TMEFF2 antibodies are designed to detect and bind TMEFF2, a 374-residue type-I transmembrane proteoglycan encoded by the TMEFF2 gene (chromosome 2q32-q33) . Key structural features include:
Domains: Two follistatin-like domains, a truncated EGF-like domain, a transmembrane region, and a short intracellular tail .
Isoforms: Full-length membrane-bound and proteolytically shed extracellular (ECD) forms, with distinct roles in cancer progression .
ADC Name | Payload | Mechanism | Efficacy (Preclinical) |
---|---|---|---|
Pr1-vcMMAE | Auristatin E | Microtubule disruption | 3–10 mg/kg: >50% tumor inhibition |
JNJ-70218902 | Bispecific | T-cell recruitment | Robust response in xenografts |
TMEFF2 exhibits context-dependent oncogenic or tumor-suppressive effects:
Oncogenic: Promotes prostate cancer progression via JAK-STAT pathway activation .
Tumor-Suppressive: Hypermethylation-linked silencing in colorectal, gastric, and bladder cancers correlates with poor prognosis .
NCT04397276: Phase I trial evaluating JNJ-70218902 in metastatic castration-resistant prostate cancer (mCRPC) .
Functional Complexity: Conflicting reports on TMEFF2’s role in prostate cancer necessitate further mechanistic studies .
Therapeutic Potential: ADCs and bispecific antibodies show promise but require validation in late-stage trials .
Diagnostic Utility: Methylation status in liquid biopsies could enable non-invasive cancer detection .
TMEFF2 is a 374-residue long single polypeptide, type-I transmembrane proteoglycan with two follistatin-like domains and one epidermal growth factor-like domain. Its value as an antibody target stems from its highly restricted expression pattern in normal tissues (primarily brain and prostate) coupled with significant overexpression in prostate cancer . Immunohistochemistry studies have confirmed significant TMEFF2 protein expression in 73-74% of primary prostate tumors and 42% of metastatic lesions from lymph nodes and bone that represented both hormone-naïve and hormone-resistant disease . More recent studies have detected TMEFF2 in 91% (41/45) of metastatic castration-resistant prostate cancer (mCRPC) samples .
Research into TMEFF2 antibodies has developed several distinct types:
Conventional monoclonal antibodies: Used primarily for detection in techniques like immunohistochemistry and flow cytometry .
Antibody-drug conjugates (ADCs): Exemplified by Pr1-vcMMAE, which combines anti-TMEFF2 antibodies with the cytotoxic agent auristatin E via a cathepsin B-sensitive valine-citrulline linker .
Bispecific T-cell engagers: JNJ-70218902 (JNJ-902) represents this approach, engaging both TMEFF2 on tumor cells and CD3 on T cells to promote T cell-mediated killing of cancer cells .
Humanized antibodies: Modified versions like huPr1-vcMMAE with reduced immunogenicity for potential clinical applications .
Validation of TMEFF2 expression typically involves multiple complementary approaches:
Binding assays: JNJ-902 demonstrated concentration-dependent binding to TMEFF2-positive LNCaP-AR cells (EC50 = 9.6 nM) with no binding to TMEFF2-negative DU145 prostate cancer cells, confirming specificity .
Immunohistochemistry (IHC): Using specific monoclonal antibodies to detect TMEFF2 protein in tissue samples, allowing for quantification of expression rates across tumor types .
Gene expression profiling: Identifying TMEFF2 as highly expressed in prostate cancer with limited normal tissue distribution .
Cross-reactivity testing: Evaluating antibody reactivity with murine TMEFF2 to assess potential off-target effects, which is particularly important given the 100% sequence homology between human and monkey TMEFF2 .
Based on current research, effective evaluation protocols include:
Incubate antibodies with healthy human donor T cells and target tumor cells at specific effector-to-target ratios (typically 3:1)
Measure caspase-3 activity as an indicator of cell death (JNJ-902 showed EC50 = 1.4 nM in such assays)
Include appropriate negative controls (TMEFF2-negative cell lines and control antibodies)
Measure concentration-dependent increases in cell surface markers (CD8+CD25+)
Quantify proinflammatory cytokine production (GM-CSF, IFN-γ, IL-10, TNF-α)
Assess granzyme B expression and proliferation markers (Ki-67)
Optimal experimental design should include:
T cell humanized NSG mice bearing LNCaP xenografts or LuCaP 86.2 patient-derived xenografts have proven effective
Models should represent clinical scenarios of interest, particularly treatment-resistant disease
Test multiple dose levels (3-10 mg/kg showed efficacy in preclinical models)
Consider step-up dosing approaches, which showed enhanced CD8+ T cell infiltration in non-human primate studies
Tumor growth inhibition (TGI) relative to control-treated mice (JNJ-902 demonstrated 72-122% TGI in preclinical models)
Intratumoral T cell infiltration analysis by flow cytometry
Assessment of T cell activation markers within the tumor microenvironment
The most informative analytical approaches include:
Flow cytometry to assess T cell infiltrates and myeloid cells in target tissues and blood
Quantification of surface markers of T cell function (activation, proliferation, and suppression)
Measurement of CD4+CD25hi Foxp3+ regulatory T cells to monitor potential immunosuppression
Assessment of pro-inflammatory cell influx (dendritic cells, myeloid cells, macrophages, and B cells) into target tissues
Correlation of pharmacokinetic parameters with biological responses
Consideration that clinical activity may not always be dose-related, as observed in the phase 1 study of JNJ-902
The multifaceted nature of TMEFF2 presents a significant challenge to researchers . A methodological approach to resolving these contradictions includes:
Context-dependent analyses: Design experiments that investigate TMEFF2 function across different cancer stages, microenvironments, and genetic backgrounds
Signaling pathway dissection: Determine how TMEFF2 interacts with various signaling networks that might lead to opposing outcomes in different cellular contexts
