TNFRSF9/4-1BB recombinant monoclonal antibodies are typically fully human or humanized IgG2 antibodies engineered for high specificity. Key structural features include:
For example, PF-05082566 (described in ) binds the 4-1BB extracellular domain, triggering receptor clustering and downstream signaling without blocking ligand interaction.
4-1BB agonistic antibodies activate immune cells through these pathways:
NF-κB Activation: Recruitment of TRAF1/2 adaptors initiates NF-κB signaling, upregulating anti-apoptotic proteins (Bcl-xL, Bfl-1) and cytokines (IL-2, IFN-γ) .
Metabolic Reprogramming: Enhances mitochondrial biogenesis and fatty acid oxidation in T cells, supporting long-term survival .
Tumor Microenvironment Modulation: Increases expression of adhesion molecules (ICAM-1, VCAM-1) on tumor vasculature, promoting T-cell infiltration .
Tumor Regression: PF-05082566 demonstrated efficacy in xenograft models, reducing tumor volume by 80–90% as monotherapy and synergizing with anti-PD-1 agents .
Cytokine Induction: MAB8382 (R&D Systems) induced IL-2 secretion in human T cells at ED50 1–10 µg/mL in combination with anti-CD3 .
Safety Profile: Early trimeric antibodies (e.g., EGFR/4-1BB bispecifics) showed reduced hepatotoxicity compared to first-generation agonists .
Hepatotoxicity: Systemic activation of 4-1BB in early trials caused liver inflammation . Solutions include:
Hyperimmune Activation: 4-1BB knockout mice paradoxically show enhanced T-cell responses, suggesting context-dependent regulation .
Combo Therapies: Synergy with PD-1/PD-L1 inhibitors and CAR-T cells is under investigation .
Next-Gen Engineering: FcγR-binding-deficient antibodies (e.g., IgG2σ) minimize effector cell depletion .
Biomarker Development: Identifying patients with high 4-1BB+ tumor-infiltrating lymphocytes may improve response rates .
The TNFRSF9/4-1BB Recombinant Monoclonal Antibody is produced through a rigorous and controlled process, ensuring its exceptional quality and specificity. The process begins with the isolation of B cells from the spleen of an immunized animal. These B cells are then stimulated with recombinant human TNFRSF9 protein as the immunogen. RNA is extracted from the B cells and converted into cDNA via reverse transcription. The TNFRSF9 antibody genes are amplified using specific primers designed for the antibody constant regions and subsequently inserted into an expression vector. This vector is then transfected into host cells, allowing for the production of the TNFRSF9/4-1BB Recombinant Monoclonal Antibody. Following a period of cell culture, the antibody is harvested from the supernatant and purified using affinity chromatography, resulting in a highly purified form suitable for diverse applications. ELISA is conducted to validate the antibody's specificity and functionality in detecting human TNFRSF9 protein. This rigorous production process ensures the generation of a reliable and effective TNFRSF9 Recombinant Monoclonal Antibody, essential for various TNFRSF9-related research and diagnostic applications.
TNFRSF9 (also known as 4-1BB, CD137, ILA, or CDw137) is a type II membrane glycoprotein with a molecular weight of approximately 30 kDa that belongs to the tumor necrosis factor receptor superfamily. Expression patterns vary significantly between resting and activated states. In humans, inducible expression is predominantly found on activated CD4+ and CD8+ T cells after stimulation. TNFRSF9 expression has also been documented on natural killer (NK) cells, dendritic cells (DCs), monocytes, and even some non-immune cells like hepatoma cells and blood vessels from individuals with malignant tumors . Notably, TNFRSF9 expression increases in human peripheral blood mononuclear cells following exposure to DNA-damaging agents such as mitomycin . Western blot analysis typically detects TNFRSF9 at approximately 32 and 40 kDa in human lymphoid tissues and cell lines .
TNFRSF9 interacts with its cognate ligand TNFSF9 (4-1BB ligand/CD137 ligand), a type II glycoprotein with a mass of 34 kDa that is expressed on activated T cells, macrophages, monocytes, dendritic cells, B cells, and B lymphomas . This interaction initiates bidirectional signaling in both the TNFRSF9-expressing cell and the TNFSF9-expressing cell.
For TNFRSF9-expressing cells (primarily activated T cells), binding triggers complex signaling cascades mediated through TNF receptor-associated factors (TRAFs), particularly TRAF1 and TRAF2 . These pathways promote:
Enhanced expression of anti-apoptotic proteins (Bcl-2, Bcl-XL, Bfl-1)
Suppression of pro-apoptotic proteins like Bim
Increased proliferation and cytokine production
Extended survival of activated T cells, particularly CD8+ T cells
Importantly, TNFRSF9 engagement on regulatory T cells can have context-dependent effects, sometimes enhancing and other times inhibiting their immunosuppressive activity . For TNFSF9-expressing cells (typically antigen-presenting cells), reverse signaling increases antigen-presenting capacity and modulates inflammatory responses .
Multiple validated techniques exist for detecting TNFRSF9 expression, each with specific sample preparation requirements and detection sensitivity:
Western Blot Analysis: Effective for detecting TNFRSF9 in cell lysates and tissue samples under reducing conditions. Research has validated detection in HDLM-2 human Hodgkin's lymphoma cell lines and human tonsil tissue, with specific bands appearing at approximately 32 and 40 kDa . Optimal results require:
2 μg/mL of anti-TNFRSF9 antibody
PVDF membrane
Reducing conditions
Appropriate immunoblot buffer systems (e.g., Immunoblot Buffer Group 1)
Flow Cytometry: Particularly useful for detecting TNFRSF9 on cell surfaces in transfected cell lines and primary immune cells. Recommended protocol:
Cell concentration: approximately 106 cells per sample
Antibody concentration: 0.25 μg per 106 cells
Use of appropriate negative controls and gating strategies based on marker expression
Validated in HEK293 cells transfected with human TNFRSF9 and activated human PBMCs
Immunocytochemistry/Immunofluorescence: Effective for visualizing TNFRSF9 cellular localization. Optimal conditions include:
8-25 μg/mL antibody concentration
Room temperature incubation (3 hours)
Fluorescent-conjugated secondary antibodies
DAPI counterstaining for nuclear visualization
Validated in PBMCs treated with PHA (positive control) and HEK293 cells (negative control)
When designing T cell activation assays to evaluate TNFRSF9 agonist antibody function, researchers should consider the following methodological approach:
Experimental Setup:
Use purified T cells (CD4+ and CD8+ separately for differential analysis)
Include appropriate controls:
Isotype control antibody (negative control)
Anti-CD3/CD28 stimulation (positive control)
Combined anti-CD3/CD28 with TNFRSF9 agonist (to assess co-stimulatory effects)
Key Parameters to Measure:
Proliferation (CFSE dilution or thymidine incorporation)
Cytokine production (IFN-γ, TNF-α, IL-2 by ELISA or intracellular staining)
Expression of activation markers (CD25, CD69 by flow cytometry)
Cell survival markers (Annexin V/PI staining)
Cytotoxicity (against target cells)
Important Variables to Control:
Antibody concentration (dose-response relationships are critical)
Timing of antibody addition relative to TCR stimulation
Crosslinking requirements (some antibodies require crosslinking for optimal activity)
Duration of culture (short-term vs. long-term effects)
Advanced Considerations:
Assess FcγR dependency of agonist activity using Fc-engineered variants
Evaluate effects on memory vs. naive T cell subsets
Monitor changes in gene expression profiles (particularly survival genes like Bcl-2, Bcl-XL)
Consider examining bidirectional signaling when APCs are present in the culture system
The epitope specificity of anti-TNFRSF9 antibodies significantly impacts their functional properties, presenting both research challenges and therapeutic opportunities. This relationship stems from several factors:
Domain-Specific Effects: The extracellular portion of TNFRSF9 contains four cysteine-rich domains (CRDs), each with distinct roles in ligand binding and signal transduction. Antibodies targeting different CRDs display varying levels of agonistic activity:
CRD1 (membrane-distal domain): Antibodies binding this region often show stronger agonistic properties
CRD4 (membrane-proximal domain): Antibodies targeting this region may affect receptor clustering differently
Clustering Mechanisms: Effective TNFRSF9 signaling requires receptor trimerization and higher-order clustering. Different epitope-binding antibodies vary in their ability to induce optimal clustering configurations. This explains why some antibodies with similar binding affinities demonstrate dramatically different functional outcomes .
Fc-Dependency Variations: The requirement for FcγR-mediated crosslinking varies substantially between antibodies binding different epitopes:
Some epitope-specific antibodies absolutely require Fc-FcγR interactions for activity
Others maintain significant activity even with F(ab')2 fragments or Fc-silenced variants
Ligand Competition or Synergy: Depending on the targeted epitope, antibodies may:
Competitively inhibit natural ligand (TNFSF9) binding
Allow simultaneous ligand binding, potentially leading to synergistic effects
Induce conformational changes affecting ligand binding affinity
These epitope-dependent variations present significant challenges for researchers, particularly when comparing results across different antibody clones or when translating preclinical findings to clinical applications. Researchers should carefully characterize the epitope specificity and functional properties of anti-TNFRSF9 antibodies in their experimental systems .
Hepatotoxicity represents one of the most significant challenges limiting the clinical development of TNFRSF9 agonist antibodies, despite their promising anti-tumor efficacy. Several proposed mechanisms have been identified through research:
FcγR-Dependent Mechanisms:
Agonistic anti-TNFRSF9 antibodies require crosslinking via FcγR for optimal activity
Liver-resident Kupffer cells express high levels of FcγRs, which may create a "hotspot" for antibody crosslinking and excessive local activation
This hypothesis is supported by observations that Fc-engineered variants with reduced FcγR binding show decreased hepatotoxicity while maintaining anti-tumor activity
TNFRSF9 Expression on Liver-Resident Cells:
Hepatocytes and sinusoidal endothelial cells can express TNFRSF9 under inflammatory conditions
Direct activation of these cells may trigger inflammatory cascades and cellular damage
Studies have shown that TNFRSF9 expression increases on hepatic cells following initial inflammatory stimuli, creating a potential feedback loop
Inflammatory Cytokine Release:
Systemic activation of TNFRSF9 on T cells and other immune populations triggers substantial inflammatory cytokine release
Elevated levels of IFN-γ, TNF-α, and IL-12 can directly mediate hepatocellular injury
This cytokine storm effect may be dose-dependent and more pronounced with higher-affinity antibodies
Polyclonal T Cell Activation in the Liver:
The liver contains a significant population of resident T cells
Non-specific activation of these cells via TNFRSF9 may trigger local inflammation
Memory T cells, which express higher levels of TNFRSF9, are particularly susceptible to activation
These mechanistic insights have guided the development of next-generation TNFRSF9-targeting approaches, including tumor-targeted bispecific antibodies, conditional agonists, and antibodies with modified Fc regions to limit systemic activation while maintaining intratumoral efficacy .
Designing tumor-targeted TNFRSF9 agonist approaches involves multiple sophisticated strategies to enhance anti-tumor efficacy while minimizing systemic toxicity:
Bispecific Antibody Approaches:
Create bispecific antibodies targeting both TNFRSF9 and tumor-associated antigens (TAAs)
This restricts full agonistic activity to the tumor microenvironment where both targets are present
Design considerations include:
Selecting TAAs with high tumor specificity and minimal expression in healthy tissues
Optimizing binding affinity for both targets (typically lower affinity for TNFRSF9 and higher for TAA)
Determining optimal antibody formats (e.g., IgG-like vs. fragment-based constructs)
Engineering Fc functions to modulate FcγR engagement based on desired activity profile
Conditional Activation Approaches:
Design antibodies that activate TNFRSF9 only under specific conditions found in the tumor microenvironment
Examples include:
pH-sensitive antibodies that change conformation at acidic tumor pH
Protease-activatable antibodies that require tumor-associated protease cleavage
Masking approaches where the binding domain is revealed only in the tumor context
Fc Engineering Strategies:
Modify the Fc portion of anti-TNFRSF9 antibodies to control FcγR engagement patterns
Options include:
Silencing Fc to eliminate FcγR binding (reducing systemic activation)
Selectively engaging specific FcγR subtypes (e.g., FcγRIIb) to modulate activity
Creating tumor-selective Fc engagement through bispecific designs
Combination with Local Delivery Methods:
Intratumoral injection to limit systemic exposure
Nanoparticle or liposome encapsulation for tumor-targeted delivery
Combination with radiation therapy to enhance local release and activation
Novel Formats Beyond Traditional Antibodies:
Researchers should employ rigorous preclinical testing of these approaches using both in vitro systems and in vivo models that recapitulate human TNFRSF9 biology to evaluate their potential for clinical translation.
The TNFRSF9-TNFSF9 axis features complex bidirectional signaling that affects both the receptor-expressing cell (forward signaling) and the ligand-expressing cell (reverse signaling), presenting unique opportunities for cancer immunotherapy research:
Forward Signaling Mechanisms (TNFRSF9 to T cells):
Engagement of TNFRSF9 on activated T cells recruits TRAF1 and TRAF2 to cytoplasmic domains
This activates downstream pathways including NF-κB, PI3K/Akt, and MAPK cascades
Functional outcomes include:
Reverse Signaling Mechanisms (TNFSF9 to APCs):
Engagement of TNFSF9 on antigen-presenting cells (APCs) triggers activation signals
This increases:
Antigen-presenting capacity
Production of pro-inflammatory cytokines (TNF-α, IL-6, IL-8, IL-12)
Expression of costimulatory molecules
Maturation of dendritic cells
Notably, TNFSF9 reverse signaling can also trigger apoptosis in some cell types, including certain tumor cells
Research Applications Leveraging Bidirectional Signaling:
TNFRSF9 Agonist Plus TNFSF9-Targeting Approaches:
Simultaneously targeting both receptor and ligand may enhance anti-tumor immunity
Combined approaches could activate both T cells and APCs in the tumor microenvironment
Research designs should include individual and combined targeting to assess potential synergies
Engineered Cell Therapies:
CAR-T cells with integrated TNFRSF9 signaling domains for enhanced persistence
Dendritic cell vaccines engineered to express TNFSF9 for improved T cell priming
Cell-based delivery of TNFRSF9 agonists to the tumor microenvironment
Biomarker Applications:
Understanding and leveraging this bidirectional signaling could lead to more effective immunotherapeutic strategies that simultaneously enhance T cell effector functions and optimize APC-mediated antigen presentation within the tumor microenvironment.
Researchers frequently encounter technical challenges when detecting TNFRSF9 in various experimental systems. These challenges and their solutions include:
Low Basal Expression Levels:
Challenge: TNFRSF9 is often minimally expressed on resting cells, making detection difficult.
Solutions:
Pre-activate T cells with anti-CD3/CD28 or PHA for 24-48 hours before analysis
Use high-sensitivity detection methods such as enzyme-amplified flow cytometry
Consider analyzing TNFRSF9 mRNA levels using qRT-PCR when protein levels are below detection limits
For tissue samples, focus on areas with activated immune cells rather than resting regions
Non-Specific Antibody Binding:
Challenge: Some anti-TNFRSF9 antibodies show cross-reactivity or high background.
Solutions:
Always include proper isotype controls and negative cell lines (e.g., HEK293 parental cells)
Titrate antibody concentration to find optimal signal-to-noise ratio
For flow cytometry, use viability dyes to exclude dead cells which often bind antibodies non-specifically
For Western blotting, validate specific bands using TNFRSF9-transfected cell lysates as positive controls
Glycosylation Variability:
Challenge: TNFRSF9 is a glycoprotein with variable glycosylation patterns, causing inconsistent band patterns.
Solutions:
Soluble TNFRSF9 Interference:
Challenge: Soluble forms of TNFRSF9 can interfere with membrane-bound detection.
Solutions:
For flow cytometry, wash samples thoroughly to remove soluble receptor
When measuring functional responses, consider pre-clearing culture supernatants
Design experiments to measure both membrane-bound and soluble forms
Sample Preparation Effects:
Challenge: Freezing/thawing or enzymatic dissociation can affect TNFRSF9 epitopes.
Solutions:
For flow cytometry, prefer mechanical dissociation over enzymatic methods when possible
Validate detection methods using both fresh and frozen samples to understand potential variations
Process samples consistently to maintain comparability across experiments
These technical insights are derived from validated experimental protocols and represent established solutions to common challenges in TNFRSF9 research .
Systematic evaluation of anti-TNFRSF9 antibody clones is essential for selecting the optimal reagent for specific research applications. This comprehensive evaluation should include:
Binding Characteristics Assessment:
Affinity Determination:
Measure binding kinetics (kon, koff, KD) using surface plasmon resonance
Compare EC50 values across antibody clones using titration in flow cytometry
Assess binding under varying conditions (temperature, pH, buffer composition)
Epitope Mapping:
Determine which domain(s) of TNFRSF9 are recognized by each antibody
Evaluate competition with the natural ligand (TNFSF9)
Assess cross-reactivity with orthologs from other species if cross-species research is planned
Functional Activity Profiling:
Agonistic Potential:
Measure T cell proliferation induced by each antibody clone
Quantify cytokine production (IFN-γ, IL-2, TNF-α)
Assess survival-promoting effects through apoptosis assays
Evaluate impact on cytotoxic activity of CD8+ T cells
Crosslinking Requirements:
Compare activity of whole IgG versus F(ab')2 fragments
Test dependency on FcγR-mediated crosslinking
Evaluate activity with different crosslinking methods (soluble vs. plate-bound)
Application-Specific Validation:
For Flow Cytometry:
Determine optimal antibody concentration for staining
Evaluate performance in different fixation/permeabilization protocols
Assess compatibility with multicolor panels (spectral overlap)
For Western Blotting:
For In Vivo Applications:
Assess half-life and biodistribution
Evaluate potential immunogenicity
Determine optimal dosing schedule
Comparative Analysis Framework:
Create a standardized assessment matrix scoring each antibody on:
Specificity (signal:noise ratio in relevant systems)
Sensitivity (minimum detectable expression level)
Reproducibility (inter-assay variability)
Versatility (performance across multiple applications)
Agonistic/antagonistic functional effects
Document batch-to-batch variability through repeated testing
Consider practical factors including cost, availability, and formulation stability
This systematic approach enables researchers to make informed selections of anti-TNFRSF9 antibodies that are optimally suited to their specific experimental goals and technical requirements .
Tumor-Restricted Activation Strategies:
Development of bispecific antibodies that simultaneously target TNFRSF9 and tumor-associated antigens
Engineering of antibodies that become activated only within the tumor microenvironment (e.g., protease-activated antibodies, pH-sensitive antibodies)
Local administration approaches to limit systemic exposure
Fc Engineering Approaches:
Creation of antibodies with modified Fc regions that selectively engage specific FcγR subtypes
Development of Fc-null variants that require alternative clustering mechanisms
Engineering of antibodies with controlled half-life to manage exposure
Combination Therapy Optimization:
Identifying synergistic combinations with checkpoint inhibitors at doses below toxicity thresholds
Sequential administration protocols to prime the immune system before TNFRSF9 engagement
Combination with targeted therapies that may enhance TNFRSF9 expression selectively on tumor-infiltrating lymphocytes
Novel Delivery Systems:
Nanoparticle-based delivery systems with tumor-targeting properties
Cell-based delivery approaches using engineered immune cells
Localized delivery methods such as intratumoral injection or implantable devices
Biomarker-Guided Patient Selection:
Identification of patient populations less susceptible to toxicity
Development of predictive biomarkers for efficacy and safety
Real-time monitoring approaches to guide dosing and management
These innovative approaches represent the cutting edge of TNFRSF9-targeted immunotherapy research and offer promising paths to overcome the clinical challenges encountered with first-generation agonist antibodies .
Emerging technologies are dramatically expanding our ability to investigate TNFRSF9 biology and develop more effective therapeutic approaches:
Single-Cell Analysis Technologies:
Single-cell RNA sequencing to identify differential responses to TNFRSF9 engagement across immune cell subpopulations
Mass cytometry (CyTOF) for high-dimensional phenotyping of TNFRSF9-expressing cells in complex tissues
Spatial transcriptomics to map TNFRSF9 expression patterns within the tumor microenvironment
These approaches provide unprecedented resolution of cellular heterogeneity and context-specific responses
Advanced Protein Engineering Platforms:
Directed evolution techniques to generate novel TNFRSF9-targeting proteins with unique properties
Structure-guided design of antibodies targeting specific epitopes to modulate receptor clustering
Computational protein design to create entirely new protein scaffolds for TNFRSF9 targeting
These technologies enable rational design of next-generation therapeutics with optimized properties
Intravital Imaging Technologies:
Multiphoton microscopy to visualize TNFRSF9-expressing cell dynamics in living tissues
Bioluminescence resonance energy transfer (BRET) to monitor receptor clustering and signaling in real-time
Intravital imaging with fluorescent reporters to track cellular responses to TNFRSF9 engagement
These approaches provide unprecedented insights into the spatiotemporal dynamics of TNFRSF9 biology
Genome Editing and Screening Technologies:
CRISPR-Cas9 screening to identify genetic modifiers of TNFRSF9 signaling
Knock-in reporter systems to monitor endogenous TNFRSF9 expression and trafficking
Precise genetic engineering to create humanized mouse models with improved translational relevance
These technologies enable systematic dissection of TNFRSF9 signaling networks and biology
Artificial Intelligence and Machine Learning Applications:
Predictive modeling of TNFRSF9 agonist activity based on structural parameters
Pattern recognition in large-scale clinical datasets to identify biomarkers of response
Integration of multi-omics data to understand system-level responses to TNFRSF9 targeting
These computational approaches accelerate discovery and optimize therapeutic development