HSV2 gB is a 130 kDa envelope glycoprotein that forms trimeric spikes on the viral surface. It mediates initial attachment to heparan sulfate proteoglycans on host cells and facilitates membrane fusion via interactions with the gH/gL heterodimer . Key functional domains include:
Domain | Role | Source |
---|---|---|
Amino-terminal | Initial binding to heparan sulfate moieties | |
Carboxy-terminal | Fusion machinery interaction with gH/gL |
The protein is conserved across HSV-1 and HSV-2, but type-specific epitopes in the amino-terminal region (aa 18–75) enable serological differentiation .
Antibodies targeting HSV2 gB disrupt viral entry and cell-to-cell spread through distinct mechanisms:
Monoclonal antibodies (mAbs) like hu2c and 2c bind discontinuous epitopes in domain I of gB, blocking fusogenic signal transmission .
Valency-dependent efficacy: Bivalent IgG or F(ab′)₂ derivatives neutralize HSV-2 more effectively than monovalent Fab/scFv fragments (Table 1) .
Antibody Form | HSV-1 Neutralization (nM) | HSV-2 Neutralization (nM) |
---|---|---|
IgG | 8 | 32 |
F(ab′)₂ | 4 | 16 |
Fab | 3,000 | 3,000 |
scFv | Ineffective | Ineffective |
Data from neutralization assays on Vero cells .
gB-specific antibodies prevent viral release from infected cells, limiting dissemination .
Complement-independent action: Unlike other anti-HSV mAbs, hu2c neutralizes HSV-2 without requiring Fc-mediated effector functions, making it suitable for immunocompromised patients .
Multidrug-resistant isolates: hu2c neutralizes clinical HSV-2 strains resistant to acyclovir, valacyclovir, and foscarnet with comparable efficacy to drug-sensitive strains .
In vivo protection: Prophylactic or therapeutic administration of hu2c prevents mortality in NOD/SCID mice challenged with lethal HSV-2 doses .
mAb | Target | Application | Status |
---|---|---|---|
HDIT101 | gB | Topical prevention | Phase I/II trials (NCT04539483) |
Humanized 2c | gB | Drug-resistant infections | Preclinical (NOD/SCID models) |
Adapted from clinical trial registries and preclinical studies .
Cross-reactivity with HSV-1 gB:
Antibody Concentration Requirements
Off-Target Effects
Combination Therapies:
Pairing anti-gB mAbs with antivirals or vaccines to enhance efficacy.
Biomarkers for Resistance:
Monitoring gB epitope mutations in clinical isolates to predict therapeutic outcomes.
Global Diagnostic Challenges:
Herpes simplex virus (HSV) enters host cells through a multi-step process involving viral glycoproteins and cellular receptors. The virus's outer envelope contains proteins that bind to specific receptors on the host cell's surface. This binding triggers the fusion of the viral envelope with the cell membrane, creating a pore. Through this pore, the virus enters the host cell. HSV entry mirrors the entry mechanisms of other viruses. Initially, matching receptors on the virus and the cell surface draw the two membranes close. An intermediate stage involves the merging of the two membranes, forming a partially fused state called hemifusion. Finally, a stable entry pore forms, allowing the contents of the viral envelope to enter the host cell.
The antibody is supplied in a solution containing 1 milligram of antibody per milliliter of phosphate-buffered saline (PBS) at a pH of 7.2. The solution also contains 0.01% sodium azide (NaN3) as a preservative.
For short-term storage (2-4 weeks), the antibody should be kept at a temperature of 4 degrees Celsius. For long-term storage, the antibody should be stored frozen at -20 degrees Celsius. To maintain antibody stability, it is crucial to minimize the number of times the antibody is thawed and refrozen.
HSV-2 gB antibody was purified from mouse ascitic fluids by Protein-A chromatography.
Mouse Anti Human Monoclonal.
Monoclonal anti HSV-2 gB IgG1 produced against a HSV-2 infected cells.
Glycoprotein B (gB) is one of the primary targets of the humoral immune response during HSV-2 infection. Research shows that gB, along with glycoproteins gD and gC, is recognized by sera from all HSV-seropositive individuals, although the intensity of response varies between patients . gB is an integral component of the multicomponent fusion system required for virus entry and cell-cell fusion . The antibody response against gB specifically contributes to virus neutralization, although surprisingly, studies have shown that only some individuals develop strong neutralizing antibodies targeting gB epitopes. In a study examining the antibody repertoire of HSV-seropositive individuals, only three out of ten samples contained neutralizing IgGs to gB epitopes, suggesting potential variability in how the immune system targets this protein during natural infection .