Proteolytic processing studies: Examine whether cleaved forms of TMEFF2 have different functions than the membrane-bound protein
Expression level effects: Investigate whether different expression levels trigger different cellular responses
Interaction partner identification: Characterize proteins that interact with TMEFF2 in different contexts to elucidate mechanistic differences
Based on current research, promising strategies include:
Step-up dosing approaches (starting with 0.075 mg/kg followed by 0.3 mg/kg one week later) demonstrated greater CD8+ T cell infiltration compared to fixed dosing in non-human primates
Exploration of different administration schedules (once weekly versus biweekly) as evaluated in clinical studies
Consider rational combinations with other immunomodulatory agents
Target the tumor microenvironment to overcome immunosuppression
Develop predictive biomarkers based on TMEFF2 expression levels
Consider tumor immune microenvironment characteristics that may influence response
Addressing translational gaps requires systematic analysis:
Pharmacokinetic/pharmacodynamic (PK/PD) assessment:
Response heterogeneity analysis:
Resistance mechanism characterization:
Serial biopsies to study adaptive changes during treatment
Development of patient-derived models from non-responders
Translational biomarker development:
Despite TMEFF2's expression in normal brain and prostate, research suggests several strategies to manage toxicity:
Leverage the cross-reactivity of anti-TMEFF2 antibodies with murine TMEFF2 to evaluate potential off-target effects
Studies with both Pr1-vcMMAE and huPr1-vcMMAE demonstrated no overt in vivo toxicity despite this cross-reactivity, suggesting a high safety profile
In the JNJ-902 clinical study, dose-limiting toxicities were observed in only 2.4% of patients
Step-up dosing approaches may improve tolerability while maintaining efficacy
Engineering antibodies with optimized binding properties
Exploring conditional activation mechanisms that restrict activity to the tumor microenvironment
When facing experimental variability, consider these methodological approaches:
Antibody validation:
T cell functionality assessment:
Target expression verification:
Quantify TMEFF2 expression levels in experimental models
Consider heterogeneity within cell populations
Experimental controls:
Given the variable expression of TMEFF2 across patient samples (73-74% of primary tumors, 42% of metastatic lesions) , researchers should consider:
Multi-region sampling to account for intratumoral heterogeneity
Correlation of expression with clinical parameters and outcomes
Establish clinically relevant expression thresholds for patient selection
Consider both percentage of positive cells and intensity of staining
Identify additional markers that may enhance patient selection
Develop algorithms that incorporate multiple parameters
Assess changes in TMEFF2 expression during disease progression and treatment
Develop liquid biopsy approaches for longitudinal assessment
While bispecific antibodies (JNJ-902) and ADCs (Pr1-vcMMAE) dominate current TMEFF2 research, several innovative approaches warrant investigation:
Trispecific antibodies: Targeting TMEFF2, CD3, and an additional immune checkpoint molecule
CAR-T cell therapy: Engineering T cells with TMEFF2-specific chimeric antigen receptors
Antibody-cytokine fusions: Combining TMEFF2 targeting with localized cytokine delivery
Proteolytically-activated antibodies: Engineered to become fully active only in the tumor microenvironment
Radioimmunotherapy conjugates: TMEFF2 antibodies coupled with therapeutic radioisotopes
Strategic combinations to explore include:
Combining TMEFF2 bispecific antibodies with PD-1/PD-L1 blockade
Addressing T cell exhaustion mechanisms observed in persistent disease
Targeting immunosuppressive cell populations
Enhancing T cell trafficking and infiltration
Investigating synergy with androgen receptor-targeted therapies
Exploring radiation therapy combinations to enhance immunogenic cell death
Determining optimal treatment sequences
Developing rational switching protocols based on response assessment
To address inevitable treatment resistance, researchers should prioritize:
Serial biopsies during treatment and at progression
Liquid biopsy approaches for noninvasive monitoring
Generation of resistant cell lines through prolonged exposure
Patient-derived xenografts from non-responding patients
TMEFF2 expression changes (downregulation, mutation, alternative splicing)
Altered signaling pathway activation
Immune escape mechanisms (T cell exhaustion, immunosuppressive cell recruitment)
Integrative analysis of multi-omics data from resistant samples
AI-assisted prediction of resistance mechanisms and rational combination strategies
Tomoregulin-2 is a transmembrane protein that contains EGF-like and follistatin-like domains. These domains are involved in various cellular processes, including cell signaling and regulation of cell growth. The protein has been shown to function as both an oncogene and a tumor suppressor, depending on the cellular context . This dual role suggests that Tomoregulin-2 may play a complex role in cancer biology, particularly in prostate cancer cell invasion .
The Mouse Anti Human Tomoregulin-2 Antibody is a monoclonal antibody derived from mouse and is used to detect human TMEFF2 in various applications, including Western blotting and immunohistochemistry . This antibody is highly specific and can be used to study the expression and localization of TMEFF2 in human tissues.
Research on Tomoregulin-2 has significant implications for understanding cancer biology, particularly in the context of prostate cancer. The ability of Tomoregulin-2 to act as both an oncogene and a tumor suppressor highlights its potential as a therapeutic target. Studies using the Mouse Anti Human Tomoregulin-2 Antibody can provide insights into the protein’s role in cancer progression and may lead to the development of new diagnostic and therapeutic strategies.