TNFRSF10B Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Target Overview: TNFRSF10B/DR5

TNFRSF10B is a transmembrane protein belonging to the TNF receptor superfamily, encoded by the TNFRSF10B gene (Entrez Gene ID: 8795; UniProt ID: O14763). Key features include:

  • Function: Binds to TNF-related apoptosis-inducing ligand (TRAIL), activating caspase-mediated apoptosis via the FADD-caspase-8 pathway .

  • Disease Association: Overexpressed in cancers such as squamous cell carcinoma, pancreatic cancer, and triple-negative breast cancer .

  • Therapeutic Potential: Selective activation of TNFRSF10B induces apoptosis in tumor cells while sparing normal cells, making it a prime target for oncology .

Research Applications

TNFRSF10B monoclonal antibodies serve as vital tools in:

  • Immunodetection: Used in Western blot, flow cytometry, and immunohistochemistry to study DR5 expression .

  • Functional Studies:

    • Dose-dependent apoptosis in Jurkat cells (ED₅₀: 2–12 ng/mL) .

    • Synergy with SMAC mimetics (e.g., birinapant) in HPV-positive head and neck cancers .

  • Pathway Analysis: Elucidate interactions between TRAIL receptors, FADD, and BIRC2/3 in apoptosis regulation .

Clinical and Preclinical Findings

Key Studies:

  • Phase I/II Trials: Lexatumumab demonstrated safety and preliminary efficacy in pediatric solid tumors and malignant pleural mesothelioma .

  • Combination Therapies: Enhanced anti-tumor activity when paired with birinapant in HPV-positive HNSCC models .

  • ADC Efficacy: Anti-TNFRSF10B-MMAF conjugates showed potent cytotoxicity in vitro (IC₅₀: <10 nM) .

Table 1: Selected Anti-TNFRSF10B Antibody-Drug Conjugates (ADCs)

Product IDAntibodyLinkerPayloadTarget Indication
ADC-W-1876LexatumumabMC (noncleavable)MMAFSolid tumors
ADC-W-1882TigatuzumabMCMMAFBreast cancer
ADC-W-1875LexatumumabSPDBDM4Lymphoma

Challenges and Future Directions

  • Resistance Mechanisms: Tumors with FADD amplification or BIRC2/3 deletions show reduced sensitivity to DR5 agonists .

  • Biomarker Development: Efforts focus on identifying patients with high TNFRSF10B expression for targeted therapies .

  • Next-Generation ADCs: Innovations in linker-payload combinations (e.g., duocarmycin SA) aim to improve therapeutic indices .

Product Specs

Form
Purified mouse monoclonal antibody in phosphate-buffered saline (PBS, pH 7.4) containing 0.02% sodium azide, 0.1 mg/ml bovine serum albumin (BSA), and 50% glycerol.
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchase method and location. For specific delivery times, please consult your local distributors.
Synonyms
Fas like protein; Apoptosis inducing protein TRICK2A/2B; Apoptosis inducing receptor TRAIL R2; CD 262; CD262; CD262 antigen; Cytotoxic TRAIL receptor 2; Death domain containing receptor for TRAIL/Apo 2L; Death domain containing receptor for TRAIL/Apo2L;

Q&A

What is TNFRSF10B and what is its functional significance?

TNFRSF10B (tumor necrosis factor receptor superfamily member 10B), also known as DR5, KILLER, TRAIL-R2, and several other aliases, is a member of the TNF-receptor superfamily that contains an intracellular death domain. This receptor is activated by tumor necrosis factor-related apoptosis inducing ligand (TNFSF10/TRAIL/APO-2L) and transduces apoptosis signals . The protein is located on chromosome 8p21.3 and plays a critical role in extrinsic apoptotic pathways. FADD (Fas-associated death domain protein), a death domain-containing adaptor protein, is essential for mediating the apoptotic signal from TNFRSF10B, as demonstrated in studies with FADD-deficient mice . Two transcript variants encoding different isoforms and one non-coding transcript have been identified for this gene, suggesting complex regulation of its expression and function. Understanding the molecular characteristics of TNFRSF10B is fundamental to developing targeted therapeutic strategies against cancer cells.

