TNFRSF9/4-1BB Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Molecular Structure and Target Specificity

TNFRSF9/4-1BB recombinant monoclonal antibodies are typically fully human or humanized IgG2 antibodies engineered for high specificity. Key structural features include:

FeatureDetails
Target EpitopeExtracellular domain of human 4-1BB (TNFRSF9)
Molecular Weight~150 kDa (full-length IgG2)
Binding AffinityHigh affinity (sub-nanomolar KD) to 4-1BB, with no cross-reactivity to other TNF receptors
Species ReactivityHuman-specific; limited cross-reactivity with murine 4-1BB (60% sequence homology)

For example, PF-05082566 (described in ) binds the 4-1BB extracellular domain, triggering receptor clustering and downstream signaling without blocking ligand interaction.

Mechanism of Action

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 .

Functional Outcomes:

  • Proliferation of CD8+ and CD4+ T cells

  • Enhanced NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC)

  • Suppression of regulatory T cell (Treg) activity

Key Preclinical Studies

  • 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 .

Clinical Trial Progress

AntibodyPhaseIndicationKey Findings
PF-05082566Phase I/IIAdvanced solid tumors, B-cell malignanciesRobust T-cell expansion; manageable toxicity
Utomilumab (PF-05082566 analog)Phase IIMelanoma, NSCLCPartial responses in 10–15% of patients

Challenges and Mitigation Strategies

  • Hepatotoxicity: Systemic activation of 4-1BB in early trials caused liver inflammation . Solutions include:

    • Tumor-Targeted Formats: Bispecific antibodies (e.g., EGFR/4-1BB) localize activity to tumor sites .

    • Dose Optimization: Lower doses with intermittent scheduling reduce off-target effects .

  • Hyperimmune Activation: 4-1BB knockout mice paradoxically show enhanced T-cell responses, suggesting context-dependent regulation .

Future Directions

  • 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 .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

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.

Form
Liquid
Lead Time
Typically, we are able to dispatch products within 1-3 working days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
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
Target Names
TNFRSF9/4-1BB
Uniprot No.

Target Background

Function
TNFRSF9/4-1BB is a receptor for TNFSF9/4-1BBL. It is thought to be active during T cell activation.
Gene References Into Functions
  1. A study of a northern Chinese Han population investigated the association of 3 SNPs (rs161827, rs161818, and rs161810) of the CD137 gene with ischemic stroke. rs161827 was significantly different between patients with and without diabetes and the controls. rs161818 and rs161810 differed significantly between patients without diabetes and the controls. All 3 SNPs were statistically significant in the combination stroke group. PMID: 28755037
  2. A designed adenovirus, LOAd703, was created with trimerized CD40L and 4-1BBL to activate the CD40 and 4-1BB pathways respectively. PMID: 28536305
  3. Tonic 4-1BB costimulation in chimeric antigen receptors has been shown to impede T cell survival and is vector-dependent. PMID: 28978471
  4. Cetuximab-mediated NK-cell expression of CD137 on tumor-infiltrating lymphocytes is dependent on FcgammaRIIIa polymorphism. In neoadjuvant cetuximab-treated patients with head and neck cancer, upregulation of CD137 by intratumoral, cetuximab-activated NK cells correlated with FcgammaRIIIa V/F polymorphism and predicted clinical response. PMID: 27496866
  5. A 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 that encoded CD3zeta (Gz) alone or with costimulatory domains derived from CD28 (G28z), 4-1BB (GBBz), or CD28 and 4-1BB (G28BBz). PMID: 27530312
  6. 4-1BB and 4-1BBL have been identified as markers for predicting patients' course and represent a valuable screening target for patients with acute myeloid leukemia at initial diagnosis. PMID: 27388616
  7. Further investigation has been conducted into the role of CD137-CRDI (cysteine rich domain I) in the binding of CD137-CD137L. 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 useful marker for identifying Mycobacterium tuberculosis (Mtb)-reactive CD4(+) T cells (Mtb-reactive CD4(+) T cells) by 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. These findings provide a rationale for future studies involving the biological functions of anti-4-1BB single chain variable fragments. PMID: 28347235
  11. Studies suggest that adoptive T cell therapy and CD137 antigen offer substantial potential to enhance the efficacy of current cancer immunotherapies. PMID: 26970765
  12. In complex with the T cell receptor, TNFRSF9/4-1BB promotes memory T cells, cell respiration, fatty acid oxidation, and mitochondrial biogenesis. PMID: 26885860
  13. Research provides 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. Research findings reveal 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. Activated, transferred human T lymphocytes express the inducible surface antigens hPD-1 and hCD137 on their plasma membrane. PMID: 26113085
  17. Research provides biological explanations for the antitumor effects of CD19 CARs and for the observations that CD19 CAR T cells incorporating the 4-1BB costimulatory domain are more persistent 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 completely dependent on Galectin-9 (Gal-9). Gal-9 directly bound to 4-1BB, in a site distinct from the binding site of antibodies and the natural ligand of 4-1BB. PMID: 24958847
  23. CD137 plays a role in the immunobiology of human cancer where it is preferentially expressed on a 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. Immunohistochemistry for CD137L reliably distinguishes small B-cell lymphomas from reactive lymphoid aggregates. PMID: 24746207
  26. Dengue virus induces CD137 signaling to enhance apoptosis by increasing TNF-alpha production via activation of p38 MAPK. PMID: 23557259
  27. This is the first study to demonstrate that CD137 is modulated by SAHA treatment in breast cancer cells. PMID: 22797667
  28. The CD137 multi-parameter flow cytometry fast assay allows for 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 improves 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 an important regulator of gammadelta T cells. PMID: 23640752
  31. The mechanisms that account for the effect of CD137 signaling on TNF-alpha production were based on a decrease of TNF-alpha production by antigen presenting cell (APC) and, perhaps, on the increase in APC apoptosis. PMID: 23437083
  32. Research reveals a new regulatory mechanism for CD137L expression that mediates immune escape by HRS cells, and identifies CD137 as a candidate target for immunotherapy of Hodgkin. PMID: 23204227
  33. Head and neck cancer patients have decreased levels of alternative co-stimulatory receptors OX40 and 4-1BB. PMID: 22204816
  34. 4-1BB (CD137), together with CD103, marks mesenteric lymph node dendritic cells (DC) with the highest level of retinal dehydrogenase (RALDH) activity, and ligation of 4-1BB maintains RALDH expression in these gut DC. PMID: 22896640
  35. CD137 protein is expressed by a select group of hematolymphoid tumors, including classical Hodgkin lymphoma, T-cell and NK/T-cell lymphomas, and follicular dendritic cells neoplasms. PMID: 22901750
  36. Treatment with CD137 agonistic antibody induces CCL21 expression and DC accumulation close to lymphatic vessels. Collectively, these findings 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 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, and thus might contribute to the reportedly compromised efficacy of Rituximab to induce NK-cell reactivity in the 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. Research shows 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 levels of sCD137 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 has 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 DC. 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 TNFRSF9/4-1BB and where is it expressed in human tissues?

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 .

How does TNFRSF9 interact with its ligand (TNFSF9/4-1BBL), and what cellular responses result from this interaction?

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 .

What are the validated detection methods for TNFRSF9 in different experimental systems?

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)

How should researchers design experiments to assess TNFRSF9 agonist antibody function in T cell activation assays?

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)

    • T cell subset composition and activation state

  • 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

How do different epitope specificities of anti-TNFRSF9 antibodies affect their agonistic activities?

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 .

What mechanisms contribute to hepatotoxicity observed with some TNFRSF9 agonist antibodies in clinical trials?

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 .

How can researchers design tumor-targeted TNFRSF9 agonist approaches to improve therapeutic index?

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:

    • TNFRSF9 ligand (TNFSF9) fusion proteins with tumor-targeting domains

    • Aptamer-based approaches with conditional activation properties

    • Cell-based delivery systems that release TNFRSF9 agonists locally within tumors

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.

What bidirectional signaling occurs between TNFRSF9 and TNFSF9, and how can this be leveraged in cancer immunotherapy research?

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:

      • Enhanced proliferation and cytokine production (IFN-γ, TNF-α)

      • Increased expression of anti-apoptotic proteins (Bcl-2, Bcl-XL, Bfl-1)

      • Reduced activation-induced cell death

      • Preferential expansion of CD8+ memory T cells

      • Enhanced cytotoxic functions against tumor cells

  • 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:

      • Soluble forms of both TNFRSF9 and TNFSF9 are elevated in certain disease states

      • Monitoring these soluble forms during immunotherapy trials may provide predictive biomarkers

      • Research protocols should include assessments of both membrane-bound and soluble forms

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.

What are common technical challenges in TNFRSF9 detection assays and how can they be addressed?

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:

      • When performing Western blot, expect bands between 32-40 kDa depending on glycosylation state

      • Consider deglycosylation treatments (PNGase F) to obtain more uniform band patterns

      • Use reducing conditions for Western blot analysis to improve consistency

  • 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 .

How should researchers evaluate and compare different anti-TNFRSF9 antibody clones for specific research applications?

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:

      • Compare performance under reducing and non-reducing conditions

      • Determine sensitivity limits with titrated protein amounts

      • Evaluate band patterns across different cell types and tissues

    • 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 .

What are emerging strategies to overcome clinical challenges with TNFRSF9-targeted immunotherapies?

  • 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 .

How can emerging technologies advance our understanding of TNFRSF9 biology and therapeutic applications?

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

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.