TNFRSF18 antibodies are immunological reagents designed to detect or modulate GITR, a cell surface receptor encoded by the TNFRSF18 gene (UniProt: Q9Y5U5). This receptor is constitutively expressed on regulatory T cells (Tregs) and upregulated on activated effector T cells, natural killer (NK) cells, and myeloid-derived suppressor cells (MDSCs) . Antibodies against TNFRSF18 enable researchers to:
Study GITR's role in immune checkpoint pathways
Modulate Treg suppression and effector T-cell activity
Develop therapeutic strategies for cancer and autoimmune conditions .
Treg Modulation: GITR activation abrogates Treg-mediated suppression, enhancing effector T-cell responses .
Co-stimulation: TNFRSF18 engagement on CD4+/CD8+ T cells amplifies IL-2, IL-4, and IFN-γ production .
Disease Models:
GITR associates with TRAF2/5 and NF-κB, promoting pro-survival signals in effector T cells while inhibiting Treg suppression .
| Antibody | Type | Clinical Phase | Key Feature |
|---|---|---|---|
| TRX518 | Agonistic | I/II | Blocks GITR/GITRL interaction |
| BMS-986156 | Agonistic | I | IgG1 with ADCC activity |
| MK-4166 | Agonistic | II | Combined with PD-1 inhibitors |
TNFRSF18, commonly known as GITR or CD357, is a member of the TNF receptor superfamily. It functions as a co-stimulatory receptor primarily expressed on various immune cells, particularly T cells. The receptor plays crucial roles in modulating immune responses by influencing T cell activation, proliferation, and survival.
GITR is part of the broader tumor necrosis factor receptor superfamily that regulates antigen presentation and adaptive immune activities. Unlike inhibitory immune checkpoints, GITR acts as a co-stimulatory molecule that can enhance anti-tumor immune responses when properly activated . The receptor's activation typically requires molecular clustering induced by its cognate ligand (GITR-L) or through antibody-mediated cross-linking, which triggers downstream signaling cascades essential for immune cell function .
TNFRSF18 antibodies are available in multiple configurations to support diverse research applications:
By host species:
By reactivity:
Cross-reactive antibodies with multiple species compatibility
By clonality:
By conjugation:
The selection of antibody epitope is critical and should be guided by the specific research question:
For structural studies:
Choose antibodies targeting specific domains:
Extracellular domain antibodies (e.g., AA 26-162) for surface expression studies
Antibodies targeting the ligand-binding region (e.g., AA 90-162) for interaction studies
C-terminal domain antibodies (e.g., AA 184-241) for signaling research
For functional studies:
For agonistic activities, select antibodies that bind regions conducive to receptor clustering
For neutralizing studies, choose antibodies that disrupt ligand-receptor interactions
For tracking receptor internalization, select antibodies to extracellular domains that remain bound during trafficking events
The epitope selection directly impacts the antibody's utility in different applications. For instance, antibodies recognizing the AA 184-241 region might be optimal for flow cytometry studies of surface expression, while those targeting functional domains may be better suited for activation or inhibition studies in functional assays .
TNFRSF18 agonistic antibodies induce receptor activation through a complex mechanism involving:
Receptor clustering mechanisms:
TNFRSF receptors are normally activated by molecular clustering induced by their cognate cell membrane ligands. Antibodies can mimic this activation through:
Direct bivalent binding: The inherent bivalency of antibodies allows them to bind two GITR molecules simultaneously, initiating receptor clustering
Fc-mediated cross-linking: The most dominant factor in determining agonistic activities involves Fc interactions with Fc gamma receptors (FcγRs), particularly FcγRIIB
Epitope-dependent effects: The specific binding epitope significantly determines the intrinsic agonistic activity of the antibody, with certain epitopes enabling more efficient clustering
The interaction between the antibody's Fc portion and FcγRIIB provides opportunities for multivalent interactions beyond the primary target. This cross-linking dramatically enhances receptor clustering and downstream signaling activation. Research suggests that Fc engineering approaches to optimize Fc binding to FcγRIIB, in combination with appropriate epitope selection, can significantly improve the agonistic activity of TNFRSF18 antibodies .
When using TNFRSF18 antibodies in cancer immunotherapy research, investigators should consider:
Balancing efficacy and toxicity:
A major challenge in clinical development of TNFRSF agonistic antibodies has been balancing their anti-tumor efficacy with potential toxicities. This requires careful experimental design and monitoring .
