Binding induces NF-κB activation via TRAF1/2 recruitment, promoting T-cell survival and proliferation .
Reverse signaling through TNFSF9 on APCs enhances antigen presentation and dendritic cell survival .
TNFRSF9 agonists are investigated for enhancing anti-tumor immunity:
CAR-T Therapy: TNFRSF9 co-stimulation improves T-cell persistence and tumor targeting .
Bispecific Antibodies: Combining TNFRSF9 agonism with PD-L1 blockade synergizes anti-tumor responses (Nature Communications, 2023) .
Immune Checkpoint Modulation: Agonists counteract T-cell exhaustion in solid tumors .
Bidirectional signaling complicates therapeutic targeting:
Protective Roles: Reduces severity in models of rheumatoid arthritis and herpetic stromal keratitis .
Pathogenic Roles: Exacerbates systemic lupus erythematosus and lacrimal gland disease .
Endotoxin Control: Essential for in vivo studies to prevent confounding inflammatory responses .
Batch Variability: EC values may differ between suppliers due to expression systems or tags .
Bidirectional Signaling: TNFRSF9 activation on T cells vs. TNFSF9 reverse signaling on APCs requires context-specific targeting .
Toxicity Concerns: Systemic agonism may induce hepatotoxicity or metabolic inflammation .
Targeted Delivery: Tumor microenvironment-specific agonists to minimize off-tumor effects .
Biomarker-Guided Trials: Stratifying patients based on TNFRSF9 expression or ligand levels .
TNFRSF9, also known as 4-1BB or CD137, is an inducible co-stimulatory receptor primarily expressed on activated T cells. The mature human TNFRSF9 consists of a 163 amino acid extracellular domain (ECD) with four TNFR cysteine-rich repeats, a 27 amino acid transmembrane segment, and a 42 amino acid cytoplasmic domain . Recombinant forms typically include amino acids Leu24-His183/Gln186 of the native protein, sometimes with additional tags (such as 6xHis or 10xHis) for purification and detection purposes . The protein exists as a disulfide-linked homodimer on cell surfaces with an apparent molecular weight of approximately 28-30 kDa, although recombinant monomeric forms may show lower molecular weights (approximately 21.8 kDa) when analyzed by techniques like SEC-MALS .
TNFRSF9 shows a complex expression pattern that varies by cellular context:
Primary expression occurs on various populations of activated T cells, including CD4+, CD8+, memory CD8+, NKT cells, and regulatory T cells
Expression is also found on myeloid and mast cell progenitors, dendritic cells, and bacterially infected osteoblasts
In human gliomas, a surprising expression pattern has been identified primarily in non-neoplastic reactive astrocytes rather than in classic immunological cell types such as lymphocytes and microglia
In glioma tissue, TNFRSF9 shows predominantly perivascular and peritumoural distribution, with significantly higher expression observed in IDH-1 mutant gliomas
The inducible nature of TNFRSF9 means its expression is highly dependent on cellular activation state rather than being constitutively present.
TNFRSF9 binds with high affinity to its ligand TNFSF9 (4-1BB Ligand), which is primarily expressed on antigen-presenting cells and myeloid progenitor cells . This interaction:
Costimulates the proliferation, activation, and survival of TNFRSF9-expressing cells
Plays critical roles in the activation, differentiation, and apoptosis of T cells
Mediates anti-tumor immune responses through enhanced activity of T cells and NK cells
May contribute to the pathogenesis of certain autoimmune diseases through excessive immune activation
Can enhance activation-induced cell death in repetitively stimulated T cells
In functional assays, recombinant TNFRSF9 proteins typically bind to TNFSF9 with EC50 values in the range of 1.011-3.702 ng/mL, indicating a high-affinity interaction .
Several methodologies can effectively characterize TNFRSF9-TNFSF9 binding:
Functional ELISA:
Add varying concentrations of TNFSF9
Detect binding using secondary detection reagents
Expected EC50 values: 1.011-3.702 ng/mL for high-quality preparations
Surface Plasmon Resonance (SPR):
Provides real-time kinetic analysis of binding events
Can determine association/dissociation rates and binding affinities
Has been successfully employed in studies characterizing TNFRSF9-targeting bispecific antibodies
Cell-Based Binding Assays:
Utilizes cells expressing TNFRSF9 and fluorescently-labeled TNFSF9
Provides information about binding in a cellular context
Can be analyzed by flow cytometry
The choice of method depends on specific research questions - ELISA is suitable for routine binding confirmation, while SPR provides more detailed binding kinetics information essential for therapeutic development.
