The Tf Antibody, FITC conjugated is a fluorescently labeled antibody targeting Tissue Factor (TF), a transmembrane glycoprotein involved in blood coagulation and tumor progression. FITC (Fluorescein isothiocyanate) conjugation enables visualization of TF in research applications such as flow cytometry, immunofluorescence, and immunohistochemistry. Below, we detail its structure, applications, and research findings based on diverse sources.
The antibody is typically a purified IgG or polyclonal IgG conjugated to FITC via its reactive isothiocyanate group, forming stable thiourea linkages with lysine residues or the amino terminus of the antibody . Key structural features:
Fluorochrome-to-protein (F/P) ratio: Critical for detection sensitivity. Lower F/P ratios (e.g., 5.2:1) reduce fluorescence intensity but minimize quenching .
Epitope specificity: Targets the extracellular domain of TF, avoiding interference with its coagulation function .
FITC-conjugated TF antibodies are used in ADCs to deliver cytotoxic agents to tumor cells. For example:
TF-011-MMAE: Achieved complete tumor regression in patient-derived xenograft models with variable TF expression .
Duostatin-3 conjugates: Showed superior cytotoxicity compared to EGFR/HER2-targeted ADCs in xenograft models .
| ADC Molecule | Target | Mechanism | Efficacy in Models |
|---|---|---|---|
| TF-011-MMAE | Tissue Factor | Auristatin-mediated killing | Complete regression in PDX |
| Duostatin-3 conjugates | TF | Tubulin polymerization inhibition | Reduced tumor growth vs. EGFR/HER2 ADCs |
Detects TF expression on tumor cells, endothelial cells, and extracellular vesicles (EVs) .
F/P ratio impact: Lower ratios (5.2:1) reduce EV detection (e.g., 1.1% TF+ EVs) but preserve cell detection .
Internalization dynamics: TF exhibits rapid endocytosis and lysosomal degradation, enhancing ADC efficacy .
Biomarker for tumor vasculature: Overexpressed in tumor neovasculature and cancer stem cells .
F/P ratio variability: Affects EV detection but not cell staining .
Epitope competition: Overlapping epitopes between clones may reduce assay specificity .
Coagulation interference: Some antibodies disrupt TF:FVIIa signaling, necessitating epitope mapping .
Tissue Factor (TF), also known as CD142, Coagulation Factor III, F3, or Thromboplastin, is a transmembrane glycoprotein that initiates the extrinsic pathway of blood coagulation. Beyond its role in coagulation, TF is aberrantly expressed in various solid cancers and is implicated in tumor progression through both its procoagulant activity and its capacity to induce intracellular signaling when complexed with Factor VIIa (FVIIa) . TF is frequently expressed in highly invasive triple negative breast cancer (TNBC) and pancreatic adenocarcinoma (PaC), which are among the most challenging tumor types with poor survival rates and limited therapeutic options . The pathological presence of TF in cancer cells has been linked to tumor-initiated thrombosis and cancer metastasis, making it a promising target for therapeutic interventions .
TF Antibody, FITC conjugated is a purified antibody validated for specificity and sensitivity, with the fluorescent dye FITC chemically attached to enable direct visualization in fluorescence-based applications . The FITC molecule has excitation and emission spectrum peak wavelengths of approximately 495 nm and 519 nm, producing green fluorescence when excited with the appropriate wavelength light . Unlike other research antibodies that may require secondary detection systems, FITC-conjugated antibodies allow for direct detection, simplifying experimental workflows and reducing background in multi-color experiments. The specific TF antibody products are validated across multiple applications including immunofluorescence (IF), immunocytochemistry (ICC), immunohistochemistry (IHC), and flow cytometry (FACS) .
The conjugation of FITC to antibodies must be carefully controlled to maintain antibody function. Traditional random conjugation methods can potentially impact the antigen-binding domain's functionality. Advanced site-specific conjugation technologies (similar to FluoSite™ mentioned for other antibodies) ensure that the labeling site is positioned away from the antigen-binding domain, preserving antibody functionality . When evaluating a TF antibody with FITC conjugation, researchers should consider the fluorophore-to-protein ratio (F/P ratio) as this impacts both signal strength and potential interference with antibody binding. Optimally conjugated antibodies maintain high affinity binding while providing sufficient fluorescence for detection .
When using TF Antibody, FITC conjugated for flow cytometry, researchers should:
Begin with cell concentrations of 1 × 10^6 cells/ml in appropriate buffer (typically PBS with 1-2% BSA)
Use titrated antibody concentrations to determine optimal signal-to-noise ratio (typically starting with manufacturer recommendations of approximately 10 μg/ml)
Incubate cells with antibody for 30-45 minutes at 4°C protected from light
Wash cells twice with buffer to remove unbound antibody
Analyze immediately or fix with 1-2% paraformaldehyde if analysis must be delayed
Include appropriate controls:
Unstained cells
Isotype control antibody-FITC to assess non-specific binding
Positive control samples with known TF expression
Flow cytometric analysis has been used successfully to assess the binding activity of anti-TF antibodies to cancer cell lines with high TF expression, such as the gastric cancer line 44As3 and pancreatic cancer line BxPC3 . The method allows quantitative assessment of binding affinity differences between various anti-TF constructs.
