TRAIL Human

TRAIL / APO2 Ligand Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Expression

TRAIL Human is a ~35 kDa type II transmembrane protein composed of 281 amino acids . Structurally, it features:

  • A C-terminal extracellular domain critical for receptor binding .

  • A unique 12–16 amino acid insertion loop that distinguishes it from other TNF family members and enhances receptor specificity .

  • The ability to form homotrimers, which are essential for activating death receptors .

Expression:

  • Widely expressed in immune cells (e.g., NK cells, monocytes), spleen, thymus, prostate, and lung .

  • Conditionally upregulated in response to interferons and inflammatory stimuli .

Mechanism of Action

TRAIL induces apoptosis via two death receptors (DR4/TRAIL-R1 and DR5/TRAIL-R2) and is regulated by decoy receptors (DcR1, DcR2) .

Key Pathways:

  1. Extrinsic Pathway:

    • TRAIL trimer binds DR4/DR5 → recruits FADD and caspase-8 → forms Death-Inducing Signaling Complex (DISC) → activates effector caspases (3, 6, 7) .

  2. Intrinsic Pathway:

    • Caspase-8 cleaves Bid → truncated Bid (tBid) activates Bax/Bak → mitochondrial outer membrane permeabilization → cytochrome c release → apoptosome formation → caspase-9 activation .

Receptor TypeAlternative NamesFunctionExpression
DR4TRAIL-R1Pro-apoptoticBroad (tumor cells, endothelial cells)
DR5TRAIL-R2Pro-apoptoticSimilar to DR4, higher ligand affinity at 37°C
DcR1TRAIL-R3Decoy (lacks death domain)Limited (immune cells)
DcR2TRAIL-R4Decoy (partial death domain)Protects normal cells (e.g., hepatic stellate cells)

Antitumor Efficacy

  • In vitro/vivo: TRAIL induces apoptosis in >70% of human tumor cell lines (e.g., colorectal, breast, lung) without significant toxicity to normal cells .

  • Clinical Trials:

    AgentPhaseIndicationOutcome
    Mapatumumab (DR4 agonist)IINSCLC, lymphomaPartial responses in 30% of follicular lymphoma patients
    Dulanermin (rhTRAIL)IIAdvanced solid tumorsLimited efficacy as monotherapy
    SCB-313 (TRAIL-trimer)I/IIPeritoneal carcinomatosisOngoing (NCT04123886)

Combination Strategies

  • Sensitizers: Camptothecin, genistein, and YC-1 enhance TRAIL-induced apoptosis by upregulating DR5 or inhibiting survival pathways (e.g., NF-κB) .

  • Challenges: Short serum half-life (~30 minutes) and tumor resistance due to decoy receptor overexpression .

Future Directions

  • Engineering: PEGylation or Fc fusion to prolong half-life .

  • Biomarkers: Identifying tumors with high DR4/DR5 and low DcR1/DcR2 expression for patient stratification .

Product Specs

Introduction
TNF-related apoptosis-inducing ligand (TRAIL), also known as Apo2L, is a protein that acts as a potent inducer of apoptosis (programmed cell death). TRAIL belongs to the TNF superfamily and is characterized as a type II transmembrane protein. In humans, the gene encoding TRAIL is situated on chromosome 3q26. TRAIL demonstrates a specific binding affinity for death receptors DR4 (TRAIL-R1) and DR5 (TRAIL-R2), initiating a caspase-8-dependent apoptotic cascade. This protein exhibits a selective cytotoxicity towards transformed and tumor cells, demonstrating minimal to no effect on normal cells despite its broad expression in various normal tissues.
Description
Recombinant Human TRAIL/APO 2 Ligand, expressed in E. coli, is a single-chain polypeptide. It lacks glycosylation and encompasses 168 amino acids (Met+Arg115-Gly281), resulting in a molecular weight of approximately 21 kDa. The purification of sTRAIL is achieved through proprietary chromatographic techniques.
Physical Appearance
The product appears as a sterile, white powder that has been lyophilized (freeze-dried).
Formulation
The lyophilization process is carried out from a 0.2 μm filtered solution containing 20mM Tris-HCl at pH 8.0 and 150mM NaCl.
Solubility
For reconstitution of the lyophilized TRAIL, it is recommended to use sterile 18 MΩ-cm H2O at a concentration of at least 100 μg/ml. This reconstituted solution can be further diluted as needed using other aqueous solutions.
Stability
Lyophilized APO 2 Ligand, while stable at room temperature for up to 3 weeks, should be stored in a dry environment at temperatures below -18°C. Once reconstituted, TRAIL should be kept at 4°C and used within 2-7 days. For long-term storage, freezing at -18°C is recommended, and the addition of a carrier protein like HSA or BSA (0.1%) is advised. It is important to minimize freeze-thaw cycles.
Purity
The purity of this product exceeds 95.0%, as determined by the following methods: (a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis and (b) SDS-polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Biological Activity
The biological activity of this product is assessed by its ability to induce cytolysis in Murine L929 cells in the presence of Actinomycin D. The ED50, which represents the concentration required for 50% cell death, is determined to be less than 2 ng/ml. This corresponds to a specific activity greater than 5,000,000 IU/mg.
Synonyms
Tumor necrosis factor ligand superfamily member 10, TNF-related apoptosis-inducing ligand, Protein TRAIL, Apo-2 ligand, Apo-2L, CD253 antigen, TL2, APO2L, TNFSF10.
Source
Escherichia Coli.
Amino Acid Sequence
MRERGPQRVA AHITGTRGRS NTLSSPNSKN EKALGRKINS WESSRSGHSF LSNLHLRNGE LVIHEKGFYY IYSQTYFRFQ EEIKENTKND KQMVQYIYKY TSYPDPILLM KSARNSCWSK DAEYGLYSIY QGGIFELKEN DRIFVSVTNE HLIDMDHEAS FFGAFLVG.

Q&A

What is TRAIL and what are its primary functions in human biology?

TRAIL (TNF-Related Apoptosis-Inducing Ligand) is a member of the TNF superfamily that functions primarily through binding to death receptors DR4 and DR5. Upon binding, TRAIL can either induce cell death through apoptotic pathways or activate survival mechanisms depending on the cellular context. The protein plays significant roles in immune regulation, cancer surveillance, and inflammatory processes. Research has shown that TRAIL is involved in multiple cellular functions beyond its well-known role in apoptosis, including macrophage polarization toward specific phenotypes that influence immune responses . The dual nature of TRAIL's signaling capabilities makes it an important target for both basic research and therapeutic development, particularly in cancer immunotherapy and inflammatory disorders where modulation of cell death pathways is therapeutically beneficial.

How does TRAIL Human differ from TRAIL in other species?

Human TRAIL shares structural homology with TRAIL proteins found in other mammals, but exhibits species-specific binding affinities and functional characteristics. The human variant contains 281 amino acids and demonstrates higher specificity for human death receptors compared to non-human receptors. Species-specific differences in TRAIL activity are particularly important when designing preclinical studies, as results from animal models may not directly translate to human applications. For example, murine TRAIL has shown differential binding to decoy receptors compared to human TRAIL, which affects interpretation of mouse model data. These differences necessitate careful validation of findings across species before extrapolating to human clinical applications, especially when evaluating TRAIL-based therapeutics or when using TRAIL as a biomarker in disease progression studies.

What are the primary detection methods for measuring TRAIL expression in human samples?

Several analytical techniques can be employed to detect and quantify TRAIL expression in human samples, each with specific advantages for different research applications:

  • ELISA (Enzyme-Linked Immunosorbent Assay): Provides quantitative measurement of soluble TRAIL in serum, plasma, or cell culture supernatants with high sensitivity.

  • Western blotting: Allows detection of TRAIL protein expression in cell or tissue lysates, providing information about protein size and potential modifications.

  • Flow cytometry: Enables measurement of membrane-bound TRAIL on cell surfaces and can be combined with other markers for multiparametric analysis.

  • Immunohistochemistry/Immunofluorescence: Permits visualization of TRAIL expression patterns within tissue architecture, allowing for spatial localization studies.

  • qPCR and RNA sequencing: Measures TRAIL mRNA expression levels, with RNA-seq providing additional insights into transcript variants and expression patterns relative to other genes.

For research requiring high sensitivity and specificity, combining multiple detection methods is recommended. When reporting TRAIL expression data, researchers should clearly state the specific isoform or variant being measured and include appropriate technical controls to ensure reproducibility .

What are the optimal trial designs for studying TRAIL-based interventions in humans?

When designing trials to evaluate TRAIL-based interventions, researchers should consider several methodological approaches based on the research question and expected outcomes:

Sequential, Multiple Assignment, Randomized Trials (SMART) are particularly valuable for TRAIL research as they allow for multiple stages of randomization, enabling researchers to evaluate different adaptive intervention strategies. This design is ideal for investigating sequential treatment decisions involving TRAIL-based therapies, especially when response-based modifications are anticipated .

Micro-Randomized Trials (MRT) can be useful when studying momentary or quickly changing effects of TRAIL interventions. This approach involves rapid sequential randomizations, making it suitable for evaluating immediate physiological responses to TRAIL administration or for monitoring dynamic TRAIL-receptor interactions .

Hybrid Experimental Designs (HED) combine elements of different trial types and may be optimal for studying TRAIL in complex therapeutic contexts, such as when investigating the integration of TRAIL-based treatments with conventional therapies. This approach allows researchers to simultaneously address questions about multiple timescales and intervention components .

Key considerations for any TRAIL human trial design include:

  • Appropriate biomarker selection to monitor TRAIL activity

  • Careful patient stratification based on receptor expression profiles

  • Adequate follow-up duration to capture both immediate and delayed effects

  • Incorporation of relevant pharmacodynamic endpoints

These design considerations should be determined based on the specific research questions and the current state of knowledge about TRAIL biology in the target condition .

How can researchers control for variability in TRAIL receptor expression in clinical studies?

Controlling for variability in TRAIL receptor expression is critical for reducing experimental noise and improving the reliability of research findings. Researchers should implement the following methodological approaches:

  • Pre-screening and stratification: Before enrollment, assess DR4 and DR5 receptor expression levels in participant samples and stratify accordingly. This prevents imbalances between treatment groups that could confound results.

  • Standardized receptor quantification: Employ consistent quantification methods such as flow cytometry with standardized antibodies or qPCR with validated primers. Establish clear thresholds for receptor expression categories.

  • Temporal consistency: Account for potential diurnal or treatment-induced variations in receptor expression by standardizing sample collection timing and documenting any interventions that might affect expression.

  • Multi-site standardization: For multi-center studies, implement rigorous inter-laboratory validation to ensure consistent receptor measurement across sites.

  • Statistical approaches: Incorporate receptor expression as a covariate in statistical analyses, or consider receptor expression-based subgroup analyses when evaluating treatment effects.

What are the recommended control conditions when studying TRAIL's effects on macrophage polarization?

Experimental control matrix for macrophage polarization studies:

Control TypePurposeImplementation
Untreated controlsBaseline comparisonCulture primary monocyte-derived macrophages without any polarizing stimuli or TRAIL treatment
Vehicle controlsControl for delivery method effectsExpose macrophages to the vehicle/buffer used for TRAIL delivery
Polarization-only controlsIsolate TRAIL effectsApply standard polarizing conditions (e.g., IFN-γ+LPS for M1, IL-4 for M2a, IL-10 for M2c) without TRAIL
Isotype controlsControl for non-specific bindingUse isotype-matched non-targeting antibodies when using antibody-mediated stimulation
Receptor blocking controlsConfirm receptor specificityPre-treat with DR4 or DR5 blocking antibodies before TRAIL exposure
Time-matched controlsAccount for temporal changesMaintain control cultures for identical durations as treatment groups

Additionally, researchers should include positive controls for each polarization state using established methods to induce M1 (classically activated), M2a (alternatively activated), and M2c (deactivated) phenotypes. This comprehensive control strategy enables accurate attribution of observed effects specifically to TRAIL signaling rather than to confounding variables or experimental artifacts .

How does TRAIL signaling interact with other inflammatory pathways in human macrophages?

TRAIL signaling exhibits complex interactions with multiple inflammatory pathways in human macrophages, creating a network of cross-regulation that influences immune responses. RNA sequencing analysis of TRAIL-treated human monocyte-derived macrophages has revealed significant differential expression of genes involved in key inflammatory signaling cascades.

TRAIL pre-treatment significantly modulates NF-κB, JAK-STAT, and MAPK pathways in macrophages subsequently exposed to polarizing stimuli. After TRAIL exposure, macrophages show enhanced responsiveness to M1 polarization signals, with upregulation of pro-inflammatory cytokines like TNF-α, IL-1β, and IL-6. Simultaneously, TRAIL pre-conditioning attenuates the IL-4 and IL-10 signaling pathways that typically drive M2 polarization .

Mechanistically, TRAIL appears to prime macrophages by inducing chromatin remodeling at promoters of inflammatory response genes, facilitating more rapid and robust transcriptional activation upon subsequent stimulation. This epigenetic reprogramming involves histone modifications that persist even after the initial TRAIL signal has dissipated.

The integration of TRAIL with other inflammatory signals is further evidenced by observed alterations in the expression of key pathway regulators, including SOCS proteins, MAPK phosphatases, and negative regulators of NF-κB signaling. These molecular interactions provide a mechanistic explanation for TRAIL's capacity to direct macrophage functional specialization within inflammatory microenvironments.

Understanding these pathway interactions has implications for therapeutic strategies targeting inflammatory disorders, particularly those where macrophage polarization plays a pathogenic role .

What statistical approaches are most appropriate for analyzing differential gene expression in TRAIL-treated human cells?

Analyzing differential gene expression in TRAIL-treated human cells requires sophisticated statistical approaches to account for biological variability while maintaining statistical power. Based on current methodological standards, the following approaches are recommended:

The most robust approach for RNA-seq data from TRAIL-treated cells involves a multi-step analytical pipeline. Initially, quality control of raw sequencing data should be performed, followed by alignment to the human reference genome and quantification of transcript abundance. For differential expression analysis specifically, the edgeR package has demonstrated particular utility in TRAIL-response studies due to its ability to handle the often-discrete nature of count data .

Proper normalization is critical, with Trimmed Mean of M-values (TMM) normalization being particularly effective for TRAIL studies as it accounts for library size differences while being robust to large expression changes in a subset of genes—a common occurrence in TRAIL-induced apoptotic or inflammatory responses .

For determining significance thresholds, current best practice involves:

  • Setting a minimum absolute log2-fold change threshold of ≥0.6 (approximately 1.5-fold change)

  • Applying a False Discovery Rate (FDR) cutoff of <0.05 using the Benjamini-Hochberg procedure

  • Requiring consistent expression changes across biological replicates (typically observed in ≥75% of samples)

Pathway enrichment analysis should follow differential expression identification, with platforms such as ClusterProfiler utilized to map genes to established pathways like those in the Kyoto Encyclopedia of Genes and Genomes (KEGG). This approach has successfully identified 238, 157, and 164 significantly enriched pathways in M0, M2a, and M2c macrophage groups, respectively, following TRAIL treatment .

For validation, statistical results from RNA-seq should be confirmed by qPCR for a subset of differentially expressed genes, using paired statistical tests appropriate for the experimental design.

How can researchers resolve data contradictions in TRAIL-related survival studies across different cancer types?

Resolving contradictory findings in TRAIL-related survival studies across cancer types requires a systematic methodological approach that addresses heterogeneity in study designs, patient populations, and analytical methods. Researchers should implement the following strategies:

By systematically applying these methodological approaches, researchers can distinguish genuine biological differences in TRAIL's role across cancer types from apparent contradictions arising from methodological inconsistencies .

What are the optimal protocols for isolating and culturing human macrophages for TRAIL research?

Successful TRAIL research on human macrophages depends on rigorous isolation and culture protocols that maintain cell viability and functional characteristics. The following methodological approach represents current best practices:

Isolation of Primary Human Monocytes:

  • Collect peripheral blood from healthy donors using approved protocols with appropriate ethical clearances.

  • Isolate peripheral blood mononuclear cells (PBMCs) using density gradient centrifugation over Ficoll-Paque.

  • Purify monocytes using either:

    • CD14-positive selection with magnetic beads (yields >95% purity)

    • Plastic adherence method (cost-effective but less pure at 85-90%)

Differentiation to Macrophages:

  • Seed monocytes at 0.5-1.0 × 10^6 cells/mL in RPMI-1640 supplemented with 10% heat-inactivated FBS, 2 mM L-glutamine, and antibiotics.

  • Add 50-100 ng/mL recombinant human M-CSF for differentiation to M0 macrophages.

  • Culture for 6-7 days in a humidified incubator at 37°C with 5% CO2.

  • Verify differentiation by morphological assessment and flow cytometry for CD68+CD14+ expression.

TRAIL Treatment Protocol:

  • After differentiation, treat macrophages with recombinant human TRAIL at 10-100 ng/mL (dose-dependent on experiment).

  • For receptor-specific experiments, use DR4-selective (4C7) or DR5-selective (5C7) variants.

  • Apply TRAIL treatment for 6-24 hours before subsequent polarization stimuli.

Polarization Conditions:

  • For M1 polarization: 20 ng/mL IFN-γ + 100 ng/mL LPS for 24 hours

  • For M2a polarization: 20 ng/mL IL-4 for 24 hours

  • For M2c polarization: 20 ng/mL IL-10 for 24 hours

Quality Control Measures:

  • Confirm macrophage viability (>90%) using Annexin V/PI staining

  • Verify polarization state by qPCR for phenotype-specific markers

  • Document donor-to-donor variability by maintaining donor identifiers

  • Include matched controls from the same donor for all experimental conditions

This standardized protocol facilitates reproducible investigation of TRAIL's effects on human macrophage biology while minimizing technical variability between studies .

How can researchers leverage "People Also Ask" data to identify emerging research questions about TRAIL Human?

"People Also Ask" (PAA) data represents a valuable but underutilized resource for identifying emerging research questions and knowledge gaps in TRAIL human research. This Google SERP feature appears in over 51.85% of searches, providing insights into what researchers and clinicians are actively questioning about TRAIL . By systematically analyzing this data, researchers can strengthen their investigations through the following methodological approach:

  • Data Collection Strategy:

    • Perform sequential searches using primary terms (e.g., "TRAIL human," "TRAIL cancer therapy," "TRAIL receptor expression")

    • Record all PAA questions that appear

    • Click through initial PAA questions to reveal additional nested questions

    • Document question frequency and relative positioning

  • Classification Framework:

    • Categorize questions into domain-specific clusters (e.g., basic biology, clinical applications, methodology)

    • Identify hierarchical relationships between questions

    • Distinguish between established knowledge questions and emerging research fronts

    • Flag questions indicating methodological uncertainties

  • Analytical Approach:

    • Track temporal changes in question patterns using longitudinal data collection

    • Compare PAA questions with formal literature gaps identified in review articles

    • Perform semantic analysis to identify conceptual connections between disparate questions

    • Cross-reference with citation metrics to identify high-interest/low-evidence areas

  • Research Application:

    • Develop research questions that address frequently asked but poorly answered questions

    • Incorporate anticipated questions into study design and reporting

    • Structure manuscript discussions to explicitly address common PAA questions

    • Create targeted knowledge translation materials addressing high-frequency questions

This systematic analysis of PAA data can reveal unrecognized knowledge gaps, identify emerging research interests before they appear in formal literature, and highlight areas where current evidence is insufficient to answer common questions. Tools like AlsoAsked can automate the collection process, while Hike SEO can provide visualization of question networks .

What are the ethical considerations specific to TRAIL-based clinical trials?

TRAIL-based clinical trials present unique ethical considerations that researchers must address systematically throughout study planning, implementation, and reporting. These considerations extend beyond standard research ethics requirements due to TRAIL's apoptosis-inducing properties and complex biological effects. The following framework outlines key ethical dimensions specific to TRAIL human research:

  • Risk-Benefit Assessment Considerations:

    • Evaluate liver toxicity risks based on preclinical data showing hepatocyte sensitivity to certain TRAIL formulations

    • Address potential for tumor-promoting effects in specific cancer contexts where TRAIL may activate pro-survival pathways

    • Consider differential risks for subjects with varying receptor expression profiles

    • Implement rigorous dose-escalation protocols with enhanced safety monitoring

  • Informed Consent Challenges:

    • Develop specialized consent processes explaining both anti-tumor and potential pro-tumor mechanisms

    • Communicate the investigational nature of TRAIL's effects on non-cancerous tissues

    • Address challenges in explaining complex biological mechanisms to research participants

    • Create validated assessment tools to verify participant comprehension of TRAIL-specific risks

  • Trial Design Ethics:

    • Justify control arm selection considering standard-of-care requirements versus placebo controls

    • Implement ethical adaptive design elements allowing rapid response to emerging safety signals

    • Establish clear stoppage rules specifically responsive to TRAIL's unique risk profile

    • Design appropriate crossover opportunities when ethically warranted

  • Special Population Considerations:

    • Address unique risk profiles for pediatric populations due to developmental differences in apoptotic pathways

    • Evaluate differential risks in patients with liver dysfunction or inflammatory conditions

    • Consider implications for pregnant women given TRAIL's role in maternal-fetal tolerance

  • Long-term Follow-up Requirements:

    • Establish monitoring protocols for delayed effects given TRAIL's influence on immune cell programming

    • Create biobanking frameworks with appropriate consent for future mechanism investigation

    • Develop participant communication plans for emerging knowledge about long-term effects

Researchers should document their approach to these considerations in trial protocols and ethics submissions, demonstrating thorough evaluation of TRAIL-specific ethical dimensions beyond standard research ethics frameworks .

How is single-cell analysis transforming our understanding of TRAIL receptor heterogeneity?

Single-cell technologies are revolutionizing our understanding of TRAIL receptor heterogeneity by revealing previously unappreciated complexity in receptor expression patterns and signaling dynamics. This methodological advancement addresses key limitations of bulk analysis techniques that obscured critical cell-to-cell variations in TRAIL responsiveness.

Recent single-cell RNA sequencing (scRNA-seq) studies have demonstrated that seemingly homogeneous cancer cell populations actually contain distinct subpopulations with dramatically different TRAIL receptor expression profiles. These analyses have identified rare cell subsets with unique DR4/DR5 expression ratios that would be undetectable in bulk analyses but may contribute disproportionately to treatment resistance.

Beyond mere expression levels, single-cell protein analysis via mass cytometry (CyTOF) has revealed complex patterns of receptor post-translational modifications that modulate TRAIL sensitivity. These studies have identified cell subpopulations with differential glycosylation patterns of DR4 and DR5, correlating with distinct apoptotic thresholds.

Single-cell trajectory analysis is further transforming our understanding of how TRAIL receptor expression evolves during cellular differentiation and disease progression. This approach has mapped how macrophage polarization states dynamically alter their TRAIL receptor expression profiles, providing temporal resolution that was impossible with previous methodologies.

Looking forward, integration of single-cell transcriptomics, proteomics, and functional assays promises to create comprehensive cellular atlases of TRAIL signaling networks. These integrated approaches will likely reveal new therapeutic opportunities by identifying targetable vulnerabilities in specific cellular subpopulations previously considered resistant to TRAIL-based interventions.

What are the implications of TRAIL's role in macrophage polarization for cancer immunotherapy?

The discovery that TRAIL promotes macrophage polarization toward specific phenotypes has significant implications for cancer immunotherapy development. Recent research has established that TRAIL can influence the functional state of macrophages beyond its classic role in inducing apoptosis, opening new therapeutic avenues .

TRAIL pre-treatment of human macrophages promotes polarization toward an enhanced M1-like phenotype, characterized by increased pro-inflammatory cytokine production and improved anti-tumor activity. RNA sequencing analyses have revealed that TRAIL exposure significantly upregulates pathways associated with antigen presentation, phagocytosis, and T-cell co-stimulation in macrophages subsequently exposed to inflammatory stimuli .

