TRAIL (114-281 a.a.) binds selectively to death receptors DR4 (TRAIL-R1) and DR5 (TRAIL-R2), initiating caspase-dependent apoptosis via the extrinsic pathway . Key steps include:
Receptor trimerization: TRAIL forms a homotrimer that clusters three receptor molecules .
DISC formation: Recruitment of FADD and procaspase-8 to receptor death domains, activating caspase-8 .
Caspase cascade: Executioner caspases (e.g., caspase-3) cleave cellular substrates, leading to DNA fragmentation and apoptosis .
Selectivity: Preferential apoptosis in cancer cells due to higher DR4/DR5 expression and reduced decoy receptor (DcR1, DcR2, OPG) activity .
Cell Line | Sensitivity | Apoptosis Induction (Concentration) | Reference |
---|---|---|---|
U937 | High | 1 µg/mL within 5 hours | |
NCI-H460 | High | IC50 <20 nM | |
MDA.MB.231 | High | 27.71 pM (sfTRAIL) | |
A549 | Resistant | No effect at 2 ng/mL |
Resistance mechanisms: Downregulation of DR4/DR5, overexpression of anti-apoptotic proteins (e.g., c-FLIP) .
Anticancer agent: Tested in Phase I/II trials for non-small cell lung cancer, colorectal cancer, and lymphomas .
Drug conjugates: PEG-TRAIL-vcMMAE shows enhanced tumor targeting and reduced toxicity in xenograft models .
Bystander effect: Secreted soluble TRAIL (sfTRAIL) induces apoptosis in neighboring cancer cells .
TRAIL Human (114-281 a.a.) refers to the extracellular domain of human TRAIL protein that mediates activation of the extrinsic apoptosis pathway. TRAIL is expressed as a homotrimeric type II transmembrane or soluble protein that binds to and activates its death receptors DR4 and DR5 . The binding induces receptor trimerization, which is essential for initiating the apoptotic signaling pathway . The extracellular domain (amino acids 114-281) retains the binding capacity to these receptors and can be produced recombinantly for research purposes.
The interaction between TRAIL and its receptors creates a cascade where the cytoplasmic domains of DR4 and DR5 serve as docking sites for adapter proteins that ultimately activate the caspase cascade leading to controlled cell death . Notably, TRAIL also interacts with three decoy receptors (DcR1, DcR2, and OPG), which can modulate its apoptotic activity .
TRAIL exhibits remarkable tumor specificity, selectively inducing apoptosis in malignant cells while generally sparing normal tissues . This selective activity is attributed to several factors:
Differential receptor expression: Cancer cells often upregulate death receptors DR4 and DR5 compared to normal cells
Decoy receptor distribution: Normal cells may express higher levels of decoy receptors (DcR1, DcR2, OPG) that bind TRAIL without triggering apoptosis
Intracellular anti-apoptotic mechanisms: Cancer cells frequently have alterations in apoptotic machinery that paradoxically make them more susceptible to death receptor activation
Research has demonstrated that this selective toxicity profile makes TRAIL an attractive candidate for cancer therapy compared to other TNF family members like FasL and TNF-α, which cause significant systemic toxicity when administered .
Several expression systems have been successfully employed to produce recombinant TRAIL (114-281 a.a.), each with specific advantages:
Human recombinant TRAIL (hrTRAIL) and metabolically labeled 15N-rhTRAIL have been successfully expressed in E. coli and purified using established protocols . For applications requiring more native-like protein structure, CHO cell expression systems have been employed, particularly when producing fusion proteins like TRAIL-Trimer, which contains disulfide bond-linked homotrimers .
When selecting an expression system, researchers should consider whether post-translational modifications are critical for their specific experimental design and whether protein yield or structural authenticity is the priority.