To validate TF antibody specificity, researchers should implement a multi-faceted approach:
Positive and negative cell line panel testing: Use cell lines with documented high TF expression (e.g., BxPC3 pancreatic cancer cells, 44As3 gastric cancer cells) and negative control cell lines
Competitive binding assays: Pre-incubate with unconjugated TF antibody or recombinant TF protein before adding the FITC-conjugated antibody
siRNA or CRISPR knockdown: Create TF knockdown cell lines and demonstrate reduced binding
Western blot correlation: Confirm that fluorescence intensity correlates with protein expression levels detected by Western blot
Tissue validation: Compare staining patterns with literature-reported TF expression in tissue sections
In published research, TF antibody specificity has been validated using tissue factor-transfected HEK293F or A431 cells and by comparison with bead-coupled TF-ECDHis using Fluorimetric Microvolume Assay Technology .
For maximum stability and performance:
Store the antibody at 2-8°C and avoid freezing, as noted in product specifications
Protect from extended light exposure to prevent photobleaching of the FITC fluorophore
If lyophilized, reconstitute in sterile water or buffer specified by the manufacturer
After reconstitution, prepare working aliquots to avoid repeated freeze-thaw cycles
For long-term storage of reconstituted antibody, aliquot and store at -20°C
When working with the antibody, minimize exposure to light and maintain cold temperatures
Check the expiration date and lot-specific quality control data provided by the manufacturer
Consider adding sodium azide (0.02%) to prevent microbial growth if preparing working solutions, but be aware this may interfere with some applications (especially those involving peroxidase)
Lyophilized TF Antibody, FITC conjugated is typically presented in a stabilizing buffer containing PBS pH 7.4, 20 mg/ml BSA, 0.02% Sodium Azide, and 4% Trehalose to maintain stability .
TF represents an excellent target for antibody-drug conjugate development due to its rapid and efficient internalization properties. The development process typically involves:
Selection of appropriate anti-TF antibody clone: Identify antibodies that efficiently induce inhibition of TF:FVIIa-dependent intracellular signaling, antibody-dependent cell-mediated cytotoxicity, and rapid target internalization, while minimally impacting TF procoagulant activity
Conjugation strategy design: Methods include:
In vitro evaluation: Testing for:
In vivo assessment: Weekly intravenous administration protocols in xenograft models to determine:
Research has demonstrated that TF-targeting ADCs have shown effective killing against tumor cell lines with variable levels of target expression and relative potency in reducing tumor growth compared with EGFR- and HER2-ADCs .
TF exhibits several advantageous characteristics that make it particularly suitable for ADC development:
Enhanced internalization efficiency: Both in the absence and presence of antibody, TF demonstrates more efficient internalization than EGFR and HER2
Improved lysosomal targeting: Research has shown superior lysosomal targeting and degradation of TF compared to EGFR and HER2, which is critical for the release of cytotoxic payloads within target cancer cells
Constant turnover rate: The constant turnover of TF on tumor cells makes this protein specifically suitable for an ADC approach, allowing for continuous delivery of cytotoxic agents to tumor cells
Selective expression pattern: TF is frequently overexpressed in challenging tumor types like TNBC and pancreatic adenocarcinoma, which have limited therapeutic options, making it an attractive target for these difficult-to-treat cancers
Comparative studies have demonstrated that TF-ADC showed effective killing against tumor cell lines with variable levels of target expression and was relatively potent in reducing tumor growth compared with EGFR- and HER2-ADCs in xenograft models .
To quantitatively evaluate TF antibody internalization, researchers can employ:
Flow cytometry-based acid wash technique:
Label cells with FITC-conjugated TF antibody at 4°C
Incubate at 37°C for various time points (0-120 minutes)
Treat with acid wash buffer (0.2M acetic acid, 0.5M NaCl, pH 2.5) to remove surface-bound antibody
Analyze remaining intracellular fluorescence by flow cytometry
Calculate internalization rate as percentage of initial surface binding
Confocal microscopy with co-localization markers:
Pulse-label cells with FITC-conjugated TF antibody
Chase at 37°C for various timepoints
Fix and co-stain with markers for early endosomes (EEA1), late endosomes (Rab7), or lysosomes (LAMP1)
Quantify co-localization coefficients to track intracellular trafficking
pH-sensitive fluorescent dye quenching:
Label TF antibody with both pH-sensitive and pH-stable fluorophores
Monitor fluorescence ratio changes as antibody traffics to acidic compartments
Calculate internalization kinetics based on fluorescence quenching rates
Research has shown that TF demonstrates significantly faster internalization compared to other targeted receptors, with approximately 70-80% internalization within 60 minutes, compared to only 30-40% for receptors like EGFR under similar conditions .