The therapeutic implications of this finding are substantial:

  • Enhanced CAR-Macrophage Therapy: TRAIL pre-conditioning of macrophages before CAR (Chimeric Antigen Receptor) engineering could potentially enhance their persistence and anti-tumor activity in solid tumors.

  • Combination Therapy Opportunities: TRAIL-based agents could be strategically combined with macrophage-targeting therapies such as CSF1R inhibitors to reshape the tumor microenvironment.

  • Biomarker Development: Macrophage polarization states in tumor biopsies following TRAIL therapy could serve as predictive biomarkers for treatment response.

  • Novel Delivery Approaches: Nanoparticle formulations that co-deliver TRAIL with macrophage-targeting agents could selectively modify tumor-associated macrophage function.

  • Overcoming Immunosuppression: TRAIL's ability to reprogram macrophages might help overcome the immunosuppressive tumor microenvironment that limits current immunotherapies.

Analysis of TCGA datasets has shown correlations between TRAIL expression and M1 macrophage markers in multiple cancer types, suggesting that this biological relationship exists in vivo and has prognostic significance . This emerging understanding challenges the traditional view of TRAIL as solely a pro-apoptotic molecule and positions it as a potential immunomodulator with applications beyond direct tumor killing.

How can trial methodology research enhance the design of studies investigating TRAIL-based therapies?

Adaptive Platform Trials offer significant advantages for TRAIL-based therapy evaluation. This methodology allows for multiple treatment comparisons within a single trial infrastructure, with the ability to add or remove treatment arms as evidence accumulates. For TRAIL research, this approach is particularly valuable given the multiple formulations, combination strategies, and potential biomarker-defined subgroups that require efficient evaluation .

Master Protocol Frameworks can streamline the investigation of TRAIL across multiple cancer types or indications. By employing basket, umbrella, or platform designs, researchers can simultaneously evaluate TRAIL's efficacy in different contexts while maintaining statistical rigor through appropriate multiple testing adjustments .

Novel Endpoint Selection is critical for TRAIL studies, where conventional endpoints may not capture the biological activity. Research in trial methodology has developed frameworks for selecting and validating surrogate endpoints, which is particularly relevant for TRAIL studies where early signals of immune modulation might precede clinical benefit .

Statistical Methods for Subgroup Identification are essential given the heterogeneous response to TRAIL. Methodological advances in adaptive enrichment designs allow for the prospective identification and enrichment of patient populations most likely to benefit from TRAIL-based therapies based on emerging data .

By implementing these methodological advances, TRAIL researchers can design more efficient trials that provide clearer answers about therapeutic potential while minimizing patient exposure to ineffective treatments. The MRC Network of Hubs for Trials Methodology Research (HTMR Network) represents a valuable resource for TRAIL investigators seeking to incorporate these innovations into their research programs .

Product Science Overview

Structure and Gene Location

TRAIL is a cytotoxic protein encoded by the TNFSF10 gene located on chromosome 3q26 in humans . It shares structural homology with other members of the TNF family, including Fas ligand (FasL) .

Mechanism of Action

TRAIL induces apoptosis by binding to two specific death receptors, DR4 (TRAIL-R1) and DR5 (TRAIL-R2) . These receptors contain death domains that initiate the apoptotic signaling cascade upon ligand binding . The apoptosis process is caspase-8-dependent, leading to the activation of downstream caspases that execute cell death .

Clinical Significance

TRAIL has garnered significant interest in cancer research due to its ability to selectively induce apoptosis in cancer cells. This property makes it a promising candidate for cancer therapy. Recombinant human TRAIL, such as dulanermin, has been evaluated in clinical trials for its efficacy and safety in treating various cancers, including non-small-cell lung cancer (NSCLC) .

Challenges and Future Directions

Despite its potential, the development of TRAIL-targeting therapies has faced challenges, including the emergence of resistance in some cancer cells and the need to avoid toxicity in normal cells . Ongoing research aims to overcome these hurdles by understanding the signaling network and developing novel drug platforms .

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.