Several methodologies have proven effective for detecting and quantifying HSV2 gB-specific antibodies, each with specific applications in research settings:
Western blotting with purified glycoproteins: This approach allows detection of antibodies against denatured epitopes of gB from both HSV-1 and HSV-2. Researchers typically use purified recombinant gB or viral lysates separated by SDS-PAGE .
Biosensor competition assays using surface plasmon resonance: This sophisticated technique determines whether and to what extent human antibodies compete with characterized monoclonal antibodies for binding to specific epitopes on gB. The approach involves capturing soluble gB on a biosensor chip, flowing human IgG samples across the chip surface, and then measuring binding of specific monoclonal antibodies .
Affinity chromatography: For isolation and quantification of gB-specific antibodies, tandem affinity chromatography using immobilized recombinant gB allows researchers to separate gB-specific IgGs from total serum IgGs, enabling determination of their specific contribution to neutralization .
ELISA-based assays: While not explicitly detailed in the provided materials, ELISA represents a standard approach for quantifying antibody titers against specific viral proteins.
The valency of anti-gB antibodies significantly influences their neutralization capacity. Research with the gB-specific monoclonal antibody MAb 2c has demonstrated that neutralization is critically dependent on cross-linkage of gB trimers, which can only be accomplished by bivalent antibody formats .
In detailed studies comparing monovalent formats (Fab and scFv) with bivalent formats (IgG and F(ab')2) of MAb 2c, researchers found that only the bivalent derivatives exhibited high neutralizing activity in vitro . This finding suggests that the mechanism of neutralization involves structural stabilization or interference with conformational changes in gB trimers rather than simple epitope blocking. The requirement for bivalency indicates that effective antibodies may function by cross-linking adjacent gB trimers or by bridging domains within a single trimer, preventing the protein from undergoing conformational changes necessary for the fusion process.
This mechanistic insight has important implications for vaccine design, as it suggests that eliciting antibodies capable of similar cross-linking effects may be crucial for protective immunity.
The relationship between epitope targeting on gB and neutralization effectiveness is complex and depends on several factors including epitope accessibility, conservation, and functional relevance. Analysis of the HSV-1 gB crystal structure in relation to neutralizing antibodies reveals that some key neutralizing epitopes are only partially accessible within the observed multidomain trimer conformation of gB, which likely represents its postfusion conformation .
Mapping studies have shown that certain epitopes are more effective targets for neutralization than others. For example, the discontinuous epitope recognized by MAb 2c is only partially accessible within the observed multidomain trimer conformation of gB . Nevertheless, antibodies targeting this region demonstrate potent neutralizing activity when in bivalent format, suggesting that they may interact with gB at stages during the fusion process when this region becomes more accessible.
Studies using biosensor competition assays with well-characterized neutralizing mouse monoclonal antibodies have also shown that during natural infection, not all neutralizing epitopes on gB induce consistent human antibody responses . This variability in epitope targeting may partially explain differences in neutralizing capacity observed among individuals.
Fusion constructs combining gB with immune modulators represent an innovative approach to enhance vaccine efficacy by directing the immunogen to responsive immune cells and modulating the immune response. A notable example is the HSV-2 gB-CCL19 fusion construct, which demonstrates several advantages over traditional approaches:
Enhanced antibody production: gB-CCL19 fusion constructs induce stronger gB-specific IgG and IgA responses in both sera and vaginal fluids compared to gB alone .
Improved neutralizing activity: The antibodies elicited by these fusion constructs show increased neutralizing activity against HSV-2 in vitro .
Balanced immune response: gB-CCL19 fusion constructs induce balanced Th1 and Th2 cellular immune responses, which may be optimal for controlling HSV-2 infection .
Improved protection: Mice vaccinated with the fusion constructs demonstrated superior protection from intravaginal lethal challenge with HSV-2 .
Enhanced mucosal immunity: Compared to co-administration of gB and CCL19, fusion constructs increased mucosal surface IgA+ cells, as well as CCL19-responsive immunocytes in spleen and mesenteric lymph nodes .