When evaluating recombinant TNFRSF9 proteins for research applications, several critical quality attributes should be assessed:
Variations from expected specifications may indicate issues with protein folding, stability, or post-translational modifications that could affect experimental outcomes.
The choice between carrier-free and carrier-containing preparations involves important methodological considerations:
Carrier-Free Preparations:
Recommended for applications where BSA or other carriers might interfere
Typically lyophilized from PBS solutions and should be reconstituted at higher concentrations (approximately 500 μg/mL)
More susceptible to adsorption losses and stability issues
Requires careful handling to prevent protein loss during dilution and storage
Essential for certain binding assays, SPR studies, and in vivo applications
Carrier-Containing Preparations:
Include stabilizers like BSA that enhance shelf-life and stability
Allow for storage at more dilute concentrations
More resistant to freeze-thaw cycles and handling stresses
Suitable for most cell culture applications and as ELISA standards
May interfere with certain downstream applications or introduce unwanted variables
For either format, it is advisable to avoid repeated freeze-thaw cycles by preparing single-use aliquots and storing at -80°C for maximum retention of activity.
TNFRSF9 is widely implemented in second-generation CAR-T cell therapy as a costimulatory domain, with several key design considerations:
Domain Selection:
The intracellular domain of TNFRSF9 provides critical costimulatory signals that enhance T cell activation and persistence
Typically, amino acids from the cytoplasmic tail are incorporated into the CAR construct downstream of the CD3ζ signaling domain
Comparative Advantages:
TNFRSF9-containing CARs typically demonstrate enhanced T cell persistence compared to CD28-based designs
These CARs show improved memory T cell formation
May provide better long-term anti-tumor responses, particularly important for solid tumors
Optimization Strategies:
To assess CAR-T cells incorporating TNFRSF9 domains, researchers should evaluate not only immediate cytolytic activity but also long-term persistence, memory formation, and resistance to exhaustion for a complete functional profile.
TNFRSF9-targeted approaches for cancer immunotherapy include several promising strategies:
Agonistic Antibodies:
Direct TNFRSF9 agonists that stimulate T cell activation and proliferation
Have shown remarkable efficacy in murine tumor models but face challenges with systemic toxicity in clinical translation
Bispecific Antibodies:
Combine TNFRSF9 agonism with targeting of tumor antigens or checkpoint molecules
Example: CD137×PD-L1 bispecific antibodies that provide context-dependent T cell costimulation and checkpoint blockade
These designs restrict TNFRSF9 activation to the tumor microenvironment, potentially improving safety
Engineered TNFSF9 Ligands:
Recombinant versions of the natural ligand with enhanced stability or targeting properties
May provide more physiological receptor activation compared to antibody approaches
Methodological Considerations:
Careful epitope selection significantly impacts agonistic activity
Antibody format affects pharmacokinetics and tissue penetration
Fc engineering can modulate FcγR binding to enhance agonistic activity
Affinity balancing is critical to prevent systemic activation and toxicity
Clinical development has been challenging due to hepatotoxicity observed with some TNFRSF9 agonists, highlighting the need for more selective targeting approaches.
Research has revealed a unique pattern of TNFRSF9 expression in human gliomas with important implications:
Expression Pattern:
TNFRSF9 is considerably upregulated in human gliomas compared to normal brain tissue
Surprisingly, expression is predominantly found in non-neoplastic reactive astrocytes rather than in tumor cells or typical immune cells
Distribution is primarily perivascular and peritumoural, with significantly higher expression in IDH-1 mutant gliomas
Research Implications:
This pattern challenges direct translation of findings from murine models where TNFRSF9-targeting approaches have shown complete tumor eradication
Identifies a novel TNFRSF9-positive reactive astrocytic phenotype that may have unique functional properties in the tumor microenvironment
Suggests TNFRSF9 might be involved in forming a reactive tumor microenvironment rather than direct anti-tumor immunity in this context
Methodological Recommendations:
Combined immunohistochemistry and immunofluorescence approaches with cell-type specific markers are essential for accurate characterization
Careful validation of antibody specificity is critical given the unexpected cellular localization
Single-cell analysis may provide further insights into heterogeneity of expression within reactive astrocyte populations
These findings highlight the importance of thorough characterization of target expression patterns in human samples before clinical translation of promising preclinical results.
