1. Introduction
Botulinum toxin type A (BoNT-A) is widely recognized for its clinical applications in neurology, pain management, and aesthetic medicine [
1]. Despite its efficacy, a significant challenge in long-term BoNT-A therapy is the development of neutralizing antibodies (NAbs), which can lead to treatment resistance and reduced therapeutic outcomes [
2,
3]. Immunogenicity in biologics, including BoNT-A, is influenced by human leukocyte antigen (HLA) molecules, which present antigenic peptides to T cells, initiating an immune response that can lead to the production of anti-drug antibodies (ADAs) [
4]. The degree of immune recognition varies among individuals due to genetic diversity in HLA alleles [
5], making it imperative to understand these interactions to mitigate immunogenicity risks in BoNT-A therapy.
Although much has been claimed regarding the immunogenicity of different BoNT-A formulations, there remains a considerable lack of comprehensive and conclusive research [
6,
7,
8,
9,
10,
11,
12,
13,
14,
15,
16,
17,
18]. Many assertions rely on speculative claims and counterclaims, often asserting the superiority of one formulation over another without substantial molecular or clinical validation in the absence of robust scientific data [
19,
20,
21,
22]. The absence of standardized methodologies in assessing BoNT-A immunogenicity has resulted in inconsistent findings, making it difficult to ascertain the true extent of immunogenic responses in different formulations. This gap in knowledge necessitates an evidence-based approach that moves beyond commercial biases and focuses on objective scientific analysis.
Computational immunogenetics provides a helpful approach for investigating the immunogenic potential of biologic proteins by integrating bioinformatics, structural modeling, and immune prediction tools. In silico techniques like epitope mapping, molecular docking, and HLA binding prediction enable identification of peptide regions likely to be presented by MHC molecules and recognized by T cells. These tools also allow for the visualization of antigen–receptor interfaces and analysis of structural features such as solvent-accessible surface area (SASA), both of which are essential for understanding immune recognition [
23,
24,
25]. These approaches have been successfully applied in the development of peptide-based vaccines, such as for SARS-CoV-2, and in the assessment of immunogenicity in therapeutic monoclonal antibodies and enzyme replacement therapies [
26,
27,
28].
In the context of BoNT-A, computational analyses facilitate the prediction of epitopes with strong binding affinities to HLA class II alleles, helping to identify immunodominant regions that may contribute to variable patient responses. These tools make it possible to assess T-cell epitope recognition and allele-specific HLA presentation—key determinants of ADA formation and immune-mediated treatment failure. The ability to forecast immunogenicity at the individual level has significant implications for personalized medicine, enabling the design of BoNT-A formulations tailored to minimize immunogenic risk and enhance therapeutic safety [
29,
30,
31,
32].
There has been an increasing suggestion of HLA-DQA1*01:02 and HLA-DQB1*06:04 in BoNT-A immunogenicity, indicating a potential role in antigen presentation and immune activation. The involvement of these specific alleles in biologic immunogenicity has been observed in various therapeutic contexts, suggesting a need for further investigation [
33]. These alleles have been linked to enhanced immune recognition of specific protein structures, due to subtle conformational changes in the protein structure, raising questions about whether certain BoNT-A epitopes are more readily presented to the immune system, contributing to NAb formation. Understanding these genetic associations could provide valuable insights into why certain individuals develop stronger immune responses to BoNT-A than others and whether specific regions of the toxin, particularly those involved in receptor binding and enzymatic function, serve as immunogenic hotspots.
Additionally, the role of specific BoNT-A epitopes in triggering immune recognition remains an area of ongoing inquiry and remains under-researched. While the structure of BoNT-A is highly conserved, subtle variations in epitope accessibility may influence how efficiently HLA molecules, based on allele variations, present antigenic regions to immune cells. It has been proposed that some epitopes located within the heavy chain could exhibit higher immunogenic potential due to their exposure during toxin trafficking [
34]. Meanwhile, epitopes in the light chain, particularly those involved in enzymatic cleavage, may also elicit immune responses depending on how they interact with antigen-presenting cells [
35]. Identifying which epitopes contribute most significantly to ADA formation is a critical aspect of understanding BoNT-A immunogenicity.