The mechanistic basis for these improvements likely relates to the ability of CCL19 to direct the immunogen to CCL19-responsive cells, improving the efficiency of antigen presentation and subsequent immune response development. This approach may be particularly valuable for developing vaccines against mucosal infections like HSV-2.
The purification and characterization of recombinant HSV2 gB for immunological studies requires careful consideration of protein structure, folding, and epitope preservation. Based on established methodologies, the following approach is recommended:
Expression system selection: Baculovirus-infected insect cells have proven effective for expressing recombinant HSV gB. For example, a truncated form of gB consisting of amino acids 31-724 with a histidine tag (gB(724tHis)) has been successfully expressed and purified from this system .
Protein engineering considerations:
Truncation at residue 724 or 730 removes the transmembrane domain while preserving the extracellular portion containing major antigenic sites
Addition of a histidine tag facilitates purification via metal affinity chromatography
Engineering a GCN4-based isoleucine zipper motif can promote proper oligomerization
Purification protocol:
Metal affinity chromatography using the histidine tag
Size exclusion chromatography to ensure homogeneity and proper oligomerization
Verification of purity by SDS-PAGE and Western blotting
Characterization methods:
Circular dichroism to assess secondary structure
Western blotting with conformation-specific antibodies to confirm proper folding
ELISA with a panel of monoclonal antibodies to verify preservation of key epitopes
Limited proteolysis, such as chymotrypsin digestion, which can provide information about domain structure and accessibility
For detailed epitope mapping studies, chymotrypsin digestion can be particularly informative. Research has shown that chymotrypsin cleaves gB between residues Leu97/Arg98 and Gln472/Ser473, effectively removing residues Ala31 to Leu97 and yielding a stable homogeneous product that can be further analyzed .
Biosensor competition assays using surface plasmon resonance (SPR) represent a powerful approach for mapping HSV2 gB antibody epitopes. Based on published methodologies, the following optimization strategies are recommended:
Immobilization strategy:
Capture recombinant gB on the biosensor chip via its histidine tag using an anti-histidine antibody or NTA surface
This orientation-specific immobilization helps preserve native epitope presentation
Avoid direct covalent coupling which may alter protein conformation or block epitopes
Experimental design:
Use a parallel flow design with test and control surfaces
Capture soluble gB on the test surface
Expose the test surface to human IgG samples to allow binding of gB-specific antibodies
In parallel, expose a control surface containing gB to buffer alone
Flow characterized monoclonal antibodies with known epitopes across both surfaces
Data analysis:
Calculate percent inhibition of monoclonal antibody binding by comparing test surface (pre-exposed to human IgG) with control surface
Reduced MAb binding indicates the presence of human antibodies targeting the same or overlapping epitope
Establish threshold values for positive competition based on signal-to-noise ratios
Validation controls:
Include non-competing antibody pairs to confirm specificity
Use dose-response experiments with purified antibodies to establish competition sensitivity
Include mouse MAbs that target distinct, non-overlapping epitopes as specificity controls
This approach allows researchers to determine whether and to what extent the binding of characterized monoclonal antibodies to gB is diminished by preincubation with human IgG samples, providing a map of the epitopes targeted during natural infection or following vaccination.
Designing rigorous in vivo experiments to evaluate gB-based vaccine candidates requires careful consideration of multiple factors to ensure reliable, translatable results:
Animal model selection:
Immunization protocol:
Route: Subcutaneous immunization has been effective for gB-based vaccines
Schedule: Prime-boost regimens with 2-3 immunizations at 2-3 week intervals
Dose: Titrate vaccine dose to determine minimum effective concentration
Controls: Include appropriate controls (adjuvant alone, irrelevant antigen, positive control)
Challenge model:
Endpoints and evaluation criteria:
Clinical scoring: Monitor animals daily for disease signs (lesions, neurological symptoms)
Viral shedding: Collect vaginal swabs for viral quantification
Latency: Examine dorsal root ganglia for viral DNA to assess protection against latent infection
Immune correlates: Collect serum and mucosal samples for antibody and T-cell response analysis
Mechanistic studies:
Ethical considerations:
Implement humane endpoints based on clinical scoring systems
Ensure proper approval from institutional animal care and use committees
Follow the 3Rs principle (Replacement, Reduction, Refinement)
These considerations will help ensure that experiments generate robust, reproducible data that can guide the development of effective HSV-2 vaccines.
