TNFRSF11A is essential for osteoclastogenesis and bone remodeling. Its partial recombinant forms are used to study:
RANKL Binding:
Signaling Pathways:
TNFSF11/TNFRSF11A Axis in Asthma:
Parameter | Specification | Sources |
---|---|---|
Endotoxin | <0.01 EU/μg (LAL test) . | |
Storage | -80°C for GST-tagged proteins; -20°C or 4°C for Fc-tagged variants . |
The Recombinant Human Tumor necrosis factor receptor superfamily member 11A protein (TNFRSF11A) is a partial-length protein encompassing amino acids 29-202 of the human TNFRSF11A. This protein was produced through the expression of a DNA fragment encoding this region in E. coli. Rigorous validation via SDS-PAGE and HPLC analyses confirmed a purity exceeding 90%. Furthermore, its biological activity was verified through its interaction with sRANK Ligand. The endotoxin level of this TNFRSF11A is below 1.0 EU/µg as determined by the LAL method. This recombinant TNFRSF11A protein is readily available, eliminating any waiting time for preparation. Custom tagged proteins are also offered upon request. This recombinant TNFRSF11A protein can be utilized for the development of specific antibodies or in research focusing on TNFRSF11A-associated cancers.
TNFRSF11A, also known as RANK, serves as the receptor for RANKL. The interaction between RANK and RANKL plays a critical role in a multitude of biological processes, including bone resorption and remodeling, bone marrow formation, development of the thymus and lymph nodes, intestinal immunity, inflammatory responses within the skin and central nervous system (CNS), tumorigenesis, and metastasis. Mutations within RANK or RANKL have been linked to the pathogenesis of bone diseases and certain types of cancer.
This receptor for TNFSF11/RANKL/TRANCE/OPGL is essential for RANKL-mediated osteoclastogenesis. It plays a significant role in regulating interactions between T-cells and dendritic cells.
TNFRSF11A (TNF receptor superfamily member 11a), commonly known as RANK (Receptor Activator of Nuclear Factor κB), is a type I transmembrane protein belonging to the tumor necrosis factor receptor family. The full-length human RANK contains a 184 amino acid extracellular domain and a 383 amino acid cytoplasmic domain, with two potential N-linked glycosylation sites in its extracellular region . TNFRSF11A serves several critical physiological functions, primarily as an essential mediator for osteoclast differentiation and lymph node development .
The protein functions as a receptor that interacts with various TRAF (TNF Receptor Associated Factor) family proteins, through which it induces the activation of key signaling pathways, particularly NF-kappa B and MAPK8/JNK . These signaling cascades are crucial for several biological processes including:
Regulation of interactions between T cells and dendritic cells in the immune system
Mediation of osteoclast formation and activation in bone remodeling
Development and organization of lymph nodes
The protein is widely expressed in human tissues, with highest expression observed in skeletal muscle, thymus, liver, colon, small intestine, and adrenal gland .
Research with neutrophils has demonstrated that synovial fluid from rheumatoid arthritis (RA) patients can induce the expression of RANK, suggesting environmental and inflammatory factors may regulate its expression . This induction appears to be part of a broader regulatory network involving RANK, RANKL (RANK ligand), and OPG (osteoprotegerin), which collectively control osteoclast differentiation and activity.
TNFRSF11A expression is also subject to alternative splicing, with several splice variants identified in breast cancer cell lines, including TNFRSF11A_delta 9 (RANK-a), TNFRSF11A_delta 8,9 (RANK-b), and TNFRSF11A_delta 7,8,9 (RANK-c) . These alternative splice variants may have distinct functional properties and could be regulated differently across tissue types.
TNFRSF11A activates multiple signaling pathways critical for cellular differentiation and function. The primary signaling mechanism involves the recruitment of TRAF family proteins to its cytoplasmic domain, which subsequently activates downstream pathways . The major signaling pathways include:
NF-κB (Nuclear Factor-kappa B) Pathway: TNFRSF11A stimulation leads to activation of both canonical and non-canonical NF-κB pathways, resulting in the translocation of transcription factors to the nucleus and expression of target genes involved in osteoclast differentiation, survival, and function .
MAPK (Mitogen-Activated Protein Kinase) Pathways: TNFRSF11A activates multiple MAPK pathways, particularly the c-Jun N-terminal kinase (JNK) pathway, which regulates cellular responses to stress, inflammatory cytokines, and growth factors .
PI3K/Akt Pathway: This pathway contributes to osteoclast survival and cytoskeletal rearrangements necessary for bone resorption.
NFATc1 (Nuclear Factor of Activated T cells c1) Pathway: TNFRSF11A signaling ultimately leads to the activation and amplification of NFATc1, a master transcription factor for osteoclastogenesis.
These signaling cascades work in concert to mediate various biological effects, including enhancement of T cell growth, dendritic cell function, induction of osteoclastogenesis, and lymph node organogenesis .
TNFRSF11A is located on the long arm of chromosome 18 at position 18q21.33 . The gene consists of 12 exons that encode the full protein . Within the genome, TNFRSF11A has been mapped between markers D18S383 and D18S51, specifically in the region of D18S60 between WI-9823 and SGC33768 .
The genomic sequence is represented by the reference sequence NC_000018.10 (62325310..62391288) on chromosome 18 . The full-length human TNFRSF11A cDNA encodes a type I transmembrane protein of 616 amino acids . The gene contains several polymorphic sites that have been identified, including variants in the 5'-UTR (-1G/A, 30T/C), coding regions (575C/T, 421C/T), and intronic regions (IVS6+79A/G) .
Understanding the genomic organization of TNFRSF11A is particularly important for mutation analysis in associated disorders and for designing gene-targeting strategies in research applications.
The study of TNFRSF11A-mediated signaling in osteoclast differentiation requires a multi-faceted approach combining molecular, cellular, and biochemical techniques. Based on current research practices, the following methodological approaches are recommended:
Cell Culture Systems:
RAW 264.7 mouse monocyte/macrophage cell line is an established model for studying RANK-mediated osteoclastogenesis
Primary bone marrow macrophages (BMMs) isolated from mice provide a physiologically relevant system
Human peripheral blood mononuclear cells (PBMCs) can be used for translational studies
Osteoclast Differentiation Assays:
Treat RAW 264.7 cells with recombinant RANKL and M-CSF to induce osteoclast differentiation
Use TRAP (tartrate-resistant acid phosphatase) staining to identify mature osteoclasts
Measure bone resorption activity using synthetic bone substrates or bone slices
Signaling Pathway Analysis:
Western blotting to detect phosphorylation of key signaling molecules (NF-κB, MAPK, Akt)
Immunoprecipitation to study protein-protein interactions between RANK and TRAF adaptor proteins
Luciferase reporter assays to quantify NF-κB activation
Chromatin immunoprecipitation (ChIP) to analyze transcription factor binding to target gene promoters
Functional Manipulation:
Expression Analysis:
These methodologies can be combined to comprehensively study the role of TNFRSF11A in osteoclast differentiation, from receptor expression and activation to downstream signaling events and functional outcomes.
Mutations in the signal peptide region of TNFRSF11A have profound effects on protein function and are directly linked to several bone pathologies. Research has identified specific tandem duplications in exon 1 of TNFRSF11A that affect the signal peptide of RANK and cause familial expansile osteolysis (FEO) and Paget's disease of bone (PDB) .
Molecular Mechanisms of Signal Peptide Mutations:
Altered Protein Processing: The identified mutations (84dup18 in FEO and 75dup27 in PDB) lead to perturbations in RANK expression levels and prevent normal cleavage of the signal peptide . This disruption in protein processing affects the proper localization and membrane insertion of the receptor.
Constitutive Activation: Both mutations cause an increase in RANK-mediated NF-κB signaling in vitro, consistent with an activating mutation . This leads to heightened downstream signaling even in the absence of RANKL stimulation.
Enhanced Osteoclastogenesis: The constitutive activation of RANK signaling results in increased osteoclast formation and activity, leading to excessive bone resorption and the characteristic osteolytic lesions observed in affected individuals.
Experimental Approaches to Study These Mutations:
Site-directed mutagenesis to introduce the duplications into wild-type TNFRSF11A cDNA
Expression of wild-type and mutant constructs in cell lines to assess differences in:
Protein localization (immunofluorescence microscopy)
Signal peptide cleavage (Western blot with antibodies against different protein domains)
NF-κB activation (luciferase reporter assays)
Osteoclast differentiation capacity (when expressed in osteoclast precursors)
Generation of knock-in mouse models carrying the human mutations to study the phenotypic consequences at the organismal level
Understanding the molecular consequences of these signal peptide mutations provides insights into both the normal function of TNFRSF11A and the pathogenesis of associated bone disorders, potentially identifying targets for therapeutic intervention.
TNFRSF11A undergoes alternative splicing, generating several variants with potentially distinct functional properties. Research has identified multiple splice variants in breast cancer cell lines, including TNFRSF11A_delta 9 (RANK-a), TNFRSF11A_delta 8,9 (RANK-b), and TNFRSF11A_delta 7,8,9 (RANK-c) . These splice variants may have significant implications in pathological conditions, particularly in cancer.
Methodological Approaches to Study Splice Variants:
Identification and Characterization:
RT-PCR with primers spanning multiple exons to detect alternative splicing events
Agarose gel electrophoresis to visualize different splice variants
Cloning and sequencing to confirm the identity of splice variants
qPCR with variant-specific primers to quantify relative expression levels
Expression Analysis in Pathological Conditions:
Comparative expression analysis in normal versus cancer cell lines
Assessment of splice variant expression in relation to clinical parameters such as tumor grade and proliferation marker Ki-67
Analysis in formaldehyde-fixed paraffin-embedded (FFPE) tissue samples from patients
Research Findings on TNFRSF11A Splice Variants in Cancer:
Research has shown that TNFRSF11A_delta 7,8,9 (RANK-c) expression varies across different breast cancer cell lines, with distinct expression patterns compared to the non-tumorigenic epithelial cell line MCF10A . This suggests a potential role for this splice variant in cancer development or progression.
In a panel of invasive ductal breast carcinoma samples, the relative expression levels of both wild-type RANK and the RANK-c variant showed relationships with tumor histological grade . This indicates that alterations in TNFRSF11A splicing may be associated with cancer aggressiveness.
Further investigation of these splice variants is warranted to understand:
Their structural and functional differences from full-length TNFRSF11A
Their impact on RANK-RANKL signaling and downstream pathways
Their potential as biomarkers or therapeutic targets in cancer
Their roles in other pathological conditions beyond breast cancer
Developing therapeutic approaches targeting TNFRSF11A requires understanding its structure-function relationships and its role in pathological conditions. Several strategies can be employed:
1. Antibody-Based Approaches:
Neutralizing antibodies against RANK to block interaction with RANKL
Agonistic antibodies (like AF683) that could potentially be modified to have partial agonist activity for fine-tuning RANK signaling
Engineering antibodies to target specific epitopes to modulate particular downstream pathways
2. Recombinant Protein Therapeutics:
Soluble RANK as a decoy receptor to compete with membrane-bound RANK for RANKL binding
Modified versions of RANK with enhanced binding affinity or altered signaling properties
Fusion proteins combining the RANKL-binding domain of RANK with other functional domains
3. Small Molecule Inhibitors:
Compounds targeting the intracellular signaling mediators of RANK (e.g., TRAF proteins)
Molecules that modulate specific downstream pathways like NF-κB or MAPK
Signal peptide-targeting compounds that could potentially correct the defects caused by the FEO and PDB mutations
4. Gene Therapy Approaches:
CRISPR-Cas9 gene editing to correct pathogenic mutations in TNFRSF11A
RNA interference to modulate expression of TNFRSF11A or its splice variants
Antisense oligonucleotides to alter splicing patterns in conditions where aberrant splicing contributes to pathology
5. Precision Medicine Strategies:
Genotyping patients for TNFRSF11A mutations or polymorphisms to guide therapeutic decisions
Monitoring expression of TNFRSF11A and its splice variants as biomarkers for disease progression or treatment response
Combining TNFRSF11A-targeted therapies with other agents for synergistic effects
Experimental Considerations:
In vitro osteoclast differentiation assays to assess efficacy
Bone resorption assays to determine functional impact
Animal models of FEO, PDB, or osteoporosis for in vivo validation
Safety assessment focusing on potential effects on immune function given RANK's role in dendritic cells and T cells
Development of these therapeutic approaches requires careful consideration of TNFRSF11A's physiological roles to minimize unwanted effects while maximizing therapeutic benefits in targeted bone disorders.
Producing high-quality recombinant TNFRSF11A protein is essential for various research applications including structural studies, binding assays, and functional characterization. Based on established methodologies in protein biochemistry and the specific characteristics of TNFRSF11A, the following protocol considerations are recommended:
Expression System Selection:
Mammalian Expression Systems (Preferred for full-length or extracellular domain):
HEK293 or CHO cells are optimal for obtaining properly folded and glycosylated TNFRSF11A
These systems ensure correct formation of disulfide bonds in the cysteine-rich extracellular domain
Transient transfection or stable cell line generation using pCDNA3.1 or similar vectors with appropriate tags (His, Fc, or FLAG)
Insect Cell Expression Systems:
Baculovirus-infected Sf9 or High Five cells represent an alternative for higher yield
Suitable for the extracellular domain with proper post-translational modifications
E. coli Expression (Limited application):
Only recommended for specific protein fragments that do not require glycosylation
May be useful for cytoplasmic domain expression for structural studies
Requires refolding protocols when expressing disulfide-containing domains
Construct Design Considerations:
Express the extracellular domain (amino acids 1-184) for receptor-ligand interaction studies
Include the signal peptide sequence for proper processing in mammalian systems
Consider a C-terminal tag rather than N-terminal to avoid interference with signal peptide processing
For structural studies, remove flexible regions that might hinder crystallization
Purification Strategy:
Affinity Chromatography:
Immobilized metal affinity chromatography (IMAC) for His-tagged proteins
Protein A/G chromatography for Fc-fusion proteins
Anti-FLAG affinity chromatography for FLAG-tagged constructs
Further Purification Steps:
Ion exchange chromatography based on TNFRSF11A's isoelectric point
Size exclusion chromatography to remove aggregates and ensure monodispersity
Consider including low concentrations of reducing agents to prevent non-specific disulfide formation
Quality Control:
SDS-PAGE and Western blot to confirm identity and purity
Mass spectrometry to verify molecular weight and post-translational modifications
Circular dichroism to assess secondary structure
Thermal shift assays to evaluate protein stability
Functional Validation:
RANKL binding assays using surface plasmon resonance or bio-layer interferometry
Cell-based assays measuring NF-κB activation in response to the purified protein
Osteoclast differentiation assays to confirm biological activity
These methodological considerations should be adapted based on the specific research questions and downstream applications of the recombinant TNFRSF11A protein.
Analysis of TNFRSF11A polymorphisms and their association with bone disorders requires a systematic approach combining genetic, statistical, and functional methods. The following comprehensive methodology is recommended:
1. Polymorphism Identification and Genotyping:
Next-Generation Sequencing (NGS): Targeted sequencing of the TNFRSF11A gene, including exons, intron-exon boundaries, and regulatory regions
PCR-RFLP (Restriction Fragment Length Polymorphism): For known polymorphisms like IVS6+79A/G
TaqMan SNP Genotyping Assays: For high-throughput analysis of specific SNPs
Sanger Sequencing: For confirmation of novel variants
PAGE (Polyacrylamide Gel Electrophoresis): Particularly useful for detecting insertions/duplications that alter fragment size, as demonstrated in the FEO studies
2. Study Design Considerations:
Case-Control Studies: Compare genotype frequencies between patients with bone disorders and healthy controls
Family-Based Association Studies: Analyze inheritance patterns within families, as demonstrated in the FEO and PDB families
Meta-Analysis: Combine results from multiple studies to increase statistical power
Sample Size Calculation: Ensure adequate power to detect associations based on expected effect sizes
3. Statistical Analysis Approaches:
Hardy-Weinberg Equilibrium Testing: To check for genotyping errors or selection bias
Chi-Square or Fisher's Exact Test: For comparing genotype and allele frequencies between cases and controls
Logistic Regression: To calculate odds ratios and adjust for potential confounding factors
Haplotype Analysis: To evaluate combinations of polymorphisms that may act together
Linkage Disequilibrium Mapping: To identify regions of TNFRSF11A associated with disease
4. Functional Validation of Associated Polymorphisms:
Luciferase Reporter Assays: To assess the impact of promoter variants on gene expression
EMSA (Electrophoretic Mobility Shift Assay): To evaluate effects on transcription factor binding
RNA Stability Assays: For variants in untranslated regions
Cell-Based Functional Assays: To determine effects on NF-κB signaling, as performed for the FEO mutations
Animal Models: Generation of knock-in mice carrying human polymorphisms for in vivo functional studies
5. Clinical Correlation:
Genotype-Phenotype Correlation Analysis: Relating specific polymorphisms to clinical features
Bone Mineral Density Measurements: DXA scans to correlate genotypes with quantitative traits
Biomarker Analysis: Measuring bone turnover markers in relation to genotypes
Imaging Studies: Advanced imaging to characterize bone structure in different genotype groups
This polymorphism was analyzed in 67 Northern-Irish patients with sporadic Paget's disease of bone and 63 age-matched controls . While the study did not find a significant association, the methodology demonstrates the principle of case-control design for TNFRSF11A polymorphism analysis.
By integrating these approaches, researchers can comprehensively evaluate the contribution of TNFRSF11A genetic variation to bone disorders, potentially identifying risk factors and novel therapeutic targets.
Selecting appropriate experimental models is crucial for investigating TNFRSF11A function in vivo. Each model system offers distinct advantages and limitations for studying different aspects of RANK biology. The following models are recommended based on research objectives:
1. Mouse Models:
Global Knockout Models:
TNFRSF11A-/- mice display severe osteopetrosis due to the absence of osteoclasts, along with defects in lymph node development and mammary gland formation
These models are valuable for understanding the fundamental physiological roles of RANK but may have limitations for studying acquired bone disorders due to developmental defects
Conditional Knockout Models:
Cell-specific deletion using Cre-loxP system (e.g., cathepsin K-Cre for osteoclast-specific deletion)
Temporal control using inducible systems (e.g., tamoxifen-inducible CreERT2)
These models allow for the study of TNFRSF11A function in specific cell types and developmental stages, avoiding the confounding effects of developmental abnormalities
Knock-in Models:
Transgenic Overexpression Models:
Overexpression of wild-type or constitutively active TNFRSF11A variants
Useful for studying gain-of-function effects and potential roles in cancer models
2. Rat Models:
Larger size facilitates certain procedures such as bone surgeries and imaging
Similar bone physiology to humans
Valuable for pharmacological studies due to more predictable drug metabolism
3. Zebrafish Models:
Rapid development and transparent embryos allow for real-time visualization of skeletal development
Amenable to high-throughput screening of chemical compounds targeting RANK signaling
CRISPR-Cas9 gene editing can be used to generate loss-of-function or knock-in mutations
4. Ex Vivo Bone Organ Cultures:
Calvarial organ cultures from neonatal mice
Maintain the complex cellular interactions within bone tissue
Suitable for short-term studies of RANK-mediated osteoclastogenesis and bone resorption
5. Humanized Mouse Models:
Immunodeficient mice reconstituted with human immune cells
Valuable for studying human-specific aspects of RANK function in immune regulation
Potential for testing human-specific therapeutics
Experimental Applications:
Bone Phenotyping:
Micro-CT analysis for bone microarchitecture
Histomorphometry for cellular parameters (osteoclast number, surface, etc.)
Biomechanical testing for functional consequences
Immune Function Assessment:
Flow cytometry analysis of dendritic cell and T cell populations
Lymph node development and organization
Immune responses to various challenges
Disease Modeling:
Inflammatory bone loss (e.g., arthritis models)
Metastatic bone disease models
Age-related bone loss
Therapeutic Testing:
Evaluation of RANK-targeted biologics
Assessment of small molecule inhibitors of downstream pathways
Testing of gene therapy approaches
Selection of the appropriate model should be guided by the specific research question, focusing on models that best recapitulate the human condition being studied while considering ethical principles and the 3Rs (Replacement, Reduction, Refinement) in animal research.
TNFRSF11A mutations play a central role in the pathogenesis of several bone disorders through dysregulation of osteoclast formation and function. Understanding these mechanisms has significant clinical implications for diagnosis, prognosis, and treatment.
Pathogenic Mechanisms:
Gain-of-Function Mutations:
Tandem duplications in the signal peptide region (84dup18 in FEO and 75dup27 in PDB) lead to constitutive activation of RANK-mediated NF-κB signaling
These mutations prevent normal cleavage of the signal peptide, resulting in altered protein processing and expression levels
Hyperactive RANK signaling promotes excessive osteoclast formation and activity, leading to enhanced bone resorption
The 84dup18 mutation has been identified in affected individuals from Northern Irish, American, and German FEO families, suggesting a common pathogenic mechanism
Loss-of-Function Mutations:
Clinical Manifestations and Diagnostic Considerations:
Familial Expansile Osteolysis (FEO):
Paget's Disease of Bone (PDB):
Osteopetrosis:
Increased bone density
Fractures despite increased bone mass due to poor bone quality
Hematopoietic failure
Neurological complications including vision and hearing loss
Therapeutic Implications:
Targeted Therapies:
Antiresorptive agents (bisphosphonates, denosumab) may be effective for conditions with increased osteoclast activity (FEO, PDB)
Recombinant RANKL might theoretically benefit patients with loss-of-function mutations
Development of small molecules targeting specific mutant RANK proteins or their downstream pathways
Personalized Medicine Approach:
Genetic testing to identify specific TNFRSF11A mutations
Tailoring treatment based on the molecular mechanism (gain vs. loss of function)
Monitoring response to therapy based on mutation status
Emerging Therapies:
Gene therapy approaches to correct specific mutations
Antisense oligonucleotides to modulate aberrant splicing
CRISPR-Cas9 gene editing for permanent correction of germline mutations
Genetic Counseling:
Identification of carriers within affected families
Preimplantation genetic diagnosis as an option for families with severe forms
Risk assessment for family members of affected individuals
Research in this area continues to evolve, with ongoing efforts to better understand genotype-phenotype correlations and develop more targeted therapeutic approaches based on the specific molecular defects in TNFRSF11A-associated bone disorders.
TNFRSF11A (RANK) has emerged as a significant factor in cancer pathogenesis, with evidence supporting its potential as a therapeutic target in various malignancies. The following summarizes current research findings and translational implications:
Evidence for TNFRSF11A in Cancer Pathogenesis:
Expression in Cancer Cells:
TNFRSF11A is expressed in various cancer cell lines, including breast cancer cell lines such as MDA-MB-231, SKBR3, MCF7, MDA-MB-468, and T47D
Alternative splice variants of TNFRSF11A (RANK-a, RANK-b, RANK-c) show differential expression patterns across cancer cell lines
TNFRSF11A expression patterns correlate with clinical parameters such as tumor grade and proliferation index (Ki-67) in breast cancer samples
Functional Role in Tumor Cells:
RANK signaling promotes tumor cell survival and proliferation through activation of NF-κB and other pro-survival pathways
RANK-RANKL axis contributes to cancer cell migration and invasion
RANK signaling may induce epithelial-to-mesenchymal transition (EMT), enhancing metastatic potential
Role in Metastasis:
RANK-expressing tumor cells preferentially metastasize to bone, where RANKL is abundantly expressed
RANK-RANKL interaction in bone microenvironment promotes a "vicious cycle" of bone destruction and tumor growth
RANK signaling contributes to the formation of premetastatic niches
Impact of Splice Variants:
Methodological Approaches to Study TNFRSF11A in Cancer:
Expression Analysis:
Functional Studies:
siRNA or CRISPR-Cas9 knockout to assess effects of RANK depletion on cancer cell behavior
Overexpression of wild-type RANK or specific splice variants to evaluate their role in cancer progression
In vitro assays for proliferation, migration, invasion, and resistance to apoptosis
In Vivo Models:
Orthotopic xenograft models to study the role of RANK in primary tumor growth
Metastasis models to evaluate its function in dissemination to distant sites, particularly bone
Genetic mouse models combining RANK alterations with cancer-driving mutations
Therapeutic Targeting Strategies:
Direct RANK Inhibition:
Anti-RANK monoclonal antibodies to block RANKL binding and subsequent signaling
Small molecule inhibitors of RANK dimerization or intracellular domain function
Peptide mimetics that disrupt RANK-RANKL interaction
Modulation of Splice Variants:
Antisense oligonucleotides to modify TNFRSF11A splicing patterns
Splice-switching oligonucleotides to promote expression of less oncogenic variants
Targeted degradation of cancer-associated splice variants
Combination Therapies:
RANK inhibitors with conventional chemotherapy
RANK-targeted agents with immune checkpoint inhibitors
Dual targeting of RANK and downstream effectors (e.g., NF-κB inhibitors)
Bone Metastasis Prevention:
Prophylactic use of RANK-targeted therapies in high-risk patients
Adjuvant therapy following primary tumor resection to prevent bone colonization
The emerging evidence for TNFRSF11A's involvement in cancer pathogenesis, particularly through alternative splicing mechanisms, represents a promising area for further research and therapeutic development. Clinical trials evaluating RANK-targeted approaches, either alone or in combination with established therapies, will be essential to translate these findings into improved patient outcomes.