How do researchers validate the apoptotic activity of TNFRSF10B monoclonal antibodies?

Validation of TNFRSF10B monoclonal antibody-induced apoptosis typically employs multiple complementary approaches:

  • Cell viability assays: MTT assays are commonly used to determine dose-dependent effects of anti-TNFRSF10B antibodies, as demonstrated in studies with MB231_CFP-YFP cells treated with varying concentrations (0-200 μg/ml) of anti-DR5 antibody for 20 hours .

  • Flow cytometry analysis: Annexin-V-APC and propidium iodide (PI) staining followed by flow cytometry provides quantitative assessment of early and late apoptotic cells after treatment. This method enables statistical analysis of apoptotic effects, with EC50 values determined using nonlinear curve fitting approaches .

  • Western blot analysis: Detection of cleaved caspase 3, caspase 8, and other apoptotic markers provides biochemical evidence of apoptotic pathway activation. In TNFRSF10B research, western blotting with antibodies specific to caspase 3 and caspase 8 can confirm the mechanism of action .

  • Concentration dependency: Establishing dose-response relationships is crucial, with typical ED50 values for TRAIL R2/TNFRSF10B monoclonal antibody-induced apoptosis ranging from 2-12 ng/mL in Jurkat human acute T cell leukemia cells .

These methodologies collectively provide robust validation of antibody functionality and specificity in research applications.

What methods are recommended for detecting TNFRSF10B expression in experimental systems?

Several techniques have been optimized for detecting TNFRSF10B expression in research contexts:

  • Western blot analysis: Using specific anti-TNFRSF10B antibodies, researchers can detect characteristic bands at approximately 44 and 52 kDa under reducing conditions. This approach has been validated in human cancer cell lines including HCT-116 colorectal carcinoma and HepG2 hepatocellular carcinoma . Optimal protocols typically use PVDF membranes probed with 1 μg/mL of mouse anti-human TRAIL R2/TNFRSF10B monoclonal antibody followed by HRP-conjugated secondary antibodies.

  • Flow cytometry: For cell surface expression analysis, fluorophore-conjugated anti-TNFRSF10B antibodies allow quantification of receptor density on intact cells. This method is particularly valuable for comparing expression levels across different cell types or treatment conditions.

  • Immunohistochemistry: For tissue samples, immunohistochemical staining provides spatial information about TNFRSF10B expression patterns, which is especially relevant when analyzing heterogeneous tumor tissues or comparing normal versus malignant tissues.

  • RT-PCR and qPCR: For transcript-level analysis, these techniques can quantify TNFRSF10B mRNA expression, complementing protein-level data and providing insights into transcriptional regulation.

Each method offers distinct advantages depending on the research question, with western blotting and flow cytometry being particularly robust for antibody characterization studies.

How can FRET-based biosensors enhance TNFRSF10B signaling research?

Förster Resonance Energy Transfer (FRET) biosensors represent a sophisticated approach to studying TNFRSF10B-mediated apoptosis with several methodological advantages:

  • Real-time monitoring: FRET biosensors enable continuous observation of signaling events in living cells. MB231_CFP-YFP cells treated with TRAIL (0-300 ng/ml) or anti-DR5 antibody (0-100 μg/ml) demonstrate quantifiable FRET responses that correlate with apoptotic activity . These biosensors allow researchers to capture immediate responses and temporal dynamics that might be missed with endpoint assays.

  • Spatial resolution: Confocal microscopy of FRET signals reveals subcellular localization of TNFRSF10B signaling events. As demonstrated in MB231_CFP-YFP cells treated with increasing TRAIL concentrations, a pronounced shift from YFP acceptor emission (yellow) to CFP donor emission (cyan) occurs with excitation at 458 nm, providing visual evidence of apoptotic pathway activation .