Discrepancies between in vitro neutralization and in vivo protection for anti-gB antibodies represent a complex challenge in HSV-2 research. Several key considerations can help researchers interpret these differences:
Fc-mediated mechanisms: Research shows that non-neutralizing antibodies can provide complete protection through Fc-mediated mechanisms. Studies with HSV-2 vaccines deleted in glycoprotein D demonstrated that protection was lost when immune serum was transferred to Fcγ receptor and FcRn knockout mice, despite minimal neutralizing activity in vitro . This suggests that:
Antibody-dependent cellular cytotoxicity (ADCC)
Complement-dependent cytotoxicity
Fc-mediated phagocytosis
May be critical in vivo mechanisms not captured by standard neutralization assays.
Epitope accessibility differences: Some epitopes may be more accessible in vivo than in the virion preparations used for in vitro assays. For example, the discontinuous epitope recognized by the potent neutralizing MAb 2c is only partially accessible within the observed multidomain trimer conformation of gB, likely representing its postfusion conformation . In vivo, this epitope may become more accessible during the dynamic fusion process.
Mucosal vs. serum antibodies: Standard neutralization assays typically use serum antibodies, but protection at mucosal surfaces may depend on locally produced IgA with different functional properties. Studies with gB-CCL19 fusion constructs showed enhanced mucosal IgA production that correlated with protection .
Antibody valency effects: The neutralization capacity of antibodies like MAb 2c is dependent on cross-linkage of gB trimers, with bivalent formats showing significantly higher neutralizing activity than monovalent formats . Standard neutralization assays may not fully capture these valency-dependent effects that are relevant in vivo.
To address these discrepancies, researchers should:
Incorporate multiple assays beyond standard neutralization, including ADCC and complement fixation
Evaluate protection in relevant animal models with intact immune systems
Consider passive transfer experiments to determine the protective capacity of antibodies in vivo
Examine mucosal antibody responses separately from systemic responses
Determining the relative contribution of anti-gB antibodies versus other glycoprotein-specific antibodies in polyclonal serum requires sophisticated methods to isolate and functionally characterize specific antibody populations. Based on published methodologies, the following approaches are recommended:
Tandem affinity chromatography separation:
Sequential affinity chromatography using immobilized recombinant glycoproteins (e.g., gD followed by gB) can be used to fractionate serum IgGs into glycoprotein-specific populations
This approach has been successfully used to isolate gD-specific and gB-specific IgGs from human sera
The isolated antibody fractions can then be tested individually for neutralizing activity
Quantitative depletion studies:
Deplete specific antibody populations using immobilized recombinant glycoproteins
Compare neutralizing activity of depleted versus non-depleted serum
The reduction in neutralizing activity represents the contribution of the depleted antibody specificity
Competitive inhibition assays:
Pre-incubate virus with excess recombinant glycoproteins to block binding of specific antibody populations
Compare neutralizing activity with and without competitive inhibition
The reduction in neutralizing activity indicates the contribution of antibodies targeting the competing glycoprotein
Western blot analysis with glycoprotein-specific fractions:
Biosensor epitope mapping:
Use surface plasmon resonance to map which epitopes on each glycoprotein are targeted by the serum
This provides information on both the breadth and focus of the antibody response
A study examining the IgGs from HSV-seropositive individuals demonstrated the application of these approaches by showing that in some individuals, gD and gB together accounted for all neutralizing activity against HSV-2, with some HSV-1 neutralization directed against gC . This type of comprehensive analysis provides important insights into the protective antibody response against HSV glycoproteins.
The differences in gB sequence and structure between HSV-1 and HSV-2 have significant implications for cross-reactive antibody responses, which is critical for understanding cross-protection and developing broadly protective vaccines. Although the search results don't provide comprehensive details on this specific topic, we can infer several important considerations based on the available information:
Test neutralization against both HSV-1 and HSV-2
Use recombinant glycoproteins from both virus types in binding and competition assays
Consider structural modeling to identify conserved and variable epitopes
Examine whether antibodies that bind both HSV-1 and HSV-2 gB have similar functional activities against both viruses
The deletion or modification of other viral glycoproteins can significantly alter the immune response to gB, potentially unmasking protective epitopes or changing the immunodominance hierarchy. The most striking example comes from studies of HSV-2 virus deleted in glycoprotein D (ΔgD−/+gD1):
Unmasking of protective antigens: Deletion of gD, which is immunodominant in wild-type HSV infection, appears to unmask viral antigens important in generating a protective humoral response . Western blot analysis showed that the predominant antigen recognized by control-immunized, HSV-2-challenged mice was gD, while ΔgD−/+gD1-vaccinated mice developed stronger responses to other antigens, including gB .
Shift in antibody functionality: The antibodies elicited by ΔgD−/+gD1 vaccination showed little neutralizing activity but demonstrated strong antibody-dependent cellular cytotoxicity (ADCC) . This suggests that modification of the viral glycoprotein composition can fundamentally alter the functional profile of the antibody response.
Change in protective mechanisms: Protection conferred by ΔgD−/+gD1 vaccination was dependent on Fc-mediated antibody functions rather than direct neutralization. This protection was lost when immune serum was transferred to FcγR and FcRn knock-out mice, highlighting the critical role of Fc receptor interactions .
Potential immunomodulatory effects: The deletion of gD may alter immune responses through mechanisms beyond simple antigen unmasking. gD interacts with herpesvirus entry mediator (HVEM) on immune cells, and this interaction may skew the immune response . In the absence of the gD-HVEM interaction, the host may mount a polyantigenic IgG2-dominant response capable of eliciting FcR-mediated protection.
These findings suggest that strategic modification of viral glycoproteins could be a promising approach for developing more effective HSV vaccines. By altering the composition or expression levels of specific glycoproteins, it may be possible to direct the immune response toward more protective targets or functional mechanisms.
Non-neutralizing anti-gB antibodies appear to play a significant role in protection against HSV-2 infection through Fc-mediated effector functions. This represents a paradigm shift from the traditional focus on neutralizing antibodies as the primary correlate of protection. Evidence for the protective role of non-neutralizing antibodies includes:
Fc receptor dependency: Studies with the HSV-2 ΔgD−/+gD1 vaccine demonstrated that protection was mediated by antibodies with little neutralizing activity but was completely dependent on Fc receptors. Protection was lost when immune serum was transferred to FcγR knock-out mice, despite the presence of HSV-specific antibodies in vaginal washes .
Antibody-dependent cellular cytotoxicity (ADCC): Serum from mice immunized with ΔgD−/+gD1 elicited cell-mediated cytotoxicity, suggesting that ADCC is an important protective mechanism . This function can be mediated by non-neutralizing antibodies that recognize viral antigens on the surface of infected cells.
FcRn-mediated transport: The neonatal Fc receptor (FcRn) plays a crucial role in transporting antibodies from serum to mucosal surfaces. Protection was lost when immune serum was transferred to FcRn knock-out mice, and HSV-specific antibodies were not detected in vaginal washes of these mice . This highlights the importance of mucosal antibody delivery for protection.
Complement activation: While not explicitly discussed in the provided materials, complement activation represents another mechanism by which non-neutralizing antibodies can contribute to protection.
These findings suggest that vaccine strategies targeting gB should aim to elicit not only neutralizing antibodies but also antibodies capable of mediating efficient Fc-dependent effector functions. This might be achieved through appropriate adjuvant selection, antigen formulation, or vaccine delivery strategies that promote particular antibody isotypes or subclasses associated with enhanced effector functions.
The protective role of non-neutralizing antibodies also has implications for evaluating vaccine candidates, suggesting that standard neutralization assays alone may not predict protective efficacy and should be complemented with assays measuring Fc-mediated functions.
Developing standardized assays for measuring anti-gB antibody functionality presents several technical challenges that researchers must address to ensure reliable and comparable results across different laboratories:
Neutralization assay variability:
Cell type differences: Neutralization efficiency can vary depending on the cell line used
Virus strain differences: HSV-1 and HSV-2 strains may show variable susceptibility to neutralization
Assay format: Plaque reduction versus colorimetric/fluorometric readouts may yield different results
Incubation conditions: Temperature, time, and medium composition can affect outcomes
Beyond neutralization functionality:
ADCC assays require appropriate effector cells and target cells expressing physiologically relevant levels of viral antigens
Complement-dependent cytotoxicity assays need standardized complement sources
Fc-mediated phagocytosis assays require consistent phagocytic cell preparation
Epitope accessibility issues:
Key neutralizing epitopes on gB may be only partially accessible in the postfusion conformation observed in the crystal structure
Different assay conditions may affect protein conformation and epitope exposure
Antibodies may recognize epitopes that are only transiently exposed during the fusion process
Recombinant protein quality:
Variation in recombinant gB preparation (truncation sites, tags, expression systems) affects epitope presentation
Protein aggregation or misfolding can expose non-native epitopes
Post-translational modifications may differ between expression systems
Recommended solutions:
Standardized reagents:
Develop and distribute reference antibodies with defined specificities and activities
Establish common recombinant protein preparations with defined characteristics
Create standardized virus stocks for neutralization assays
Comprehensive assay panels:
Implement multiple complementary assays measuring different aspects of antibody functionality
Include both neutralization and Fc-mediated function assays
Validate assays with antibodies of known functionality
Detailed protocols:
Publish comprehensive methodologies including all critical parameters
Conduct inter-laboratory comparison studies to identify and address variables affecting reproducibility
Establish accepted positive and negative controls for each assay
Data normalization approaches:
Develop statistical methods to compare results across different assay formats
Establish reporting standards that include all relevant experimental parameters
Create conversion factors to allow comparison between different assay readouts
By addressing these challenges, researchers can develop more reliable and comparable methods for measuring the diverse functional activities of anti-gB antibodies, facilitating more effective vaccine development and evaluation.
Distinguishing between type-common and type-specific antibody responses to HSV gB requires systematic approaches that can identify antibodies recognizing conserved versus variable epitopes. The following methodological approaches are recommended:
Comparative binding assays:
ELISA using purified recombinant gB from both HSV-1 and HSV-2
Compare binding curves and calculate relative affinities
Antibodies with similar binding to both proteins likely target conserved epitopes
Western blotting against purified glycoproteins from both serotypes can reveal type-specific recognition patterns
Cross-neutralization testing:
Test antibody samples against both HSV-1 and HSV-2 in neutralization assays
Calculate neutralization titers against each virus type
Compare neutralization efficiency to identify type-biased versus broadly neutralizing antibodies
Studies have shown that some human sera contain antibodies that neutralize both HSV-1 and HSV-2, while others show type-preference
Epitope mapping strategies:
Biosensor competition assays using well-characterized type-common and type-specific monoclonal antibodies
Peptide mapping with overlapping peptides covering variable regions of gB
Domain-swapping experiments with chimeric gB proteins containing domains from HSV-1 and HSV-2
Chymotrypsin digestion of gB followed by epitope mapping can provide information about domain-specific recognition
Absorption/depletion studies:
Sequential absorption with recombinant gB from one virus type followed by testing reactivity against the other type
Depletion with type-specific peptides to remove antibodies targeting variable regions
Analysis of the remaining antibody fraction for cross-reactivity
Structural analysis:
Computational mapping of conserved and variable regions on the gB structure
Correlation of antibody binding sites with these regions
Prediction of accessible epitopes that might elicit type-common versus type-specific responses
These approaches can be combined to provide a comprehensive analysis of the type-specificity of anti-gB antibody responses, which is crucial for understanding cross-protection and developing broadly protective vaccines. The data obtained can be presented in tables comparing binding and neutralization against both virus types, and mapping results can be visualized on structural models highlighting conserved and variable epitopes.
The development of gB-based universal vaccines against multiple herpesvirus infections represents an ambitious but potentially achievable goal, given certain characteristics of this glycoprotein across the herpesvirus family:
Structural and functional conservation:
gB is one of the most conserved glycoproteins across the herpesvirus family
It plays a critical role in the fusion mechanism required for virus entry across all herpesviruses
The core fusion machinery likely maintains similar structural features despite sequence divergence
Evidence for cross-reactive immunity:
Some studies have demonstrated cross-reactivity between antibodies targeting gB from different herpesviruses
The success of HSV-2 gB-based vaccines in providing protection against both HSV-1 and HSV-2 challenges suggests potential for broader protection
The identification of conserved neutralizing epitopes could form the basis for universal vaccine design
Strategies for enhancing cross-protection:
Structure-based design of immunogens focusing on conserved regions of gB
Multivalent constructs incorporating gB from multiple herpesviruses
Fusion with immune modulators like CCL19 to enhance both local and systemic immunity
Deletion of immunodominant variable regions to focus the immune response on conserved epitopes
Challenges to overcome:
Sequence divergence between gB from distant herpesvirus family members
Different tissue tropism and transmission routes of various herpesviruses
Potential for type-specific neutralizing epitopes dominating the immune response
Need for different immune effector mechanisms for controlling different herpesviruses
Innovative approaches:
Computational design of "consensus" gB sequences representing conserved features
Structure-based engineering of stabilized prefusion conformations to expose conserved neutralizing epitopes
Nanoparticle presentation of multiple conserved epitopes from different herpesvirus gB proteins
Combination with other conserved herpesvirus antigens to broaden protection
The successful development of gB-CCL19 fusion constructs that provide protection against HSV-2 challenge and the demonstration that antibody-mediated protection can occur through non-neutralizing Fc-mediated mechanisms provide promising platforms upon which to build universal herpesvirus vaccine strategies. Future research should focus on identifying the most conserved functional epitopes across herpesvirus gB proteins and developing immunization strategies that elicit antibodies targeting these regions.