Target-specific optimization strategies:
Leveraging interactions between antibodies and inhibitory Fc receptor FcγRIIB to optimize co-stimulation activities selectively in the tumor microenvironment
Exploring cross-linking through tumor antigen binding with bispecific antibody approaches
Fc engineering considerations:
The role of FcγRIIB in TNFRSF cross-linking and clustering bridged by agonistic antibodies is critical
Multiple Fc engineering approaches can optimize FcγRIIB binding in the context of proper Fab and epitope selection
Cross-linking antibody (xLinkAb) models may enhance efficacy while minimizing systemic toxicity
Experimental model selection:
Researchers should select models that appropriately recapitulate the human immune environment and tumor microenvironment to accurately assess antibody functionality and safety profiles.
When encountering inconsistent results with TNFRSF18 antibody-based assays, researchers should systematically evaluate:
Sample preparation factors:
Ensure proper sample fixation for immunohistochemistry applications
Verify cell preparation protocols for flow cytometry to maintain epitope integrity
For protein analysis, confirm appropriate lysis buffers that preserve the target epitope
Technical optimization:
Titrate antibody concentrations to determine optimal signal-to-noise ratios
Adjust incubation conditions (time, temperature, buffer composition)
For flow cytometry, ensure appropriate compensation and gating strategies
For ELISA, optimize blocking and washing steps to reduce background
Antibody validation:
Confirm antibody specificity using positive and negative controls
Consider using multiple antibodies targeting different epitopes to verify results
Validate results using complementary techniques (e.g., flow cytometry results with Western blot)
Biological variables:
Account for differential expression of TNFRSF18 across cell activation states
Consider potential receptor internalization or shedding affecting detection
Evaluate potential cross-reactivity with similar family members
For optimal detection of TNFRSF18 by flow cytometry:
Sample preparation:
Harvest cells gently to minimize receptor shedding
Wash cells in cold PBS + 1% BSA or FBS
If using fresh blood samples, perform red blood cell lysis
Adjust cell concentration to 1-5 × 10⁶ cells/mL
Staining protocol:
Block Fc receptors with 10% normal serum from the antibody host species
Incubate with primary TNFRSF18 antibody at optimal dilution (determined experimentally)
For unconjugated antibodies, wash and add appropriate secondary antibody
Include viability dye to exclude dead cells
Include appropriate isotype controls and FMO (fluorescence minus one) controls
Analysis considerations:
Set appropriate compensation using single-stained controls
Use isotype controls to determine positive staining thresholds
Consider including known positive control samples (such as activated T cells)
For activated cells, compare TNFRSF18 expression levels between resting and stimulated populations
TNFRSF18 expression can be significantly affected by cell activation status, so careful experimental design is crucial for meaningful interpretation .
When designing functional assays to evaluate TNFRSF18 antibody agonistic activity:
T cell activation assay design:
Isolate primary T cells or use appropriate T cell lines
Pre-coat plates with suboptimal anti-CD3 (for TCR stimulation)
Add the TNFRSF18 agonistic antibody at varying concentrations
Include appropriate controls:
Isotype control antibody
Known agonist positive control
Anti-CD28 as alternative co-stimulatory control
Measure activation markers (CD25, CD69), proliferation (CFSE dilution), and cytokine production (IL-2, IFN-γ)
Considerations for Fc-dependent activity assessment:
Test the antibody in presence/absence of FcγRIIB-expressing cells
Include Fc-mutant versions of the antibody to determine Fc dependency
Compare activities across different antibody isotypes with varying FcγR binding profiles
Evaluation in complex immune cell mixtures:
Test antibody in PBMC cultures to assess effects on multiple cell types
Measure changes in Treg suppressive function
Assess dendritic cell maturation and function as secondary readouts
In vivo functional assessment:
Design dose-response studies in appropriate animal models
Monitor immune cell populations in peripheral blood, lymphoid organs, and tumor (if applicable)
Assess changes in immune cell activation status and functional parameters
To differentiate between activities of different TNFRSF18 antibody clones:
Epitope mapping approaches:
Conduct competitive binding assays to determine if antibodies recognize overlapping or distinct epitopes
Perform domain-specific truncation analyses to identify binding regions
Use peptide arrays to identify specific binding sequences
Functional profiling:
Compare dose-response curves for T cell activation
Assess differences in receptor internalization kinetics
Evaluate differential effects on distinct T cell subsets (conventional T cells vs. Tregs)
Measure qualitative differences in downstream signaling activation
Cross-linking dependency analysis:
Test antibodies in the presence/absence of cross-linking agents or FcγR-expressing cells
Compare activities when presented in solution versus immobilized on surfaces
Assess the impact of various Fc engineering modifications on each clone's activity
Data analysis and visualization:
Create comparison tables showing:
| Clone ID | Epitope Region | Intrinsic Agonistic Activity | FcγRIIB Dependency | T Cell Activation (EC50) | Treg Modulation |
|---|---|---|---|---|---|
| 6H8B9 | AA 184-241 | Low/Medium/High | Strong/Moderate/Weak | xx nM | Effect size |
| 4H2D6 | AA 26-162 | Low/Medium/High | Strong/Moderate/Weak | xx nM | Effect size |
| 2H4 | AA 26-115 | Low/Medium/High | Strong/Moderate/Weak | xx nM | Effect size |
Such comparative analyses provide crucial insights into the mechanistic differences between antibody clones and guide selection for specific research applications .
The isotype of TNFRSF18 antibodies significantly impacts experimental outcomes through:
FcγR engagement profiles:
Different isotypes (IgG1, IgG2a/b, IgG4) exhibit distinct binding affinities for activating and inhibitory FcγRs, directly affecting:
Cross-linking efficiency and consequent receptor clustering
Potential for antibody-dependent cellular cytotoxicity (ADCC)
Complement activation
In vivo half-life and tissue distribution
For TNFRSF18 agonistic antibodies, the interaction with inhibitory FcγRIIB is particularly important for optimal cross-linking and agonistic activity . Mouse IgG1 antibodies typically have lower affinity for activating FcγRs compared to mouse IgG2a, resulting in different functional profiles in vivo.
Species-specific considerations:
Human IgG1 and mouse IgG2a have strong affinity for activating FcγRs
Human IgG4 and mouse IgG1 generally have reduced FcγR binding
Human IgG2 shows selective engagement patterns
Researchers should select antibody isotypes based on:
The desired mechanism of action (pure agonism vs. additional effector functions)
The experimental model (human vs. mouse systems)
The availability of FcγR-expressing cells in the experimental system
Proper validation of TNFRSF18 antibodies requires comprehensive controls:
Specificity controls:
TNFRSF18 knockout or knockdown cells/tissues (negative control)
TNFRSF18 overexpression systems (positive control)
Pre-absorption with recombinant TNFRSF18 protein
Comparison with established reference antibodies
Testing on cells known to express or not express TNFRSF18
Technical controls:
Isotype-matched control antibodies to assess non-specific binding
Secondary antibody-only controls (for unconjugated primaries)
Titration series to determine optimal working concentration
Multiple application testing (if claiming multi-application utility)
Biological controls:
Resting vs. activated T cells (TNFRSF18 is upregulated upon activation)
Different T cell subsets (conventional T cells vs. Tregs)
Tissues with known expression patterns
Cross-species reactivity testing if claimed
Functional validation:
Correlation of binding with functional outcomes
Comparison of agonistic/antagonistic effects with known standards
Validation in physiologically relevant models
Documentation of these validation steps provides critical evidence of antibody performance and reliability for specific research applications .
Engineering TNFRSF18 antibodies with enhanced agonistic function involves:
Fc engineering approaches:
Enhancing FcγRIIB binding:
Introducing specific amino acid substitutions in the Fc region
Creating Fc variants with selective FcγRIIB engagement
Optimizing the antibody's Fc glycosylation pattern
Cross-linking antibody (xLinkAb) models:
Structural optimization strategies:
Epitope selection targeting receptor domains that facilitate clustering
Optimizing antibody flexibility through hinge region modifications
Developing antibody fragments with enhanced penetration into tissues while maintaining clustering capability
Comparative efficacy assessment:
Engineered antibodies should be assessed against conventional formats to quantify improvements in:
Receptor clustering efficiency
Downstream signaling activation
T cell proliferation and cytokine production
Anti-tumor activity in relevant models
To effectively monitor TNFRSF18 receptor dynamics following antibody engagement:
Real-time imaging approaches:
Live-cell confocal microscopy with fluorescently labeled antibodies
FRET-based assays to monitor receptor-receptor interactions
Single-molecule tracking to observe individual receptor movements
Total internal reflection fluorescence (TIRF) microscopy for surface clustering visualization
Biochemical and molecular techniques:
Proximity ligation assays to detect closely associated receptor molecules
Immunoprecipitation followed by native PAGE to preserve receptor complexes
Cross-linking studies to capture transient interaction partners
Pulse-chase experiments to track receptor internalization and trafficking
Signaling activation indicators:
Phospho-specific Western blotting for downstream signaling components
Reporter cell lines expressing TNFRSF18-responsive promoter elements
Calcium flux assays for immediate signaling readouts
Transcriptional profiling to identify gene expression changes
Experimental design considerations:
Include appropriate time courses (seconds to hours)
Compare different antibody clones and formats
Assess effects of Fc receptor availability
Evaluate impact of simultaneous TCR engagement
Emerging technologies transforming TNFRSF18 antibody research include:
Advanced antibody engineering platforms:
Structure-guided antibody design based on TNFRSF18 crystal structures
High-throughput screening systems for identifying optimal agonistic clones
Site-specific conjugation technologies for precise payload delivery
Computational approaches to predict and optimize antibody-receptor interactions
Novel functional assessment tools:
Organoid and 3D culture systems for more physiologically relevant testing
Microfluidic systems for analyzing cell-cell interactions in controlled environments
Advanced intravital imaging for real-time visualization of antibody activities in vivo
Single-cell approaches to characterize heterogeneous responses across immune populations
Combination therapy approaches:
Rational design of TNFRSF18 antibodies with complementary checkpoint inhibitors
Development of multi-specific antibodies targeting TNFRSF18 and other immunomodulatory receptors
Integration with cellular therapies like CAR-T cells
Combination with cancer vaccines to enhance tumor-specific responses
The future of TNFRSF18 antibody research lies in developing increasingly sophisticated reagents with enhanced specificity, potency, and safety profiles through these emerging technologies and approaches.
ELISA Protocol for TNFRSF18 Quantification:
Materials:
Anti-TNFRSF18 capture antibody (e.g., clones targeting AA 26-115)
Biotinylated or HRP-conjugated detection antibody (ideally targeting a different epitope)
Recombinant TNFRSF18 protein standards
High-binding 96-well plates
Blocking buffer (1% BSA in PBS)
Wash buffer (0.05% Tween-20 in PBS)
TMB substrate and stop solution
Plate reader capable of measuring absorbance at 450nm
Procedure:
Plate coating:
Dilute capture antibody to optimal concentration (typically 1-5 μg/mL) in coating buffer
Add 100 μL per well and incubate overnight at 4°C
Wash 3 times with wash buffer
Blocking:
Add 300 μL blocking buffer per well
Incubate for 1-2 hours at room temperature
Wash 3 times with wash buffer
Sample addition:
Prepare standards using recombinant TNFRSF18 (typical range: 0-1000 pg/mL)
Dilute samples appropriately in blocking buffer
Add 100 μL of standards and samples in duplicate
Incubate for 2 hours at room temperature
Wash 5 times with wash buffer
Detection:
Add 100 μL of diluted detection antibody
Incubate for 1-2 hours at room temperature
Wash 5 times with wash buffer
If using biotinylated detection, add streptavidin-HRP and incubate for 30 minutes
Wash 5 times with wash buffer
Development and analysis:
This protocol can be optimized based on specific antibody pairs and sample types being analyzed.
When conducting multiplexed analysis with TNFRSF18 antibodies:
Panel design considerations:
Select TNFRSF18 antibodies validated for multiplexed applications
Choose compatible fluorophores or tags based on:
Available instrument configuration
Spectral overlap minimization
Relative abundance of targets (brightest fluorophores for least abundant markers)
Include markers to identify relevant cell populations (e.g., CD3, CD4, CD8 for T cells)
Add functional markers to correlate with TNFRSF18 expression (e.g., activation markers, cytokines)
Technical optimization steps:
Conduct antibody titrations in single-stain format before multiplexing
Test for potential antibody interactions or blocking effects
Optimize staining buffers and conditions for all markers
Perform compensation controls for each fluorophore
Include FMO controls to set accurate gates
Consider live/dead discrimination and doublet exclusion
Analysis approaches:
Conventional manual gating strategies
Unsupervised clustering algorithms (e.g., t-SNE, UMAP)
Supervised machine learning for population identification
Correlation analyses between TNFRSF18 and other markers
Application-specific considerations:
| Application | Key Considerations |
|---|---|
| Flow Cytometry | Compensation, spectral overlap, optimal antibody concentration |
| Mass Cytometry | Metal isotope selection, signal interference, antibody conjugation quality |
| Multiplex IHC/IF | Antibody order, antigen retrieval compatibility, panel design |
| Single-cell sequencing with protein | Antibody-oligo conjugate quality, background control |