  • Quantitative analysis: FRET efficiency can be calculated and normalized against treatment concentrations to generate dose-response curves. This approach allowed researchers to determine EC50 values using nonlinear curve fitting to normalized FRET values plotted against logarithmic concentrations of TRAIL and anti-DR5 antibody .

  • Validation methodology: FRET biosensor results should be validated against established techniques. For example, researchers confirmed that MB231_CFP-YFP cells retain sensitivity to TRAIL and anti-DR5 antibody by comparing FRET results with MTT assays and Annexin-V/PI staining, establishing correlation between FRET signals and conventional apoptosis measures .

These biosensors are particularly valuable for high-throughput screening of novel TNFRSF10B-targeting agents and for dissecting the kinetics of receptor activation and downstream signaling events.

What is the relationship between genomic alterations in TNFRSF10B and therapeutic response?

The relationship between TNFRSF10B genomic alterations and therapeutic response is complex and tissue-specific:

  • Copy number variations (CNVs): Analysis of TCGA datasets has revealed that TNFRSF10B exhibits distinct CNV patterns across cancer types. In head and neck squamous cell carcinoma (HNSCC), HPV-negative tumors show higher percentages of one-copy loss of TNFRSF10A/B/C/D (TRAILRs), while HPV-positive tumors more frequently display neutral status or one-copy gain . These differences suggest that HPV status influences the genetic landscape of TRAIL receptors.

  • Site-specific patterns: Tumors from different anatomical sites show varying patterns of TNFRSF10B genetic alterations. Oropharyngeal (OP) tumors, which are often HPV-positive, exhibit higher frequencies of one-copy gain of TNFRSF10A/B/C/D, while laryngeal (LR) and oral cavity (OC) tumors more commonly show one-copy deletions . These patterns have implications for predicting sensitivity to TNFRSF10B-targeting therapies.

  • Therapeutic implications: The high percentage loss of FADD and BIRC2/3 coupled with gain in TRAIL receptors in HPV-positive OP tumors suggests these tumors might differ in sensitivity to agents targeting TRAILRs . This hypothesis provides a rationale for personalized therapeutic approaches based on the genetic profile of TNFRSF10B and related apoptotic pathway components.

  • Methodological considerations: Researchers investigating the relationship between TNFRSF10B alterations and therapeutic response should employ multiparametric approaches combining genomic analysis (CNV assessment, sequencing), expression profiling (RNA/protein levels), and functional assays (apoptosis induction, drug sensitivity) to establish clinically relevant correlations.

This genomic information can guide patient stratification for clinical trials of TNFRSF10B-targeting therapies and contribute to the development of companion diagnostics.

How does HPV status influence TNFRSF10B expression and function in cancer?

Human papillomavirus (HPV) status significantly impacts TNFRSF10B biology in cancer, particularly in head and neck squamous cell carcinoma (HNSCC):

  • Distinct genomic alteration patterns: Comprehensive analysis of TCGA datasets has revealed that HPV-positive and HPV-negative HNSCC tumors exhibit significant differences in the copy number variation (CNV) components of TNFRSF10A/B/C/D genes. HPV-negative tumors show a higher percentage of one-copy loss, while HPV-positive tumors more frequently display neutral status or one-copy gain of these receptors . This suggests fundamental differences in the genetic regulation of TRAIL receptors depending on HPV status.

  • Anatomical site correlation: The distribution of TNFRSF10B genetic alterations correlates with anatomical sites that have different HPV prevalence. Oropharyngeal tumors, which are enriched for HPV-positive HNSCC, showed the highest percentage of one-copy loss of FADD and BIRC2/3 genes (apoptosis regulators) while simultaneously exhibiting gains in TRAIL receptors . This unique genetic signature may contribute to the distinct biological behavior of HPV-positive tumors.

  • Therapeutic sensitivity implications: The genetic alterations associated with HPV status may predict differential responses to TNFRSF10B-targeting therapies. HPV-positive tumors with one-copy gain of TRAIL receptors coupled with loss of anti-apoptotic factors (BIRC2/3) might exhibit enhanced sensitivity to TRAIL receptor agonists . This provides a rationale for HPV status as a potential biomarker for response to TNFRSF10B-targeted therapies.

  • Experimental approach: To investigate HPV-related differences in TNFRSF10B function, researchers should consider comparative studies using matched HPV-positive and HPV-negative cell lines, patient-derived xenografts, or organoid models. Functional assays should assess baseline receptor expression, apoptotic pathway activation, and sensitivity to TNFRSF10B-targeting agents.

Understanding these HPV-dependent variations in TNFRSF10B biology can inform the design of more effective therapeutic strategies for different cancer subtypes.

What are the current approaches for developing antibody-drug conjugates targeting TNFRSF10B?

Antibody-drug conjugates (ADCs) targeting TNFRSF10B represent an advanced therapeutic strategy combining the specificity of monoclonal antibodies with the cytotoxicity of payload drugs:

  • ADC components optimization: Current approaches focus on optimizing three key components:

    • Antibody selection: Human anti-TNFRSF10B antibodies like Lexatumumab (IgG1-lambda) provide the targeting component with reduced immunogenicity concerns .

    • Linker chemistry: Both cleavable and non-cleavable linkers are being explored. For example, the maleimidocaproyl (MC) linker represents a non-cleavable option that affects the mechanism of drug release within target cells .

    • Cytotoxic payload: Microtubule inhibitors such as MMAF (monomethyl auristatin F) are commonly utilized. These agents bind to tubulins, interrupt microtubule dynamics, and induce cell death after being delivered into cancer cells via receptor-mediated endocytosis .

  • Mechanism of action: TNFRSF10B-targeted ADCs operate through a dual mechanism:

    • Direct apoptosis induction through receptor clustering and TRAIL pathway activation

    • Delivery of cytotoxic payloads that exert additional killing effects through alternative mechanisms

  • Experimental validation: Researchers developing TNFRSF10B-targeted ADCs typically assess:

    • Binding specificity to TNFRSF10B-expressing cancer cells

    • Internalization efficiency using fluorescently labeled antibodies

    • Cytotoxicity in vitro across multiple cancer cell lines

    • In vivo efficacy in xenograft models, with assessment of tumor regression, survival, and toxicity profiles

  • Considerations for resistance mechanisms: As with other targeted therapies, understanding and addressing resistance mechanisms is crucial. This includes studying TNFRSF10B downregulation, altered internalization kinetics, and efflux of payload drugs through increased expression of drug transporters.

This multi-faceted approach to TNFRSF10B-targeted ADC development illustrates the complexity and potential of these advanced therapeutic agents in cancer treatment.

What are the optimal conditions for validating TNFRSF10B antibody specificity?

Establishing antibody specificity is critical for reliable TNFRSF10B research. Optimal validation protocols include:

  • Western blot validation:

    • Sample preparation: Use cell lysates from multiple cell lines with varying TNFRSF10B expression levels (e.g., HCT-116 colorectal carcinoma and HepG2 hepatocellular carcinoma cells) .

    • Electrophoresis conditions: SDS-PAGE under reducing conditions using appropriate immunoblot buffer systems (e.g., Immunoblot Buffer Group 2) .

    • Antibody concentration: Optimal primary antibody concentration typically ranges from 0.5-1 μg/mL (e.g., 1 μg/mL was effective for detection in HCT-116 and HepG2 lysates) .

    • Expected bands: Look for characteristic bands at approximately 44 and 52 kDa, which represent different glycosylation states of TNFRSF10B .

  • Positive and negative controls:

    • Positive controls: Include cell lines with known TNFRSF10B expression (e.g., Jurkat cells, which are sensitive to TNFRSF10B-mediated apoptosis) .

    • Negative controls: Use TNFRSF10B knockout cells or cells with confirmed low expression.

    • Peptide competition: Pre-incubation of antibody with immunizing peptide should eliminate specific binding.

  • Cross-reactivity assessment:

    • Test reactivity against related TRAIL receptors (TNFRSF10A/TRAIL-R1, TNFRSF10C/TRAIL-R3, TNFRSF10D/TRAIL-R4).

    • Confirm specificity using immunoprecipitation followed by mass spectrometry.

  • Functional validation:

    • Evaluate antibody's ability to induce apoptosis in dose-dependent manner in sensitive cell lines like Jurkat (ED50 typically 2-12 ng/mL) .

    • Confirm apoptosis induction using multiple assays (e.g., Resazurin viability assay, caspase activation, Annexin V/PI staining) .

These comprehensive validation approaches ensure that experimental findings accurately reflect TNFRSF10B biology rather than non-specific effects.

How can researchers effectively measure TNFRSF10B-mediated apoptosis kinetics?

Measuring the kinetics of TNFRSF10B-mediated apoptosis requires time-resolved approaches that capture the temporal dynamics of cell death signaling:

  • Real-time FRET biosensor systems:

    • System setup: MB231_CFP-YFP cell-based FRET biosensors provide quantifiable responses to TNFRSF10B activation. These systems detect caspase activation through the cleavage of FRET probes, offering temporal resolution of apoptotic signaling .

    • Time-course considerations: TRAIL treatments typically require 2.5 hours for optimal response, while anti-DR5 antibody treatments may require longer incubation (up to 20 hours) to achieve comparable effects .

    • Quantification: FRET efficiency changes can be plotted against time to generate kinetic curves, with EC50 values determined by nonlinear curve fitting to normalized FRET values .

  • Live-cell imaging approaches:

    • Confocal microscopy: Time-lapse imaging of cells treated with TRAIL or anti-TNFRSF10B antibodies allows visualization of morphological changes associated with apoptosis.

    • Fluorescent reporters: Annexin V conjugated to fluorophores can detect phosphatidylserine externalization, an early apoptotic event, in real-time.

    • Analysis parameters: Quantify time to initial morphological changes, rate of cell shrinkage, membrane blebbing, and complete cell death to generate comprehensive kinetic profiles.

  • Biochemical time-course studies:

    • Sequential sampling: Collect cells at multiple time points after treatment with TNFRSF10B agonists.

    • Molecular markers: Assess the activation sequence of caspase 8 (initiator), caspase 3 (effector), PARP cleavage, and DNA fragmentation through western blotting and other techniques .

    • Data representation: Generate time-response curves showing the percentage of cells exhibiting specific apoptotic markers at each time point.

  • Flow cytometry time-course:

    • Analyze cells at multiple time points post-treatment using Annexin-V-APC and propidium iodide staining to distinguish early apoptotic (Annexin V+/PI-) from late apoptotic/necrotic (Annexin V+/PI+) populations .

    • Calculate the rate of transition between different cell death stages.

These methodologies allow researchers to characterize not only the extent but also the temporal dynamics of TNFRSF10B-mediated apoptosis, providing insights into the efficiency and mechanisms of cellular response to receptor activation.

What are common challenges in TNFRSF10B antibody experiments and how can they be addressed?

Researchers working with TNFRSF10B antibodies frequently encounter several technical challenges that can be systematically addressed:

  • Variable apoptotic responses across cell lines:

    • Challenge: Different cancer cell lines show widely varying sensitivity to TNFRSF10B antibody-induced apoptosis.

    • Solution: Perform initial dose-response characterization across multiple cell lines. For example, MB231_CFP-YFP cells might require anti-DR5 antibody concentrations up to 200 μg/ml for optimal response . Select appropriate positive controls (e.g., Jurkat cells) that reliably respond to TNFRSF10B activation .

    • Approach: Determine EC50 values using nonlinear curve fitting and establish cell line-specific working concentrations before proceeding to complex experiments .

  • Inconsistent antibody performance:

    • Challenge: Batch-to-batch variation or storage conditions may affect antibody performance.

    • Solution: Validate each new antibody lot using western blot against reliable positive controls (e.g., HCT-116 or HepG2 cell lysates) . Confirm the presence of characteristic bands at approximately 44 and 52 kDa under reducing conditions .

    • Approach: Prepare aliquots of validated antibody lots to avoid freeze-thaw cycles and store according to manufacturer recommendations.

  • Background signal in detection methods:

    • Challenge: High background can obscure specific TNFRSF10B detection, particularly in western blotting.

    • Solution: Optimize blocking conditions and antibody dilutions. For western blotting, 1 μg/mL of primary antibody has been validated for specific detection with minimal background .

    • Approach: Include appropriate negative controls and consider using specialized buffer systems (e.g., Immunoblot Buffer Group 2 for western blotting) .

  • Distinguishing direct apoptosis from payload effects in ADC research:

    • Challenge: When studying TNFRSF10B-targeted ADCs, it can be difficult to distinguish receptor-mediated apoptosis from cytotoxicity caused by the payload.

    • Solution: Include appropriate controls including unconjugated antibody, free payload, and non-targeting ADC with the same payload.

    • Approach: Perform mechanistic studies examining markers specific to receptor-mediated apoptosis (e.g., caspase-8 activation) versus payload effects (e.g., microtubule disruption for MMAF-containing ADCs) .

Systematic troubleshooting through these approaches can significantly improve reproducibility and reliability in TNFRSF10B antibody research.

How are genomic approaches advancing TNFRSF10B antibody research?

Genomic approaches are transforming TNFRSF10B antibody research by enabling more precise targeting strategies and patient selection:

  • Integration of multi-omics data:

    • Genomic alterations: Analysis of TCGA datasets has revealed distinct patterns of copy number variations (CNVs) in TNFRSF10B across different cancer types. HPV-positive and HPV-negative head and neck squamous cell carcinomas (HNSCC) show significant differences in CNV profiles, with HPV-negative tumors exhibiting higher percentages of one-copy loss and HPV-positive tumors displaying more neutral status or one-copy gain .

    • Methodology: Researchers should combine DNA sequencing, RNA expression analysis, and protein assessment to comprehensively characterize TNFRSF10B status in experimental models and clinical samples.

    • Application: This multi-dimensional characterization can guide the selection of appropriate cell lines and patient-derived models for antibody testing.

  • Pathway-level genomic analysis:

    • Contextual assessment: Evaluate TNFRSF10B in the context of related death receptor pathway genes. Studies in HNSCC have demonstrated that combining TNFRSF10B alterations with FADD, BIRC2/3, and XIAP status provides more predictive power for therapeutic response than single-gene assessment .

    • Approach: Use pathway enrichment analysis and gene set variation analysis (GSVA) to assess the functional status of death receptor signaling.

    • Implication: This comprehensive approach helps explain why some tumors with high TNFRSF10B expression still resist TRAIL-induced apoptosis due to alterations in downstream pathway components.

  • Biomarker development:

    • Predictive signatures: Develop genomic signatures that predict sensitivity to TNFRSF10B antibodies by correlating response data with baseline genomic profiles.

    • Validation method: Test candidate signatures across multiple cell line panels and patient-derived models.

    • Clinical translation: Utilize these signatures to design basket trials grouping patients by molecular profile rather than cancer type.

  • CRISPR-based functional genomics:

    • Systematic screening: Perform genome-wide CRISPR screens to identify genes that modulate sensitivity to TNFRSF10B antibodies.

    • Validation strategy: Confirm hits through isogenic cell line models with specific gene knockouts or mutations.

    • Therapeutic application: Target identified resistance mechanisms to enhance TNFRSF10B antibody efficacy.

These genomic approaches are advancing beyond correlative studies to provide mechanistic insights and actionable strategies for improving TNFRSF10B-targeted therapies.

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.