Emerging research has begun to elucidate the epigenetic regulation of TNFRSF9:
DNA Methylation:
Multiple CpG sites within the TNFRSF9 gene show differential methylation patterns that correlate with expression levels
At least twelve CpG sites have been identified as potentially relevant for regulation
Methylation patterns may correlate with response to immunotherapy, particularly checkpoint inhibitors in melanoma
Methodological Approaches:
Bisulfite sequencing provides single-nucleotide resolution of methylation status across the TNFRSF9 gene
Methylation-specific PCR can rapidly assess methylation at specific CpG sites
Pyrosequencing offers quantitative assessment of methylation levels
Methylation arrays allow for genome-wide analysis including the TNFRSF9 locus
Research Implications:
Correlation analyses between methylation patterns and gene expression are critical to establish functional significance
Integration with clinical outcome data may identify predictive biomarker signatures
Combined analysis with other epigenetic marks (histone modifications, chromatin accessibility) provides a more complete regulatory picture
Understanding epigenetic regulation of TNFRSF9 may lead to novel therapeutic strategies combining epigenetic modifiers with immunotherapeutic approaches.
TNFRSF9 has emerging potential as a biomarker in several contexts:
Predictive Biomarker for Immunotherapy:
DNA methylation patterns at the TNFRSF9 locus may correlate with response to anti-PD-1 immunotherapy in melanoma
Expression levels on tumor-infiltrating lymphocytes may indicate immune activation status
Soluble forms of TNFRSF9 are elevated in certain disease states and may serve as liquid biopsy markers
Prognostic Biomarker:
Expression patterns in tumor microenvironments may correlate with clinical outcomes
The distribution pattern (perivascular/peritumoural) appears to have biological significance in gliomas
Higher expression in IDH-1 mutant gliomas suggests correlation with molecular subtypes
Methodological Considerations:
Standardized assays with validated cutoff values are needed for clinical application
Tissue-based versus circulating biomarker approaches have different strengths and limitations
Integration with other immune markers may provide more robust predictive signatures
Sample collection timing relative to treatment may significantly impact biomarker performance
Further research correlating TNFRSF9 methylation, expression, and clinical outcomes across multiple cancer types is needed to fully establish its biomarker utility.
Several innovative strategies are being developed to mitigate the hepatotoxicity and other adverse effects associated with TNFRSF9 agonism:
Conditional Activation Approaches:
Tumor-targeted bispecific antibodies that activate TNFRSF9 only in the presence of specific tumor antigens
pH-sensitive antibodies that preferentially activate in the acidic tumor microenvironment
Protease-activated antibodies that become functional only in the presence of tumor-associated proteases
Engineering Strategies:
Fc domain modifications that fine-tune FcγR interactions and subsequent agonistic activity
Alternative antibody formats with optimized valency and binding properties
Antibody fragments with limited systemic exposure but retained tumor penetration
Combination Approaches:
Lower-dose TNFRSF9 agonism combined with complementary immunotherapies
Sequential rather than concurrent administration protocols
Local rather than systemic administration for specific tumor types
These approaches require sophisticated preclinical models, including humanized mouse models and ex vivo human tissue systems, to accurately predict both efficacy and toxicity before clinical translation.
Single-cell technologies offer unprecedented opportunities to resolve TNFRSF9 biology in complex tissues:
Single-cell RNA Sequencing:
Reveals cell type-specific expression patterns of TNFRSF9 and TNFSF9
Identifies co-expressed receptors and downstream signaling components
Characterizes heterogeneity in expression levels within nominally similar cell populations
CITE-seq and Protein Analysis:
Simultaneously assesses TNFRSF9 protein levels and transcriptional profiles
Correlates protein expression with activation states and functional markers
Identifies post-transcriptional regulatory mechanisms
Spatial Transcriptomics:
Maps TNFRSF9 expression within tissue architecture
Correlates expression with specific microenvironmental niches
Particularly valuable for understanding the perivascular/peritumoural distribution observed in gliomas
Epigenetic Analysis at Single-cell Level:
Single-cell ATAC-seq reveals chromatin accessibility at the TNFRSF9 locus
Single-cell methylation analysis identifies cell-specific regulatory patterns
Integrative multi-omic approaches connect epigenetic state to expression and function
These technologies could resolve contradictory findings between animal models and human samples, such as the unexpected expression of TNFRSF9 on reactive astrocytes in human gliomas, potentially leading to more precisely targeted therapeutic strategies.