A significant challenge with native TRAIL (114-281 a.a.) is its relatively rapid clearance in vivo, which limits its therapeutic potential. Several methodological approaches have been developed to address this limitation:
Trimer-Tag fusion: In-frame fusion of human C-propeptide of α1(I) collagen (Trimer-Tag) to the C-terminus of mature human TRAIL creates a disulfide bond-linked homotrimer with significantly improved pharmacokinetic profiles
Receptor-selective variants: Engineering TRAIL variants with preferential binding to either DR4 or DR5 based on the cancer type being targeted (some cancers preferentially signal through one receptor over the other)
The TRAIL-Trimer approach has demonstrated particular promise, retaining similar bioactivity and receptor binding kinetics as native TRAIL in vitro while showing 4–5 orders of magnitude superior activity compared to dimeric TRAIL-Fc constructs . Most importantly, TRAIL-Trimer manifests more favorable pharmacokinetic and antitumor pharmacodynamic profiles in vivo than native TRAIL, with direct evidence that antitumor efficacy correlates with systemic drug exposure .
Accurate quantification of TRAIL is essential for consistent experimental results. Several complementary analytical methods are available:
LC-MS/MS analysis: Antibody-free liquid chromatography-tandem mass spectrometry provides highly specific protein analysis of TRAIL with potential for absolute quantification
Bioactivity assays: Functional testing using TRAIL-sensitive cell lines to determine biological activity rather than just protein concentration
Receptor binding kinetics: Surface plasmon resonance (SPR) or similar techniques to measure binding affinity to DR4/DR5 receptors
For the most comprehensive characterization, researchers should employ multiple orthogonal methods. A particular advantage of LC-MS/MS approaches is the ability to perform antibody-free analysis, which can be especially valuable when antibody availability or specificity is limiting .
Understanding receptor selectivity is crucial when studying TRAIL mechanisms. While it was initially assumed that TRAIL-R2 (DR5) played a dominant role in initiating apoptosis, more recent evidence suggests that receptor preference varies by cancer type . Some methodological approaches to evaluate receptor selectivity include:
Comparative receptor expression analysis: Quantify DR4, DR5, and decoy receptor levels on target cells
Receptor-specific blocking antibodies: Selectively inhibit either DR4 or DR5 to determine their relative contribution to apoptosis
Receptor knockdown/knockout: Use RNAi or CRISPR-based approaches to specifically reduce receptor expression
Receptor-selective TRAIL variants: Compare the activity of TRAIL mutants engineered for preferential binding to either DR4 or DR5
Research has demonstrated that some cancer types, such as chronic lymphocytic leukemia (CLL), predominantly signal to cell death via TRAIL-R1 (DR4), challenging the previous assumption of TRAIL-R2 dominance . Importantly, the relationship between receptor expression levels and response to TRAIL-R1 or TRAIL-R2 activating compounds is not straightforward and requires careful experimental design to elucidate .
Despite TRAIL's promise as a cancer therapeutic, many tumors exhibit intrinsic or acquired resistance. Several key resistance mechanisms have been identified:
Inhibitor of Apoptosis Proteins (IAPs): Many cancer types, including pancreatic carcinoma, overexpress IAPs such as X-linked inhibitor of apoptosis (XIAP), which prevents apoptosis by binding to and inhibiting activated caspase-3 and caspase-9
Decoy receptor expression: Increased expression of non-apoptotic TRAIL receptors (DcR1, DcR2, OPG) can sequester TRAIL without triggering cell death
Death receptor downregulation: Reduced expression or internalization of DR4/DR5 receptors limits TRAIL sensitivity
Defects in apoptotic signaling: Alterations in downstream components of the apoptotic machinery can prevent effective signal transduction
XIAP represents a particularly important resistance factor as it acts at the core of the apoptotic machinery, blocking cell death at the effector phase . This makes therapeutic modulation of XIAP a potentially valuable strategy to overcome TRAIL resistance in cancer treatment protocols .
Several methodological strategies have shown promise in addressing TRAIL resistance:
Combination treatments: Many standard chemotherapeutics can sensitize resistant cells to TRAIL through various mechanisms, including upregulation of death receptors or downregulation of anti-apoptotic proteins
IAP antagonists: Small molecule inhibitors that target XIAP and other IAPs can restore TRAIL sensitivity in resistant cancer cells by removing the block at the effector phase of apoptosis
Epigenetic modifiers: HDAC inhibitors and DNA methyltransferase inhibitors can increase death receptor expression in some cancer types
Improved TRAIL formulations: Novel delivery systems or engineered TRAIL variants with enhanced stability and receptor binding properties
The selection of sensitizing agents should be guided by the specific resistance mechanism present in the experimental model. For instance, IAP antagonists would be most appropriate when dealing with XIAP-mediated resistance, whereas epigenetic modifiers might be more suitable for cases with epigenetically silenced death receptors.
Despite encouraging preclinical data, TRAIL-based therapies have shown modest efficacy in clinical trials . Several factors may explain this disconnect:
Pharmacokinetic limitations: Evidence suggests that the clinical failures may be due to rapid systemic clearance of native TRAIL, limiting effective drug exposure at tumor sites
Receptor agonist potency: Dimeric agonist monoclonal antibodies targeting DR4/DR5 have shown poor apoptosis-inducing potency despite their long serum half-lives
Tumor heterogeneity: Variability in receptor expression and resistance mechanisms within and between tumors may limit consistent responses
Decoy receptor interference: The presence of decoy receptors in the tumor microenvironment may sequester therapeutic TRAIL
Research has provided direct evidence that in vivo antitumor efficacy of TRAIL correlates with systemic drug exposure, suggesting that approaches to extend half-life while maintaining or enhancing receptor activation could improve clinical outcomes .
When designing clinical studies involving TRAIL (114-281 a.a.), researchers must navigate complex regulatory requirements for human subjects research:
Clinical trial definition: Studies must determine whether they meet the NIH definition of a clinical trial, which encompasses "a research study in which one or more human subjects are prospectively assigned to one or more interventions to evaluate the effects of those interventions on health-related biomedical or behavioral outcomes"
Regulatory compliance: Research must follow institutional and national guidelines for human subjects research, including IRB approval and informed consent procedures
Trial registration: Clinical trials involving TRAIL must be registered on ClinicalTrials.gov with appropriate documentation of protocol and results reporting
Does the study involve human participants?
Are the participants prospectively assigned to an intervention?
Is the study designed to evaluate the effect of the intervention on the participants?
Is the effect being evaluated a health-related biomedical or behavioral outcome?
LC-MS/MS offers powerful capabilities for TRAIL analysis without relying on antibodies, which can be particularly valuable when antibody availability, specificity, or cross-reactivity is a concern:
Sample preparation: For complex matrices like serum or tissue lysates, protein precipitation, immunodepletion of abundant proteins, or solid-phase extraction may be necessary
Peptide selection: Identify unique peptides (signature peptides) from TRAIL that can serve as quantitative surrogates for the intact protein
Isotope dilution: Use of metabolically labeled 15N-rhTRAIL as an internal standard enables accurate quantification
Sensitivity enhancement: Techniques such as multiple reaction monitoring (MRM) can improve detection limits for TRAIL peptides in complex backgrounds
LC-MS/MS approaches have been successfully applied to quantify other proteins in human serum with lower limits of quantification (LLOQ) in the nanomolar range, suggesting this methodology could be adapted for TRAIL quantification in clinical samples .
Identifying predictive biomarkers for TRAIL sensitivity remains an active area of research. Several potential markers have emerged:
Death receptor expression: While not a perfect predictor, DR4/DR5 expression levels provide baseline information about potential TRAIL responsiveness
Decoy receptor levels: The ratio of death receptors to decoy receptors may offer better predictive value than absolute expression of either alone
XIAP and other IAP expression: Higher levels typically correlate with resistance
C-reactive protein (CRP): Some studies have investigated relationships between inflammatory markers like CRP and therapeutic responses, though direct links to TRAIL sensitivity require further investigation
Researchers should consider using a panel of biomarkers rather than relying on a single predictor, given the complex and multifactorial nature of TRAIL response mechanisms in cancer cells.
An exciting frontier in TRAIL research involves its intersection with cancer immunotherapy:
TRAIL-immune checkpoint interactions: Emerging evidence indicates that the TRAIL pathway may interact with immune checkpoint proteins, including programmed death-ligand 1 (PD-L1), potentially modulating PD-L1-based tumor immunotherapies
TRAIL in immune surveillance: Beyond direct apoptosis induction, TRAIL expressed by immune cells plays a critical role in tumor surveillance
Combination approaches: TRAIL-based therapies may complement immune checkpoint inhibitors by targeting cancer cells through orthogonal mechanisms
These interactions suggest potential synergies between TRAIL-targeted therapies and immune checkpoint blockade strategies, opening new avenues for combination treatments that simultaneously activate apoptotic pathways and reinvigorate anti-tumor immune responses .
Several innovative approaches are being explored to overcome the pharmacokinetic limitations of native TRAIL:
Engineered fusion proteins: The TRAIL-Trimer approach using C-propeptide of α1(I) collagen has already demonstrated improved pharmacokinetics while maintaining receptor binding efficiency
Nanoparticle delivery systems: Encapsulation or surface presentation of TRAIL on various nanoparticles may improve stability and tumor targeting
Cell-based delivery: Engineered cells expressing TRAIL could provide sustained local delivery in the tumor microenvironment
Gene therapy approaches: Viral vectors encoding TRAIL could enable long-term expression within tumors
The promising results with TRAIL-Trimer, which showed significantly improved pharmacokinetic and antitumor pharmacodynamic profiles compared to native TRAIL, provide proof-of-concept that addressing the rapid clearance issue can enhance therapeutic efficacy .
Consistency in TRAIL preparations is crucial for reproducible research. Several approaches can minimize variability:
Standardized expression protocols: Maintain consistent culture conditions, induction parameters, and harvest timing
Quality control metrics: Implement multiple orthogonal QC assays including:
SDS-PAGE for purity assessment
Size exclusion chromatography to confirm trimeric structure
Bioactivity assays on reference cell lines
Mass spectrometry for identity confirmation
Reference standards: Maintain well-characterized internal reference standards to calibrate new batches
Storage optimization: Investigate and standardize buffer compositions and storage conditions that maximize stability
Researchers should document batch characteristics comprehensively and include appropriate controls when comparing results across different TRAIL preparations.
Differentiating TRAIL-specific apoptosis from other cell death mechanisms requires careful experimental design:
Blocking controls: Use TRAIL-neutralizing antibodies or soluble decoy receptors to confirm specificity
Receptor dependence: Employ receptor-specific blocking antibodies or receptor knockdown to verify the involvement of DR4/DR5
Apoptosis markers: Assess hallmarks of apoptosis including caspase activation, PARP cleavage, and phosphatidylserine externalization
Caspase inhibition: Determine whether pan-caspase inhibitors (e.g., z-VAD-fmk) prevent the observed cell death
Alternative death pathways: Evaluate markers of necroptosis, pyroptosis, or other death mechanisms to rule out non-apoptotic death
TRAIL is a type II transmembrane protein that can be cleaved to form a soluble protein. The human TRAIL gene is located on chromosome 3q26. The recombinant form of TRAIL, specifically the fragment spanning amino acids 114 to 281, is expressed in Escherichia coli and is often used in research due to its high purity and biological activity .
TRAIL induces apoptosis by binding to its death receptors, DR4 (TRAIL-R1) and DR5 (TRAIL-R2). Upon binding, these receptors undergo trimerization and recruit adaptor proteins such as FADD (Fas-Associated Death Domain), which in turn activate caspase-8. This activation triggers a cascade of downstream caspases, ultimately leading to cell death .
Interestingly, TRAIL can also bind to decoy receptors DcR1 (TRAIL-R3) and DcR2 (TRAIL-R4), which do not induce apoptosis. These decoy receptors can inhibit TRAIL-induced apoptosis by competing with death receptors for TRAIL binding .
TRAIL is unique among the TNF superfamily members due to its ability to selectively induce apoptosis in cancer cells while sparing normal cells. This selective cytotoxicity makes TRAIL a promising candidate for cancer therapy. Studies have shown that recombinant human TRAIL can induce apoptosis in various cancer cell lines, including those resistant to conventional therapies .
Recombinant human TRAIL (114-281 a.a.) is widely used in research to study apoptosis mechanisms and to develop potential cancer therapies. Its high purity and biological activity make it suitable for various applications, including: