Recombinant Human Tumor necrosis factor receptor superfamily member 9 protein (TNFRSF9),Partial (Active)

Shipped with Ice Packs
In Stock

Description

Core Features

PropertyDetailsSource
Expression RegionLeu24-Gln186 (partial extracellular domain)
Host SystemMammalian cells (CHO or HEK293)
TagC-terminal 6xHis-tag or Fc fusion (varies by construct)
Theoretical MW18.1–21.8 kDa (monomer)
Observed MW25–38 kDa (SDS-PAGE, reducing conditions)
Purity>95% (SDS-PAGE, Coomassie Blue/HAL method)
Endotoxin Level<1.0 EU/μg (LAL method)

Mechanistic Insights:

  • 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 .

Cancer Immunotherapy

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 .

Autoimmune Diseases

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 .

Quality and Handling Protocols

ParameterRecommendationSource
Storage-20°C (lyophilized) or 2–8°C (reconstituted)
ReconstitutionPBS (500 μg/mL for His-tag; 400 μg/mL for Fc Avi-tag)
StabilityAvoid freeze-thaw cycles; use manual defrost freezers

Critical Considerations:

  • Endotoxin Control: Essential for in vivo studies to prevent confounding inflammatory responses .

  • Batch Variability: EC50_{50} values may differ between suppliers due to expression systems or tags .

Therapeutic Hurdles

  • 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 .

Innovation Opportunities

  • Targeted Delivery: Tumor microenvironment-specific agonists to minimize off-tumor effects .

  • Biomarker-Guided Trials: Stratifying patients based on TNFRSF9 expression or ligand levels .

Comparative Analysis of Recombinant Constructs

Construct TypeAdvantagesLimitationsSource
His-tag (Partial)High solubility, low immunogenicityRequires optimization for multivalent binding
Fc ChimeraEnhanced stability, Fc-mediated dimerizationPotential steric hindrance in assays
Fc Avi-tagBiotinylation compatibility, ultra-high affinityRequires streptavidin conjugation steps

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Please reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% of glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers may use this as a reference.
Shelf Life
The shelf life is influenced by numerous factors, including storage conditions, buffer ingredients, storage temperature, and the inherent stability of the protein itself. Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt; aliquoting is necessary for multiple use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
4 1BB; 4 1BB ligand receptor; 4-1BB ligand receptor; 4-1BB Ligand Receptor T Cell; 4-1BB, mouse, homolog of; Antigen 4-1BB Homolog; CD 137; CD137; CD137 antigen; CDw137; HLDA VI; Homolog of mouse 4 1BB; ILA; induced by lymphocyte activation (ILA); Induced by lymphocyte activation; Interleukin activated receptor homolog of mouse Ly63; Ly63, mouse, homolog of; MGC2172; OTTHUMP00000044294; Receptor protein 4 1BB; T cell antigen 4 1BB homolog; T cell antigen ILA; T-cell antigen 4-1BB homolog; T-cell antigen ILA; TNF receptor superfamily member 9; TNFRSF9; TNR9_HUMAN; Tumor necrosis factor receptor superfamily member 9
Datasheet & Coa
Please contact us to get it.
Expression Region
19-184aa
Mol. Weight
17.7 kDa
Protein Length
Partial
Purity
>97% as determined by SDS-PAGE.
Research Area
Cancer
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Receptor for TNFSF9/4-1BBL. Potentially active during T cell activation.
Gene References Into Functions
  1. Three single nucleotide polymorphisms (SNPs) (rs161827, rs161818, and rs161810) within the CD137 gene and their association with ischemic stroke were investigated in a northern Chinese Han population. rs161827 exhibited a significant difference between individuals with and without diabetes and the control group. rs161818 and rs161810 differed significantly between patients without diabetes and the controls. All three SNPs were statistically significant in the combined stroke group. PMID: 28755037
  2. This study presents LOAd703, a designed adenovirus equipped with trimerized CD40L and 4-1BBL, which respectively activate the CD40 and 4-1BB pathways. PMID: 28536305
  3. Continuous 4-1BB costimulation in chimeric antigen receptors hinders T cell survival and is vector-dependent. PMID: 28978471
  4. Cetuximab-mediated NK-cell expression of CD137 on tumor-infiltrating lymphocytes is contingent upon FcgammaRIIIa polymorphism. In patients with head and neck cancer receiving neoadjuvant cetuximab treatment, upregulation of CD137 by intratumoral, cetuximab-activated NK cells correlated with FcgammaRIIIa V/F polymorphism and predicted clinical response. PMID: 27496866
  5. This study systematically evaluated a series of CAR constructs targeting glypican-3 (GPC3), which is selectively expressed on several solid tumors. The study compared GPC3-specific CARs encoding CD3zeta (Gz) alone or in combination with costimulatory domains derived from CD28 (G28z), 4-1BB (GBBz), or CD28 and 4-1BB (G28BBz). PMID: 27530312
  6. 4-1BB and 4-1BBL serve as markers for predicting patients' course and represent a valuable screening target for individuals with acute myeloid leukemia at initial diagnosis. PMID: 27388616
  7. The role of CD137-CRDI (cysteine-rich domain I) in the binding of CD137-CD137L was further investigated. PMID: 27430526
  8. Egr2-driven cell surface proteins LAG-3 and 4-1BB can identify dysfunctional tumor antigen-specific CD8(+) TIL. PMID: 28115575
  9. Findings indicate that CD137 antigen is a valuable marker for identifying Mycobacterium tuberculosis (Mtb)-reactive CD4(+) T cells (Mtb-reactive CD4(+) T cells) through flow cytometry. PMID: 28218958
  10. Anti-4-1BB single-chain variable fragments enhanced surface CD69 expression and interleukin-2 production in stimulated CCRF-CEM cells, confirming the agonistic effect of the selected single-chain variable fragments. The data from this study provide a basis for further investigations into the biological functions of anti-4-1BB single-chain variable fragments in future studies. PMID: 28347235
  11. Studies suggest that adoptive T cell therapy and CD137 antigen offer significant opportunities to enhance the efficacy of current cancer immunotherapies. PMID: 26970765
  12. In conjunction with the T cell receptor, CD137 promotes memory T cells, cell respiration, fatty acid oxidation, and mitochondrial biogenesis. PMID: 26885860
  13. These studies provide the first direct evidence that ligation of tumor necrosis factor superfamily members and their cognate receptors is crucial for controlling viral lytic replication. PMID: 26467721
  14. Our findings identify a novel, TNFRSF9-positive, reactive astrocytic phenotype in human gliomas. PMID: 24606203
  15. Human genetic evidence supports the involvement of CD137 in atherosclerosis. PMID: 25032953
  16. Upon activation, transferred human T lymphocytes express the inducible surface antigens hPD-1 and hCD137 on their plasma membrane. PMID: 26113085
  17. Our results provide biological explanations for the antitumor effects of CD19 CARs and for the observations that CD19 CAR T cells incorporating the 4-1BB costimulatory domain exhibit greater persistence than those incorporating CD28 in clinical trials. PMID: 25939063
  18. Upregulation of CD137 expression through LMP1 by EBV promotes cell survival in T or NK cells. PMID: 25409517
  19. Based on CD137 or CD154 expression. PMID: 25367298
  20. High expression of CD137 is associated with type 1 diabetes. PMID: 24797972
  21. DENV C disrupts Daxx and NF-kappaB interaction to induce CD137-mediated apoptosis during DENV infection. PMID: 25019989
  22. The action of agonist anti-4-1BB in suppressing autoimmune and allergic inflammation was entirely dependent on Galectin-9 (Gal-9). Gal-9 directly bound to 4-1BB at a site distinct from the binding site of antibodies and the natural ligand of 4-1BB. PMID: 24958847
  23. A role for the TNFR-family member CD137 in the immunobiology of human cancer where it is preferentially expressed on tumor-reactive subset of tumor-infiltrating lymphocytes. PMID: 24045181
  24. Monocytes interact with iNKT cells to increase expression of 4-1BBL and 4-1BB, and in conjunction with this pathway, maintain their numbers at baseline. PMID: 24639347
  25. Findings demonstrate that immunohistochemistry for CD137L can reliably distinguish small B-cell lymphomas from reactive lymphoid aggregates. PMID: 24746207
  26. Dengue virus induces CD137 signaling to enhance apoptosis by increasing TNF-alpha production through activation of p38 MAPK. PMID: 23557259
  27. This is the first study to demonstrate that this member of the TNF superfamily, CD137, is modulated by SAHA treatment in breast cancer cells. PMID: 22797667
  28. The CD137 multi-parameter flow cytometry fast assay enables phenotypic and functional determination of alloreactive precursor frequencies of both CD4+ and CD8+ T cells with high sensitivity and specificity. PMID: 23750604
  29. Co-stimulation through 4-1BB/CD137 enhances the expansion and function of CD8(+) melanoma tumor-infiltrating lymphocytes for adoptive T-cell therapy. PMID: 23560068
  30. These data provide evidence that the 4-1BB signal is a critical regulator of gammadelta T cells. PMID: 23640752
  31. The mechanisms responsible for the effect of CD137 signaling on TNF-alpha production are attributed to a decrease in TNF-alpha production by antigen-presenting cells (APCs) and potentially an increase in APC apoptosis. PMID: 23437083
  32. Our results uncover a new regulatory mechanism for CD137L expression that mediates immune escape by HRS cells and identify CD137 as a potential target for immunotherapy of Hodgkin lymphoma. PMID: 23204227
  33. Patients with head and neck cancer exhibit decreased levels of alternative co-stimulatory receptors OX40 and 4-1BB. PMID: 22204816
  34. 4-1BB (CD137), in conjunction with CD103, marks mesenteric lymph node dendritic cells (DCs) with the highest level of retinal dehydrogenase (RALDH) activity, and ligation of 4-1BB sustains RALDH expression in these gut DCs. PMID: 22896640
  35. CD137 protein is expressed by a specific group of hematolymphoid tumors, including classical Hodgkin lymphoma, T-cell and NK/T-cell lymphomas, and follicular dendritic cell neoplasms. PMID: 22901750
  36. Treatment with a CD137 agonistic antibody induces CCL21 expression and DC accumulation near lymphatic vessels. Collectively, our results demonstrate that the inflammatory function of lymphatic vessels can be regulated by CD137. PMID: 22593548
  37. CD137:CD137L interactions regulate the innate and adaptive immune response of the host against M. tuberculosis. PMID: 21747409
  38. A statistically significant positive correlation exists between CD137 expression and complex coronary stenosis morphology. PMID: 21396356
  39. Data indicate that 4-1BBL mediates NK-cell immunosubversion in CLL, potentially contributing to the reportedly compromised efficacy of Rituximab in inducing NK-cell reactivity in this disease. PMID: 22144129
  40. CD137 activity is directly proportional to colorectal cancer stage. Surgical resection of the tumor results in increased CD134 and CD137 expression. PMID: 22343199
  41. This study demonstrates that the inflammatory and cytotoxic function of CD4(+)CD28(null) T cells can be inhibited by blocking OX40 and 4-1BB costimulatory receptors. PMID: 22282196
  42. The sCD137 levels correlate with the probability of complications and lethality. The association of sCD137, a product of activated T cells, with the severity of acute pancreatitis suggests that T cells contribute to the pathogenesis of acute pancreatitis. PMID: 21963611
  43. CD137 plays a role in breast cancer, and its specific antibody can be used to enhance trastuzumab efficacy. PMID: 22326955
  44. Conditioned medium from Lewis Lung Carcinoma cells caused significant upregulation of 4-1BB in mast cells. PMID: 22343053
  45. Data indicate that ex4-1BBL augments 4-1BB expression not only on the primed T cell but also on DCs. PMID: 21745658
  46. The measurement of a single gene expressed by tumor cells (LMO2) and a single gene expressed by the immune microenvironment (TNFRSF9) powerfully predicts overall survival in patients with diffuse large B-cell lymphoma. PMID: 21670469
  47. This work is the first to demonstrate the contribution of CD137 signaling to DENV-mediated apoptosis. PMID: 21669186
  48. CD137 ligand can also be expressed as a transmembrane protein on the cell surface and transmit signals into the cells on which it is expressed (reverse signaling). PMID: 20643812
  49. Results suggest a two-step model of M cell differentiation, with initial CD137-independent commitment to the M cell lineage followed by CD137-CD137L interaction of M cells with CD137-activated B cells or dendritic cells for functional maturation. PMID: 20616340
  50. Data support a role for CD137 in the recruitment of monocytes to inflammatory tissues. PMID: 20347151

Show More

Hide All

Database Links

HGNC: 11924

OMIM: 602250

KEGG: hsa:3604

STRING: 9606.ENSP00000366729

UniGene: Hs.738942

Subcellular Location
Membrane; Single-pass type I membrane protein.
Tissue Specificity
Expressed on the surface of activated T-cells.

Q&A

What is TNFRSF9 and what is its molecular structure?

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 .

What cell types express TNFRSF9 and under what conditions?

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.

How does the TNFRSF9-TNFSF9 signaling axis function?

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 .

What are the optimal methods for assessing TNFRSF9-TNFSF9 binding interactions?

Several methodologies can effectively characterize TNFRSF9-TNFSF9 binding:

Functional ELISA:

  • Immobilize recombinant TNFRSF9 (typically 50 ng/mL)

  • 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.

What are the critical quality attributes for recombinant TNFRSF9 proteins?

When evaluating recombinant TNFRSF9 proteins for research applications, several critical quality attributes should be assessed:

AttributeAssessment MethodExpected Specifications
PuritySDS-PAGE, SEC>95% purity
Endotoxin ContentLAL test<1.0 EU/μg protein
Binding ActivityFunctional ELISA with TNFSF9EC50: 1.011-3.702 ng/mL
Molecular WeightSEC-MALS, SDS-PAGE~21.8-28 kDa depending on glycosylation
Oligomeric StateSEC-MALSPredominantly monomeric in solution
Post-translational ModificationsMass spectrometryPresence of expected glycosylation patterns

Variations from expected specifications may indicate issues with protein folding, stability, or post-translational modifications that could affect experimental outcomes.

How should researchers handle carrier-free versus carrier-containing preparations of TNFRSF9?

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.

How is TNFRSF9 utilized in CAR-T cell engineering and what are the design considerations?

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.

What approaches are being developed for TNFRSF9-targeted cancer immunotherapy?

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.

What is the significance of TNFRSF9 upregulation in gliomas?

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.

How is TNFRSF9 expression regulated at the epigenetic level?

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.

What is the potential for TNFRSF9 as a biomarker in immunotherapy?

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.

What are the emerging approaches for addressing toxicity challenges in TNFRSF9-targeted therapies?

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.

How might single-cell technologies advance understanding of TNFRSF9 biology?

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.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.