TMEFF2 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Structure and Mechanism of TMEFF2 Recombinant Monoclonal Antibodies

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 .

Antibody CloneApplicationsConjugate/LabelSource
Janssen patent anti-TMEFF2ELISA, Flow Cytometry, FunctionalJanelia Fluor 585Bio-Techne
huPr1-vcMMAEAntibody-Drug Conjugate (ADC)Auristatin E (vcMMAE linker)Genentech
CSB-RA883439MA1HUELISA, Western BlotUnconjugatedCUSABIO
ARC2117 (MA5-38106)IHC, Flow CytometryUnconjugatedThermo Fisher
6G6 (MA5-50086)IHC, Western BlotUnconjugatedThermo Fisher

Research Applications and Therapeutic Potential

TMEFF2 antibodies have been employed in preclinical and clinical studies to explore their diagnostic and therapeutic utility:

Cancer Therapy

  • 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 is overexpressed in prostate and endometrial cancers but hypermethylated and downregulated in colorectal, gastric, and pancreatic cancers . Antibodies targeting TMEFF2 may exploit this differential expression for selective killing.

Neuroprotection

  • TMEFF2 binds amyloid-β (Aβ) oligomers, inhibiting their neurotoxicity in Alzheimer’s disease models. Antibodies blocking this interaction could modulate Aβ-related pathways .

Signaling Pathway Modulation

  • 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 Hypermethylation in Cancers

  • TMEFF2 promoter methylation is observed in colorectal, ovarian, and glioblastoma cancers, correlating with poor prognosis . Antibodies detecting methylation status could serve as biomarkers.

Dual Role in Cancer Progression

  • 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 .

Clinical Relevance of TMEFF2 Antibodies

  • 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 .

Future Directions

  • 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 .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

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.

Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
Cancer/testis antigen family 120 member 2 antibody; CT120.2 antibody; HPP 1 antibody; HPP1 antibody; Hyperplastic polyposis protein 1 antibody; TEFF 2 antibody; TEFF2 antibody; TEFF2_HUMAN antibody; TENB 2 antibody; TENB2 antibody; TMEFF2 antibody; Tomoregulin antibody; Tomoregulin-2 antibody; TPEF antibody; TR 2 antibody; TR antibody; TR-2 antibody; Transmembrane protein containing epidermal growth factor and follistatin domains antibody; Transmembrane protein TENB2 antibody; Transmembrane protein with EGF like and two follistatin like domains 2 antibody; Transmembrane protein with EGF-like and two follistatin-like domains antibody
Target Names
Uniprot No.

Target Background

Function
TMEFF2 potentially acts as a survival factor for hippocampal and mesencephalic neurons. Its shedded form upregulates cancer cell proliferation, likely by promoting ERK1/2 phosphorylation.
Gene References Into Functions
  1. Differential TMEFF2 processing from a single transmembrane protein might be a general mechanism to modulate transmembrane protein levels and domains, dependent on the repertoire of ADAMs or TTSPs expressed by the target cell. PMID: 28762604
  2. TMEFF2 methylation is associated with clear cell renal cell carcinoma. PMID: 28128743
  3. Research suggests that TMEFF2 is a brain-enriched endogenous modulator of Abeta neurotoxicity and an enhancer of alpha-secretase processing of AbetaPP. PMID: 26402097
  4. TMEFF2 regulates the non-canonical activin/BMP4 signaling, PI3K, and Ras/ERK1/2 pathways. PMID: 25573902
  5. c-Myc contributes to the epigenetic regulation of HPP1 via the dominant recruitment of HDAC3. PMID: 24919179
  6. TMEFF2 acts as a tumor suppressor in gastric cancer through direct interaction with SHP-1 and could serve as a potential biomarker of carcinogenesis. PMID: 24987055
  7. TMEFF2 and SARDH cooperate to modulate one-carbon metabolism and invasion of prostate cancer cells. PMID: 23824605
  8. Androgen signaling promotes eIF2alpha phosphorylation and subsequent translation of TMEFF2 via a mechanism that requires uORFs in the 5'-UTR of TMEFF2. PMID: 23405127
  9. Methylation of the TMEFF2 gene is associated with colorectal neoplasia in ulcerative colitis and Crohn's colitis. PMID: 22532293
  10. Findings suggest that methylation-associated downregulation of the TMEFF2 gene may be involved in lung tumorigenesis, and TMEFF2 methylation can serve as a specific blood-based biomarker for NSCLC. PMID: 22814847
  11. Several genes expressed at exceptionally high levels were identified as being associated with early oocyte development, including TMEFF2, the Rho-GTPase-activating protein oligophrenin 1 (OPHN1), and the mitochondrial-encoded ATPase6 (ATP6). PMID: 22238370
  12. TMEFF2 can function to regulate PDGF signaling; it is hypermethylated and downregulated in glioma and several other cancers. PMID: 21559523
  13. The tumor suppressor activity of TMEFF2 requires the cytoplasmic/transmembrane portion of the protein and correlates with its ability to bind to SARDH and modulate sarcosine levels. PMID: 21393249
  14. GDF15, HSPA2, TMEFF2, and VIM were identified as epigenetic biomarkers for bladder cancer. PMID: 20975101
  15. Hypermethylation of HPP1 is associated with hMLH1 hypermethylation in gastric adenocarcinomas. PMID: 12384516
  16. Aberrant methylation of the HPP1 gene is a relatively common early event in ulcerative colitis-associated colorectal carcinoma. PMID: 12460892
  17. An inverse correlation exists between TMEFF2 and c-Myc expression. PMID: 12729735
  18. Hypermethylation of p16, RUNX3, and HPP1 in Barrett's esophagus may represent independent risk factors for the progression of Barrett's esophagus to esophageal cancer. PMID: 15824739
  19. A secreted form of TMEFF2 is expressed from the TMEFF2 locus and may functionally interact with full-length TMEFF2, or its binding partners. This form might also influence current immune-based treatment strategies. PMID: 16439095
  20. Hypermethylation of HPP1 is associated with primary adenocarcinomas of the small bowel. PMID: 16619216
  21. Methylation testing of fecal DNA using a panel of epigenetic markers (methylated SFRP2, HPP1, and MGMT) could be a simple and promising non-invasive screening method for colorectal carcinoma and precancerous lesions. PMID: 17352030
  22. Data provides evidence to support the role of HPP1 as a tumor suppressor gene; activation of the STAT1 pathway likely represents the principal mediator of HPP1's tumor suppressive properties. PMID: 18059030
  23. Methylated PAX6- or TPEF-promoters could represent biomarkers for bladder cancer. PMID: 18070176
  24. Distinct TPEF/HPP1 (transmembrane protein containing epidermal growth factor) gene methylation patterns in gastric cancer indicate a field effect in gastric carcinogenesis. PMID: 18799374
  25. The promoter of the TPEF gene is frequently hypermethylated, and associated with loss of TPEF mRNA expression in esophageal squamous cell carcinoma. PMID: 19040536
  26. Methylation of CLDN6, FBN2, RBP1, RBP4, TFPI2, and TMEFF2 in esophageal squamous cell carcinoma. PMID: 19288010
  27. Analysis of DNA from peripheral blood revealed that TPEF methylation was detectable in colorectal tumor patients and patients with early or pre-neoplastic lesions, but not in healthy volunteers. PMID: 15068392

Show More

Hide All

Database Links

HGNC: 11867

OMIM: 605734

KEGG: hsa:23671

STRING: 9606.ENSP00000272771

UniGene: Hs.144513

Protein Families
Tomoregulin family
Subcellular Location
[Isoform 1]: Membrane; Single-pass type I membrane protein.; [Isoform 2]: Membrane; Single-pass type I membrane protein.; [Isoform 3]: Secreted.
Tissue Specificity
Highly expressed in adult and fetal brain, spinal cord and prostate. Expressed in all brain regions except the pituitary gland, with highest levels in amygdala and corpus callosum. Expressed in the pericryptal myofibroblasts and other stromal cells of nor

Q&A

What is TMEFF2 and why is it an important research target?

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

  • One-carbon metabolism modulation in prostate tissue

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 .

What are the main applications of TMEFF2 monoclonal antibodies in research?

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 .

How is TMEFF2 protein typically detected in experimental systems?

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 LineConditionsAntibodyDetected Bands (kDa)Notes
LNCaPNon-reducing2H-843Endogenous TMEFF2
PC3 (overexpressing)Non-reducing2H-848, 52, 97V5/His tagged TMEFF2
CHO (overexpressing)ReducingAnti-V514, 71V5/His tagged TMEFF2
HEK293, LNCaP (media)Not specifiedAnti-TMEFF260-75Shed form in media
HEK293 (overexpressing)Not specifiedAnti-V510, 14, 22Cell 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.

How can I reconcile contradictory findings regarding TMEFF2's role in cancer progression?

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:

    • Full-length TMEFF2 decreases cell proliferation by 20-30% compared to control cells

    • The TMEFF2 ectodomain fails to bind SARDH and modulate sarcosine levels, reversing the tumor suppressor phenotype

  • 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.

What are the considerations for using TMEFF2 antibodies for immunoprecipitation studies?

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 .

How can TMEFF2 antibodies be utilized in studying the relationship between TMEFF2 expression and cancer progression?

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 .

What validation steps should be performed when using TMEFF2 antibodies for research?

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:

    • Include recombinant TMEFF2 as a positive control

    • Use appropriate negative controls (isotype control antibodies)

    • Evaluate binding under reducing and non-reducing conditions, as TMEFF2 detection varies significantly between these conditions

  • Epitope Mapping:

    • Confirm which domain of TMEFF2 the antibody recognizes (EGF-like domain, follistatin-like domains, transmembrane region, or cytoplasmic domain)

    • This is critical as different domains may exhibit different functions

  • Batch-to-Batch Consistency Testing:

    • Verify consistent performance across different antibody lots using standardized positive controls

How can I design experiments to investigate the dual role of TMEFF2 in oncogenesis and tumor suppression?

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.

What are the key considerations when developing TMEFF2 antibody-drug conjugates for therapeutic applications?

Developing TMEFF2 antibody-drug conjugates (ADCs) requires careful consideration of several methodological factors:

  • Target Expression Profile:

    • Confirm target specificity in disease tissues vs. normal tissues

    • TMEFF2 shows significant expression in 74% of primary prostate cancers and 42% of metastatic lesions but limited normal tissue distribution

    • Quantify expression levels to predict potential efficacy and toxicity

  • Antibody Characteristics:

    • Select antibodies with high affinity and specificity for TMEFF2

    • Evaluate internalization efficiency, as ADCs require internalization for payload delivery

    • Consider humanization to reduce immunogenicity (as demonstrated with huPr1-vcMMAE)

  • Linker-Drug Chemistry:

    • Select appropriate linker chemistry (e.g., cathepsin B-sensitive valine-citrulline linker used in Pr1-vcMMAE)

    • Choose payload based on potency requirements and mechanism of action

    • Optimize drug-antibody ratio for efficacy/toxicity balance

  • 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:

    • Determine cross-reactivity with murine TMEFF2 to better predict toxicity

    • Past research noted no overt in vivo toxicity with either murine or human ADC despite significant cross-reactivity with murine TMEFF2

  • 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.

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.