may not necessarily inhibit vector function. Neutralizing antibodies specifically prevent the vector from transducing target cells, effectively neutralizing its therapeutic potential . Additionally, researchers should consider cell-mediated immunity responses, which can be detected using enzyme-linked immunospot (ELISpot) assays to identify AAV capsid-specific T-lymphocyte responses . These different immune responses have varying implications for gene therapy efficacy and safety profiles.
Pre-existing immunity against AAV serotypes is remarkably common. According to multicenter epidemiologic studies, neutralizing antibodies against AAV8, AAV2, and AAV5 are present in approximately 46.9%, 53.1%, and 53.4% of adult participants, respectively . These values tend to remain relatively stable over time. Co-prevalence of neutralizing antibodies to at least two serotypes occurs in approximately 40% of individuals, while around 38.2% have antibodies against all three common serotypes (AAV2, AAV5, and AAV8) . The seroprevalence can vary substantially between different AAV types, with lower binding antibody levels detected against AAV6, AAV5, AAV12, and AAV9, while higher levels are typically found against AAV10, AAV8, AAV1, and AAV2 .
Research indicates that demographic factors may influence anti-AAV antibody prevalence in specific ways. While sex does not appear to have a significant impact on binding or neutralizing antibody levels, age shows a more notable correlation . Studies have observed a trend toward higher binding antibody levels in older individuals against certain AAV types and a clear positive correlation between neutralizing antibody titers and age . Interestingly, disease status relating to neuromuscular disorders does not appear to have a meaningful impact on antibody levels, suggesting that age rather than specific health conditions may be the more influential factor in determining pre-existing immunity to AAV vectors .
The influence of peptide modifications or shuffling of AAV capsids on antibody binding and neutralization is complex and appears to be dependent on the parental AAV serotype . These modifications can alter the antigenic profile of the vector, potentially reducing recognition by pre-existing antibodies. Research indicates that some modified AAV capsids demonstrate significantly lower binding of neutralizing antibodies compared to their wild-type counterparts . This phenomenon varies considerably between different modifications and parental serotypes, suggesting that careful selection and engineering of AAV capsids may provide opportunities to circumvent pre-existing immunity. Researchers should conduct comprehensive serological screening when developing novel AAV variants to identify those with reduced immunogenicity profiles.
Charge detection mass spectrometry (CD-MS) represents a cutting-edge approach for investigating antibody binding to AAV capsids at the molecular level . Unlike traditional methods, CD-MS provides a label-free approach that allows monitoring of individual binding events, with each event indicated by a shift of the antibody-antigen complex to a higher mass. This technique uniquely reveals the distribution of antibodies bound on capsids, enabling identification of AAV subpopulations with different affinities . Interestingly, CD-MS studies have revealed unexpected phenomena, such as a substantial decrease in charge upon binding of the first antibody to AAV8, suggesting significant structural changes, followed by charge increases with subsequent binding events . This method also allows observation of agglutination at high antibody concentrations, where antibodies can link AAV capsids to form dimers and higher-order multimers.
A comprehensive screening approach for anti-AAV antibodies should include multiple complementary assays. The primary testing schematic typically involves a sequence of assays starting with screening assays, followed by confirmation and titration assays when applicable . For humoral immunity, enzyme-linked immunosorbent assay (ELISA) is commonly used to detect binding antibodies, with serial dilutions of purified monoclonal antibodies incubated with immobilized AAV proteins or particles . The relative affinity is determined by measuring the concentration required to achieve EC50 . For neutralizing antibodies, cell-based neutralization assays using AAV pseudoviruses are the gold standard . Additionally, cell-mediated immunity should be assessed using ELISpot assays to detect AAV capsid-specific T-lymphocyte responses . Surface plasmon resonance (SPR) can provide complementary kinetic data on antibody-antigen interactions, measuring association (kon) and dissociation (koff) rates to determine binding affinity (Kd) .
The analysis and reporting of immunogenicity data for anti-drug antibodies (ADAs) in clinical trials should follow a structured approach. According to current guidelines, immunogenicity data should be converted from SDTM (Study Data Tabulation Model) format to CDISC ADaM (Clinical Data Interchange Standards Consortium Analysis Data Model) structure to support various analysis requirements . Key parameters to include in analysis datasets are:
These parameters enable comprehensive characterization of the immunogenicity profile and support regulatory reporting requirements.
Researchers can employ several strategies to overcome pre-existing immunity against AAV vectors:
Alternative Serotype Selection: Utilizing AAV serotypes with lower prevalence of pre-existing antibodies, such as AAV5, AAV12, and AAV9, which show lower neutralizing antibody levels in population studies .
Capsid Engineering: Developing peptide-modified or shuffled AAV capsids that may evade recognition by pre-existing antibodies. The effectiveness of this approach appears to be dependent on the parental AAV serotype .
Patient Screening: Implementing comprehensive screening protocols to identify patients with low pre-existing antibody titers against the intended AAV vector before enrollment in clinical trials .