Emerging technologies in antibody engineering and analysis have the potential to significantly advance HSV2 gB research, offering new tools for understanding antibody responses and developing therapeutic approaches:
Single B cell analysis and antibody repertoire sequencing:
Next-generation sequencing of B cell receptors from HSV-infected or vaccinated individuals
Identification of expanded B cell clones targeting specific gB epitopes
Tracking of antibody maturation pathways to understand how protective responses develop
Comparison of repertoires between individuals with different levels of protection
High-throughput antibody isolation and characterization:
Single-cell sorting of gB-specific B cells followed by antibody gene cloning
Microfluidic platforms for rapid screening of antibody binding and neutralization
Deep mutational scanning to map critical residues for antibody recognition of gB
Creation of comprehensive panels of monoclonal antibodies targeting different gB epitopes
Advanced structural techniques:
Cryo-electron microscopy to visualize gB-antibody complexes in different conformational states
X-ray crystallography of gB bound to novel neutralizing antibodies
Hydrogen-deuterium exchange mass spectrometry to map conformational changes induced by antibody binding
Computational modeling of antibody-antigen interactions to guide vaccine design
Antibody engineering applications:
Creation of bispecific antibodies targeting multiple epitopes on gB or multiple glycoproteins
Fc engineering to enhance ADCC, complement activation, or tissue penetration
Development of antibody-drug conjugates for targeted delivery to HSV-infected cells
Engineering of antibodies with extended half-lives for prophylactic applications
In vivo antibody imaging:
Labeled antibodies for tracking biodistribution and tissue penetration
Intravital microscopy to visualize antibody interactions with infected cells
PET imaging with radiolabeled antibodies to monitor infection sites and antibody targeting
Novel therapeutic modalities:
Antibody-based CAR-T cells targeting gB-expressing infected cells
mRNA-encoded antibodies for direct in vivo expression
Nanobodies and alternative scaffold proteins with enhanced tissue penetration These technologies will facilitate a more comprehensive understanding of protective anti-gB antibody responses and enable the development of novel therapeutic and prophylactic approaches. The ability to rapidly isolate and characterize potent neutralizing antibodies from infected individuals could lead to new passive immunotherapy options, while structural insights gained from antibody-antigen complexes will inform rational vaccine design strategies.
Herpes Simplex Virus Type 2 (HSV-2) is a member of the Herpesviridae family, which is known for causing genital herpes in humans. One of the key components of HSV-2 is Glycoprotein B (gB), a crucial protein involved in the virus’s ability to enter host cells and facilitate cell-to-cell transmission. This article delves into the background of HSV-2 gB and the development of mouse anti-human antibodies targeting this glycoprotein.
Glycoprotein B (gB) is an envelope glycoprotein that plays a pivotal role in the viral life cycle of HSV-2. It is involved in the initial attachment of the virus to host cells, fusion of the viral envelope with the host cell membrane, and subsequent entry into the host cell. gB is also essential for the spread of the virus between cells, making it a critical target for therapeutic interventions .
Mouse anti-human antibodies are monoclonal antibodies developed in mice that specifically target human proteins. These antibodies are produced using hybridoma technology, where mice are immunized with the target antigen (in this case, HSV-2 gB), and antibody-producing B cells are fused with myeloma cells to create hybrid cells capable of producing large quantities of the desired antibody .
The development of mouse anti-human antibodies targeting HSV-2 gB involves several steps:
These antibodies have several applications in research and clinical settings: