TMEFF2 recombinant mAbs are engineered to bind specifically to the extracellular domain (ECD) of TMEFF2, which contains two follistatin-like (FS) domains and one epidermal growth factor (EGF)-like domain. These antibodies are typically produced via recombinant DNA technology, enabling high specificity and reproducibility .
TMEFF2 antibodies have been employed in preclinical and clinical studies to explore their diagnostic and therapeutic utility:
Antibody-Drug Conjugates (ADCs):
The huPr1-vcMMAE ADC, conjugating anti-TMEFF2 mAb to the cytotoxic agent auristatin E, demonstrated significant tumor inhibition in prostate cancer xenografts (3–10 mg/kg doses) .
JNJ-70218902, a TMEFF2 x CD3 bispecific antibody, showed robust T-cell responses against prostate cancer cells in early-phase trials .
Tumor-Specific Expression:
TMEFF2 binds amyloid-β (Aβ) oligomers, inhibiting their neurotoxicity in Alzheimer’s disease models. Antibodies blocking this interaction could modulate Aβ-related pathways .
TMEFF2 binds PDGF-AA via its FS domains, inhibiting receptor activation and downstream proliferation signals . Antibodies disrupting this interaction may suppress PDGF-driven tumor growth.
TMEFF2 promoter methylation is observed in colorectal, ovarian, and glioblastoma cancers, correlating with poor prognosis . Antibodies detecting methylation status could serve as biomarkers.
Tumor-Suppressive: Overexpression inhibits STAT3 phosphorylation and Mcl1 expression in pancreatic cancer, promoting apoptosis .
Oncogenic: Shed ECD fragments activate ERK1/2 and JAK-STAT pathways, driving proliferation .
Diagnostic Utility: Methylation-specific antibodies may enable early detection of colorectal cancer .
Therapeutic Challenges: TMEFF2’s dual role necessitates context-specific targeting. ADCs and bispecific antibodies are being explored to mitigate off-tumor toxicity .
Bispecific Antibodies: Combining TMEFF2 targeting with T-cell engagement (e.g., CD3) may enhance immunotherapy efficacy .
Biomarker Development: Validating TMEFF2 methylation as a predictive biomarker for cancer subtypes could personalize treatment strategies .
Neurological Applications: Exploring TMEFF2 antibodies in Alzheimer’s disease models to modulate Aβ toxicity .
This TMEFF2 monoclonal antibody was produced using recombinant human TMEFF2 protein as the immunogen. The TMEFF2 monoclonal antibody cDNA was sequenced, revealing the gene, which was subsequently cloned into a plasmid vector. This vector, carrying the TMEFF2 monoclonal antibody gene, was then transfected into a host cell using an appropriate method. The resulting recombinant TMEFF2 monoclonal antibody was then purified by affinity chromatography. Its specificity was confirmed through ELISA, demonstrating binding to recombinant human TMEFF2 (CSB-MP883439HU) with an EC50 range of 2.129-2.956 ng/mL. This antibody is capable of reacting with the TMEFF2 protein.
TMEFF2 is expressed in various tissues, including the brain, lung, prostate, and breast. Its primary function is as a tumor suppressor, as it has been shown to inhibit cell proliferation and induce apoptosis in specific cancer cell types, including prostate and breast cancer. TMEFF2 is also believed to play a role in neuronal differentiation and migration during brain development and may be involved in regulating synaptic transmission.
TMEFF2 is a 374-residue long single polypeptide, type-I transmembrane proteoglycan containing an EGF-like domain and two follistatin-like domains. It has attracted considerable research interest due to its differential expression in various cancers and potential roles in diverse physiological processes .
TMEFF2 is expressed in several tissues including brain, lung, prostate, and breast, with reported functions spanning a wide range of physiological and pathological spectra . It has been identified as having roles in:
Corticotropin release hormone stimulation in the anterior pituitary gland
Neuroprotection in Alzheimer's disease by binding amyloid β protein derivatives
JAK-STAT pathway signaling in colorectal cancer
The protein's tumor-specific expression pattern makes it a particularly attractive target for therapeutic antibody development, especially given its high overexpression in prostate cancer with limited distribution in normal tissues .
TMEFF2 monoclonal antibodies serve multiple critical research functions:
Protein Detection: Used in western blotting, immunohistochemistry, and flow cytometry to detect TMEFF2 expression in cell lines and tissues
Functional Studies: Help elucidate TMEFF2's role in cellular processes by blocking or activating its function
Therapeutic Development: Serve as platforms for developing antibody-drug conjugates for targeted cancer therapy
Diagnostic Research: Study methylated TMEFF2 as a potential diagnostic biomarker in various cancers
Mechanistic Research: Investigate protein-protein interactions, such as the TMEFF2-SARDH interaction
Validated applications for commercially available antibodies typically include ELISA, flow cytometry, and functional assays .
TMEFF2 detection methods vary depending on the experimental context. Western blot analysis reveals that TMEFF2 appears at different molecular weights depending on cell type, sample preparation conditions, and the specific antibody used:
Cell Line | Conditions | Antibody | Detected Bands (kDa) | Notes |
---|---|---|---|---|
LNCaP | Non-reducing | 2H-8 | 43 | Endogenous TMEFF2 |
PC3 (overexpressing) | Non-reducing | 2H-8 | 48, 52, 97 | V5/His tagged TMEFF2 |
CHO (overexpressing) | Reducing | Anti-V5 | 14, 71 | V5/His tagged TMEFF2 |
HEK293, LNCaP (media) | Not specified | Anti-TMEFF2 | 60-75 | Shed form in media |
HEK293 (overexpressing) | Not specified | Anti-V5 | 10, 14, 22 | Cell lysate bands |
This variability reflects the complex post-translational modifications of TMEFF2 including glycosylation and proteolytic processing . For consistent results, researchers should carefully select antibodies validated for their specific application and experimental system.
The literature presents seemingly contradictory data about TMEFF2's role in cancer, with evidence supporting both oncogenic and tumor-suppressive functions. To reconcile these findings, consider:
Cellular Context Dependency: TMEFF2 function appears highly context-dependent. In prostate cancer studies, full-length TMEFF2 demonstrates tumor suppressor properties while the shed ectodomain exhibits oncogenic characteristics .
Differential Protein Processing: The protein undergoes ectodomain shedding, which generates fragments with potentially opposing functions. Research indicates that:
Methodological Differences: Different detection methods, cell models, and functional assays contribute to variable findings.
When designing experiments, carefully consider which protein form you're studying (full-length vs. ectodomain), the cellular context, and the specific signaling pathways being examined. Conducting parallel experiments with both forms can help clarify these seemingly contradictory roles.
When conducting immunoprecipitation (IP) studies with TMEFF2 antibodies, researchers should consider:
Epitope Accessibility: TMEFF2's complex structure with multiple domains may affect epitope accessibility. Select antibodies that target epitopes known to be accessible in native conditions.
Protein-Protein Interactions: TMEFF2 interacts with proteins like SARDH . The antibody chosen should not interfere with the interaction being studied or deliberately block it depending on experimental goals.
Cross-Reactivity: Verify specificity through proper controls. The high homology between TMEFF1 and TMEFF2 (35.8%) necessitates validation of antibody specificity.
Post-Translational Modifications: Consider how glycosylation may affect antibody binding. TMEFF2 exhibits multiple bands on Western blots due to extensive glycosylation .
Buffer Conditions: Optimize lysis and washing conditions to maintain protein interactions while minimizing non-specific binding.
For interaction studies with SARDH specifically, using antibodies targeting epitopes away from the SARDH-binding region would be ideal to avoid disrupting the interaction being studied .
TMEFF2 antibodies can be valuable tools for investigating the complex relationship between TMEFF2 expression and cancer progression through several methodological approaches:
Tissue Microarray Analysis: Immunohistochemistry using validated TMEFF2 antibodies can quantify expression across cancer stages. Past studies have shown significant TMEFF2 protein expression in 74% of primary prostate cancers and 42% of metastatic lesions from lymph nodes and bone, representing both hormone-naïve and hormone-resistant disease .
Correlation with Methylation Status: TMEFF2 methylation increases with breast, colon and gastric cancer progression . Combining antibody-based protein detection with methylation analysis can reveal epigenetic regulation mechanisms.
Comparative Expression Studies: Using antibodies to compare TMEFF2 expression in:
Normal vs. tumor tissue
Primary vs. metastatic sites
Treatment-responsive vs. resistant samples
Functional Blocking Studies: Antibodies that block specific domains of TMEFF2 can help determine which regions are critical for its tumor-suppressive or oncogenic functions.
Circulating TMEFF2 Detection: Developing assays to detect shed TMEFF2 in blood samples as potential liquid biopsy markers.
For prostate cancer specifically, research indicates that the TMEFF2 downregulation signature equals and sometimes outperforms the Gleason and pathological scores, suggesting its potential value as a prognostic marker .
Thorough validation of TMEFF2 antibodies is essential for reliable experimental results. The following methodological validation steps are recommended:
Specificity Testing:
Western blot analysis using positive controls (e.g., LNCaP cells with known endogenous TMEFF2 expression)
Comparison of signals between TMEFF2-expressing and non-expressing cell lines
Knockdown/knockout validation to confirm specificity
Testing for cross-reactivity with TMEFF1 due to the 35.8% homology
Application-Specific Validation:
For immunohistochemistry: Include tissue samples with known TMEFF2 expression patterns
For flow cytometry: Compare staining patterns with isotype controls
For functional assays: Verify blocking or activating effects with appropriate biological readouts
Technical Controls:
Epitope Mapping:
Batch-to-Batch Consistency Testing:
Verify consistent performance across different antibody lots using standardized positive controls
To effectively investigate TMEFF2's dual role, consider these methodological approaches:
Compare Full-Length vs. Ectodomain:
Express full-length TMEFF2 and the shed ectodomain separately in the same cell line
Assess differential effects on proliferation, migration, and invasion
Examine downstream signaling pathways activated by each form
Research has shown that full-length TMEFF2 decreases cell numbers by 20-30% compared to controls, while the ectodomain shows no tumor suppressor activity .
Domain-Specific Function Analysis:
Generate domain deletion constructs to identify regions responsible for tumor suppression or oncogenic activity
Create chimeric proteins swapping domains between TMEFF1 and TMEFF2 to identify critical functional domains
Context-Dependent Studies:
Test TMEFF2 function across multiple cell lines representing different cancer stages
Investigate effects under various conditions (hormone treatment, hypoxia, inflammation)
Mechanistic Investigations:
Examine TMEFF2-SARDH interaction and sarcosine regulation
Investigate ectodomain shedding mechanisms and regulation
Study effects on one-carbon metabolism pathways
In vivo Models:
Compare xenograft growth with full-length vs. ectodomain TMEFF2 expression
Evaluate metastatic potential and response to therapy
When designing these experiments, include appropriate controls and consider using inducible expression systems to study the temporal effects of TMEFF2 expression.
Developing TMEFF2 antibody-drug conjugates (ADCs) requires careful consideration of several methodological factors:
Target Expression Profile:
Antibody Characteristics:
Linker-Drug Chemistry:
Preclinical Testing Strategy:
Test in models expressing varying levels of TMEFF2
Include TMEFF2-negative models as specificity controls
Evaluate potential on-target, off-tumor toxicity
In preclinical studies, doses of 3-10 mg/kg of Pr1-vcMMAE resulted in significant and sustained tumor growth inhibition in xenografted prostate cancer models, while an isotype control ADC had no significant effect .
Cross-Species Reactivity Assessment:
Resistance Mechanisms:
Investigate potential resistance mechanisms such as decreased target expression or increased efflux
These considerations should guide the methodological approach to developing effective TMEFF2-targeted therapeutic ADCs.