Local Administration: When feasible, delivering AAV vectors to immunoprivileged sites such as the corpus vitreum or brain can circumvent systemic immunity to a high degree .
Immunomodulation: Temporary immunosuppression regimens may help reduce the impact of pre-existing immunity or prevent the development of treatment-induced immunity.
Vector Dose Adjustment: For patients with low to moderate antibody titers, increasing the vector dose may overcome neutralization, though this approach must balance efficacy with potential toxicity concerns.
Different AAV serotypes demonstrate distinct pre-existing immunity profiles in human populations. Among wild-type capsid AAVs, the lowest binding antibody levels are typically detected against AAV6, AAV5, AAV12, and AAV9, while the highest binding antibody levels are found against AAV10, AAV8, AAV1, and AAV2 . For neutralizing antibodies, AAV12, AAV5, AAV9, AAV7, AAV8, and AAV10 demonstrate the lowest levels, while AAV13, AAV2, and AAV3 show the highest neutralizing antibody levels . These serological differences provide important considerations for vector selection in gene therapy applications, particularly for systemic administration routes. Researchers should consider these pre-existing immunity profiles alongside the tissue tropism and transduction efficiency characteristics of each serotype when designing therapeutic approaches.
Accurate detection and characterization of cellular immune responses against AAV vectors typically employ enzyme-linked immunospot (ELISpot) assays to identify AAV capsid-specific T-lymphocyte responses . Studies indicate that approximately 38.3% of participants have detectable cell-mediated immunity by ELISpot, although interestingly, no consistent correlations have been observed between these cellular responses and humoral responses . Additional methods to characterize cellular immunity may include:
Intracellular cytokine staining and flow cytometry to identify cytokine-producing T cells after AAV peptide stimulation
Lymphocyte proliferation assays to measure T cell activation in response to AAV antigens
Cytotoxicity assays to assess the functional activity of AAV-specific cytotoxic T lymphocytes
MHC tetramer analysis to quantify antigen-specific T cells
These complementary approaches provide a comprehensive assessment of cellular immunity, which is particularly important given the potential for cell-mediated responses to eliminate transduced cells even in the absence of neutralizing antibodies.
The design and characterization of multispecific antibodies for therapeutic applications involve several key considerations and methodological approaches. Based on successful examples like trispecific antibodies targeting HIV-1, researchers should:
Rational Design Strategy: Engineer multispecific antibodies in appropriate formats (such as DVD-Ig format) that allow a single molecule to interact with multiple independent targets . This typically involves cloning sequences for scFvs in frame with sequences encoding connecting linkers (such as G4S linkers) on both the N and C termini of the full IgG1 antibody .
Construct Optimization: Test different combinations of variable domains and linker sequences to identify optimal configurations that maintain the binding activity of each component antibody.
Expression System Selection: Utilize appropriate expression systems (such as HEK293F cells) with optimized transfection ratios of heavy and light chain plasmids (e.g., 1:1.5 molar ratio) .
Comprehensive Binding Analysis: Employ ELISA to evaluate binding activity against all target antigens, confirming that the multispecific construct maintains favorable binding activity to each intended target .
Functional Characterization: Conduct functional assays relevant to the therapeutic application, such as neutralization assays for anti-viral antibodies.
Biophysical Characterization: Assess properties like thermal stability, aggregation propensity, and solution behavior to ensure manufacturability and storage stability.
In vivo Validation: Perform in vivo experiments in appropriate animal models to confirm efficacy and pharmacokinetic properties.
This systematic approach ensures that multispecific antibodies maintain the desired binding and functional properties of each constituent antibody while offering enhanced therapeutic potential through multi-target engagement.
When designing clinical trials involving AAV vectors, researchers should prioritize several key considerations to optimize safety and efficacy:
Pre-existing Immunity Screening: Implement comprehensive screening for pre-existing neutralizing antibodies against the selected AAV serotype, as high levels can prevent efficient gene transduction .
Serotype Selection: Choose AAV serotypes with lower prevalence of pre-existing antibodies in the target population, such as AAV5, AAV12, or AAV9, or consider engineered capsids designed to evade immune recognition .
Administration Route: Consider the implications of administration route, as systemic application by intravenous infusion faces greater immune challenges than local administration to immunoprivileged sites .
Immunogenicity Monitoring: Include robust immunogenicity monitoring throughout the trial, with standardized analysis and reporting of anti-drug antibody data following regulatory guidelines .
Age Stratification: Consider age-based stratification of participants, as research shows a positive correlation between neutralizing antibody titers and age .
Longitudinal Monitoring: Implement longitudinal monitoring of immune responses, as approximately 10% of initially seronegative individuals may develop neutralizing antibodies within one year .
Cellular Immunity Assessment: Include evaluation of cell-mediated immunity alongside humoral responses, as approximately 38.3% of individuals have detectable cellular responses that may impact long-term expression .
These considerations help ensure that AAV-based gene therapy trials are designed to maximize the potential for successful outcomes while addressing the significant challenge of host immune responses.