Considerable debate exists regarding the role of non-toxic neurotoxin-associated proteins (NAPs), i.e., accessory proteins, in modulating BoNT-A immunogenicity. While some argue that these proteins enhance the activity of BoNT-A [
36,
37,
38], certain reports suggest that NAPs could contribute to antibody formation, potentially triggering immune responses rather than mitigating them [
21,
39]. The precise role of these proteins remains controversial, and despite claims of their protective function, no definitive molecular mechanism has been provided to explain how they might shield BoNT-A from immune detection. The ongoing debate underscores the need for empirical investigations that rely on molecular and structural assessments, rather than assumption-based conclusions regarding the impact of accessory proteins.
Although computational immunogenetics has been critiqued for its lack of experimental follow-through, it remains a powerful approach for hypothesis generation and mechanistic insight. By addressing unanswered molecular questions, such in silico frameworks can effectively guide and prioritize future experimental validation. This study aims to provide a rigorous and objective in silico analysis of BoNT-A immunogenicity, leveraging computational immunogenetics to identify and validate immunogenic epitopes. Through a detailed investigation of HLA–epitope interactions, this study seeks to clarify the mechanisms underpinning BoNT-A immune recognition and contribute to a more evidence-driven approach to biologic formulation and administration.
3. Discussion
The findings of this study offer important insights into the immunogenicity of BoNT-A, particularly in relation to its interaction with HLA class II proteins derived from allele variations and the influence of accessory proteins on antigen presentation. The computational analyses revealed that specific epitopes, including L11, N25, and C10, exhibit strong binding affinities with HLA-DQA1*01:02, HLA-DQB1*06:04, and HLA-DQA1*03:03.
The present computational and structural analyses refute the oversimplified notion that NAPs shield BoNT-A from immune detection. SASA calculations confirm that epitopes remain exposed, which may lead to immune recognition, while electrostatic potential mapping indicates that NAPs do not significantly alter BoNT-A’s charge distribution in a way that would enhance immune recognition.
The analysis further demonstrates that while NAPs do not completely obscure BoNT-A epitopes, they also do not enhance antigenicity in a statistically significant manner. Although subtle electrostatic modifications were observed, they did not correlate with increased immunogenicity (p > 0.05). These findings suggest that NAPs do not enhance neutralizing antibody production, though they may modulate antigen presentation, potentially leading to non-neutralizing antibody formation, a hypothesis requiring further study.
The claims surrounding NAPs and BoNT immunogenicity—whether advocating for their protective role or suggesting an immunogenic influence—must be critically evaluated using scientific evidence rather than assumption or marketing narratives which are not supported by robust scientific evidence. The emphasis on “purity” in certain formulations as an immunological advantage remains unsubstantiated [
40]. Transparent, comparative studies should take precedence over commercial rhetoric, ensuring that clinical and regulatory decisions are guided by rigorous, evidence-based assessments rather than unverified claims.
In parallel, neurotoxin-associated proteins (NAPs), particularly HA-33, have been suggested to enhance the endopeptidase activity of BoNT-A and BoNT-E, though the mechanistic basis remains unverified and renders the claims no more than a hypothesis [
36,
37]. Experimental studies report increased catalytic efficiency with in vitro assays and synaptosomal cleavage models, fueling speculation that NAPs influence BoNT function beyond their established structural role. However, these findings remain inconclusive and require careful interpretation and, on the whole, remain scientifically unsubstantiated. The absence of reducing agents in some experiments raises the possibility that HA-33 stabilizes BoNT rather than actively enhancing enzymatic function. Without direct structural validation, it is unclear whether HA-33 interacts with the active site or merely preserves BoNT’s functional conformation.
The continued success of BoNT-A as a therapeutic agent depends on addressing challenges such as variability in patient responses, immunogenicity, and the limitations of pharmacovigilance systems [
41,
42]. The pharmaceutical industry must move beyond unverified claims surrounding accessory proteins and instead lean into the wealth of long-term clinical data affirming the safety and efficacy of BoNT-A formulations. This shift requires a commitment to objective, science-driven discourse, rather than reliance on assumptions and unsubstantiated hypotheses about the role of accessory proteins in immunogenicity.
A major strength of this study is the comprehensive integration of computational techniques, including molecular docking, epitope–HLA interaction analyses, and dynamic structural assessments. The use of multiple predictive models strengthens the reliability of the findings, offering a nuanced view of BoNT-A’s immunogenic profile and direct scientific investigations. However, despite the robustness of these computational predictions, the absence of direct experimental validation remains a limitation. Further in vitro and in vivo studies are required to confirm the predicted interactions and assess their clinical significance.
The implications for clinical practice are substantial. By identifying immunogenic epitopes with high HLA affinity, this study supports the potential for HLA-based patient stratification, where genotyping could guide personalized BoNT-A therapy, minimizing the risk of immune response development. Additionally, the findings challenge the widely accepted belief that formulations containing accessory proteins inherently offer superior immunogenic protection. This reinforces the need for head-to-head clinical trials that directly compare formulations with and without NAPs, ensuring that treatment decisions are driven by clinical efficacy rather than commercial positioning.
Although HLA-based stratification holds promise for improving treatment outcomes, it also raises ethical considerations regarding equitable access to care. The implementation of genotype-driven therapeutic decisions must be guided by clear ethical frameworks to avoid reinforcing disparities in healthcare access or prioritization based on genetic profiles. These approaches must be validated in diverse populations and accompanied by policies that ensure clinical utility does not translate into clinical exclusion.
Future research should prioritize the experimental validation of the identified epitope–HLA interactions through assays such as Enzyme-Linked ImmunoSpot (ELISpot), T-cell proliferation assays, and structural Cryo-Electron Microscopy (cryo-EM) [
43] studies to confirm the precise nature of NAP–epitope interactions. Additionally, long-term pharmacovigilance studies assessing antibody responses in BoNT-A-treated individuals will be critical in determining whether non-neutralizing antibodies influence therapeutic longevity or alter treatment efficacy over time. Expanding genetic association studies will further refine the understanding of HLA-driven immunogenicity, broadening insights into patient-specific immune responses.
5. Methods
5.1. Computational Resources
This study employed a comprehensive immunoinformatics workflow involving a suite of specialized tools for structural modeling, epitope prediction, molecular docking, and statistical analysis. To enhance clarity, details of each software tool—including version numbers, functions, and sources—are presented within their relevant methodological sections (
Section 5.1,
Section 5.2,
Section 5.3,
Section 5.4 and
Section 5.5). A summary table listing all computational tools used in this study, including the detailed workflow, is provided in
Supplementary Material SC S1.
5.2. Epitope Identification and Cross-Validation
The immunogenic epitopes conveyed by allele variation to BoNT-A were identified and validated using computational epitope mapping and structural analysis. Extensive research by Atassi et al. systematically mapped epitopes within BoNT-A that were recognized as antigenic determinants [
34]. These epitopes serve as critical reference points for evaluating immunogenic potential through in silico methodologies. The epitopes are distributed across the heavy chain (HC) and light chain (LC), encompassing functional domains responsible for toxin activity and neuronal receptor binding. The specific BoNT-A epitopes with strong and medium immunogenic potential, as identified by Atassi et al. and examined in this study, are presented in
Table 7.
To complement this literature-driven selection, a multi-step immunoinformatic workflow was applied to validate and functionally characterize the epitopes across different HLA class II alleles. The identification was not based on arbitrary sequence scanning but rather built upon the empirically validated immunogenic domains reported by Atassi et al., which were used as biologically meaningful input sequences. These sequences were subjected to HLA binding prediction using NetMHCpan 4.1 and IEDB MHC-II binding tools, which evaluate peptides based on known binding motifs, position-specific scoring matrices, and eluted ligand datasets. The software identifies high-affinity binders by computationally estimating the interaction potential between input peptide sequences and selected HLA alleles. The epitope IDs (e.g., L11, C10, N25) used in this manuscript represent internal nomenclature reflecting both domain localization and index position for tracking throughout modeling and docking stages. This selection strategy ensured that epitopes included in this study are not only relevant but also computationally validated for HLA class II presentation.
The epitope sequences were retrieved from NCBI FASTA files (accession: AF488749, AF461540, M30196, X52066, and X73423) and aligned against IEDB’s empirically validated BoNT-A epitopes to confirm conservation and functional relevance. Three-dimensional structural mapping of the epitopes was performed using Chimera and PyMOL [
44], with structural data obtained from Protein Data Bank (PDB ID: 3BTA, 5VGV, and 6F0O) [
45,
46,
47]. SASA calculations [
48] were conducted using the dual signal subspace projection (DSSP) algorithm to determine whether the epitopes were exposed or buried within the toxin structure [
49], as only exposed epitopes are likely to be recognized by the immune system. A threshold of at least 20% surface exposure was set to classify epitopes as antigenically significant [
50].
5.3. HLA Binding Prediction and Molecular Docking
To assess the ability of these epitopes to be presented by antigen-presenting cells (APCs), binding affinity predictions were performed against a panel of HLA class II allele variations. NetMHCpan 4.1 [
51] and IEDB-3D 2.0 [
52] MHC-II binding tools were used for peptide–HLA affinity prediction.
Specific attention was given to the alleles HLA-DQA1
01:02 and
HLA-DQB106:04, as these have been suggested to play a role in BoNT-A immunogenicity. Additionally, other alleles, including HLA-DRB1
15:01,
HLA-DQB103:01, and HLA-DQA1*03:03, were incorporated into the analysis due to their potential involvement in antigen presentation. The classification of epitope–HLA binding affinity followed a stringent threshold, with IC50 values below 500 nM categorized as strong binders, those between 500 and 1000 nM classified as medium binders, and sequences exceeding 1000 nM excluded from further consideration [
53,
54].
To further substantiate the interaction between BoNT-A epitopes and HLA molecule variations, molecular docking simulations were performed using AutoDock Vina 1.2.0, with structural HLA models obtained from the PDB [
55]. Docking was carried out using a flexible docking approach, allowing full conformational flexibility for epitopes while maintaining the HLA receptor in a fixed conformation. Docking scores were analyzed as indicators of binding affinity, where scores below −8.0 kcal/mol were considered indicative of strong interactions, while those between −6.5 and −8.0 kcal/mol were classified as moderate [
56,
57]. Binding interactions were further examined using LigPlot+ to quantify the number of hydrogen bonds, van der Waals interactions, and hydrophobic contacts contributing to epitope stability [
58,
59]. Below are two further examples of molecular docking between N25 and C10 epitopes with HLA-DQA101:02 (
Figure 7).
5.4. Assessment of NAPs and Their Immunogenic Contribution
The potential influence of NAPs on BoNT-A immunogenicity was systematically investigated using an integrative computational approach incorporating molecular docking, MD simulations, solvent-accessibility calculations, and electrostatic potential mapping. The accessory proteins assessed in this study included hemagglutinin (HA) proteins—HA-33, HA-17, and HA-70—as well as non-toxic non-hemagglutinin (NTNH).
To evaluate whether NAPs obscure or enhance the accessibility of BoNT-A antigenic sites, molecular docking simulations were performed using AutoDock Vina and ClusPro [
60]. Structural models for HA-33, HA-17, HA-70, and NTNH were retrieved from NCBI GenBank with additional homology modeling conducted via Swiss-Model where required [
61]. The NAP structures were obtained from the Protein Data Bank (PBD ID: 3V0B,4LO0, 4LO1, 4LO2, 4LO3, 4LO4, 4LO5, 4LO6, 4LO7, 4LO8) [
62,
63]. Each docking simulation was designed to allow flexible conformational adjustments of NAPs while treating BoNT-A as a stable reference structure. The top five lowest-energy docking configurations for each NAP-BoNT-A interaction were selected for further analysis.
To quantify changes in epitope accessibility in the presence of NAPs, SASA calculations were performed using DSSP and PyMOL. The solvent exposure of each BoNT-A epitope was assessed under two conditions: bound to NAPs and unbound. The accessibility of epitopes was categorized as fully exposed (>20% SASA coverage), partially exposed (5–20%), or buried (<5%).
To further analyze the structural influence of NAPs on BoNT-A immunogenicity, 100-nanosecond MD simulations were conducted using GROMACS 2021.4 with the CHARMM36m force field in explicit TIP3P water models [
64,
65,
66]. Each simulation underwent energy minimization, followed by equilibration in NVT [(Canonical Ensemble)—Constant Number, Volume, and Temperature] and NPT [(Isothermal–Isobaric Ensemble)—Constant Number, Pressure, and Temperature] ensembles, ensuring stability before entering the production phase [
67]. Root Mean Square Deviation (RMSD) and Root Mean Square Fluctuation (RMSF) calculations were used to assess conformational changes in BoNT-A epitopes when complexed with NAPs [
68]. Additionally, Molecular Mechanics Poisson–Boltzmann Surface Area (MM-PBSA) calculations were performed to estimate the binding free energy of NAP-BoNT-A interactions and determine whether these interactions impact antigen presentation [
69].
This study also examined whether NAPs interact directly with HLA class II molecules, potentially altering antigen processing and presentation. To assess this, molecular docking simulations were conducted with HLA-DQA1*01:02, HLA-DQB1*06:04, HLA-DRB1*15:01, HLA-DQB1*03:01, and HLA-DQA1*03:03. High-resolution structural models of these alleles were retrieved from the Protein Data Bank when available (PDB: 6DIG, 8TBP, 4D8P) [
70,
71,
72]. Docking experiments allowed for flexible docking of NAPs, while HLA molecules were held rigid to maintain the structural integrity of the peptide-binding groove. NetMHCIIpan 4.1 was used to predict whether BoNT-A epitopes exhibited altered binding affinities to HLA molecules when associated with NAPs. Binding affinity was classified as strong (IC50 < 500 nM), moderate (500–1000 nM), or weak (>1000 nM). MM-PBSA calculations were further performed to determine potential thermodynamic shifts in epitope–HLA interactions due to NAP presence.
To investigate whether NAPs influence HLA recognition through electrostatic modifications, electrostatic potential mapping was conducted using the Adaptive Poisson–Boltzmann Solver (APBS) [
73]. Surface charge distribution changes were analyzed under three conditions: BoNT-A epitopes in isolation, BoNT-A epitopes bound to HLA, and BoNT-A epitopes bound to HLA in the presence of NAPs. Electrostatic surface maps were visualized using ChimeraX and PyMOL to assess whether charge redistribution at the peptide-binding interface could affect antigen recognition.
5.5. Data Entry and Workflow Transparency
All epitope sequences were entered manually one by one into the modeling pipeline to prevent automation errors. Each docking run (epitope–HLA or epitope–NAP) was executed independently and repeated three times to ensure consistency. The data obtained (binding affinities, docking scores, interaction energies, electrostatic maps) were compiled and analyzed manually in structured datasets for further statistical analysis and visualization.
5.6. Statistical and Computational Validation and Visualization
All molecular docking experiments were conducted in triplicates to account for variability in docking scores and binding affinities. The statistical significance of docking results was evaluated using one-way ANOVA, comparing binding affinities of epitopes across different HLA alleles [
74]. To account for multiple hypothesis testing and reduce the risk of Type I error, Bonferroni correction was applied to all multi-allele and multi-epitope comparisons. The significance threshold was adjusted accordingly, with corrected
p-values (α/n) reported for ANOVA and correlation tests.
To validate the stability of the epitope–HLA complexes under physiological conditions, molecular dynamics simulations were conducted using GROMACS over a 100-nanosecond timeframe. Each complex was subjected to energy minimization and equilibrium assessments under CHARMM36m force fields. The stability of interactions was measured through RMSD, RMSF, and MM-PBSA binding free energy calculations. Stability criteria were defined based on an RMSD threshold of less than 3.5 Å, ensuring that only complexes with minimal conformational fluctuations were considered robust antigenic determinants.
Statistical correlation analyses were conducted to compare computational binding predictions with empirically validated epitope–HLA affinities available in the IEDB (Immune Epitope Database). Pearson correlation coefficients were calculated to determine the predictive accuracy of the docking models.
To ensure that the selected epitopes were not strain-specific, multiple sequence alignments were performed using Clustal Omega 1.2.2 to compare BoNT-A sequences across different toxin-producing Clostridium strains. Shannon entropy values were computed to quantify sequence variability, where values below 0.2 indicated highly conserved epitopes and those exceeding 1.5 suggested significant sequence variability.
The results from NetMHCpan 4.1, IEDB MHC-II binding predictions, AutoDock Vina docking simulations, MD stability assessments via GROMACS, and structural analyses in Chimera and PyMOL were systematically processed to produce visualizations using RAWGraphs 2.0 [
75].