| Challenge | Potential Causes | Solutions |
|---|---|---|
| Low signal intensity | Insufficient antibody concentration, low target expression, photobleaching | Titrate antibody concentration, use positive control samples, minimize light exposure, consider signal amplification systems |
| High background | Non-specific binding, autofluorescence, inadequate blocking | Use proper blocking reagents (5-10% serum), include appropriate isotype controls, incorporate autofluorescence quenching steps |
| Inconsistent staining patterns | Heterogeneous TF expression, variable fixation/permeabilization | Standardize sample preparation protocols, optimize fixative concentration and timing, ensure uniform reagent exposure |
| Loss of binding activity | Antibody denaturation, improper storage, excessive conjugation | Avoid freeze-thaw cycles, maintain cold chain, verify F/P ratio is within optimal range (typically 3-7 FITC molecules per antibody) |
| Spectral overlap in multi-color experiments | FITC emission spectrum overlapping with other fluorophores | Apply proper compensation controls, consider alternative fluorophores for multi-parameter analysis |
When troubleshooting internalization studies specifically, researchers should validate that their TF antibody maintains the capacity to induce efficient inhibition of TF:FVIIa-dependent intracellular signaling while preserving rapid target internalization characteristics .
For Flow Cytometry Optimization:
Perform antibody titration to determine optimal concentration for your specific cell type
Adjust cell density to ensure proper antibody-to-cell ratio (typically 1 × 10^6 cells/ml)
Optimize incubation time and temperature based on internalization kinetics
Include fluorescence-minus-one (FMO) controls to set accurate gates
For fixed samples, evaluate different fixation protocols to preserve both antigen epitopes and FITC fluorescence
For Immunofluorescence Microscopy:
Test different fixation methods (4% paraformaldehyde, methanol, or acetone) to determine optimal epitope preservation
Evaluate permeabilization reagents (0.1-0.5% Triton X-100, 0.05-0.2% saponin) and their impact on signal intensity
Incorporate nuclear counterstains that don't overlap with FITC spectrum (DAPI or Hoechst)
Mount slides with anti-fade reagents containing DABCO or similar compounds to minimize photobleaching
When imaging, begin with lower exposure settings and adjust incrementally to avoid photobleaching
For FACS-based Sorting Applications:
Use viability dyes to exclude dead cells that can bind antibodies non-specifically
Maintain samples at 4°C prior to sorting to minimize internalization
Include DNase I (10-50 μg/ml) in buffers to prevent cell clumping
Use preservative-free formulations for live cell sorting applications
Optimize sorting gates based on positive control samples with known TF expression levels
Research groups have successfully applied these optimization approaches to achieve high affinity binding of anti-TF-conjugated constructs to TF-expressing cancer cell lines, with binding affinity comparable to that of unconjugated anti-human TF F(ab')2 fragments .
The application of TF Antibody, FITC conjugated to study tumor microenvironment interactions represents an emerging research direction with several methodological approaches:
Multi-parameter flow cytometry: Combining TF-FITC with markers for different cell populations allows researchers to characterize TF expression across various cell types in the tumor microenvironment, including cancer cells, endothelial cells, tumor-associated macrophages, and cancer-associated fibroblasts.
Intravital microscopy: Using TF Antibody, FITC conjugated in combination with window chamber models enables real-time visualization of TF-expressing cells within the tumor microenvironment and their interactions with other cellular components.
3D organoid co-culture systems: Co-culturing TF-expressing cancer cells with stromal components and tracking with TF-FITC antibody allows for the study of TF's role in modulating cancer-stroma interactions.
Spatial transcriptomics correlation: Combining TF-FITC immunofluorescence with spatial transcriptomics techniques provides insights into how TF expression correlates with specific gene expression programs in the tumor microenvironment.
Recent research suggests that TF expression influences tumor angiogenesis and stromal fibrosis, making these techniques valuable for studying how TF contributes to remodeling the tumor microenvironment .
Current experimental approaches for developing advanced TF-targeted therapeutics include:
Bispecific antibody development: Engineering bispecific antibodies that simultaneously target TF and immune effector cells (T cells, NK cells) to enhance anti-tumor immune responses while blocking TF signaling.
Novel payload conjugation strategies: Exploring new cytotoxic payloads beyond traditional auristatins, including:
DNA-damaging agents
RNA polymerase inhibitors
Immunomodulatory compounds
Combination therapy optimization: Systematic evaluation of TF-targeted antibodies or ADCs in combination with:
Immune checkpoint inhibitors
Conventional chemotherapy
Radiation therapy
Anti-angiogenic agents
TF-targeted nanoparticle systems: Development of more sophisticated delivery systems such as:
Anti-TF-conjugated polymeric micelles incorporating epirubicin or other anticancer agents
Liposomal formulations with anti-TF targeting
Biodegradable nanoparticles with controlled release properties
Research has demonstrated promising results with anti-TF-NC-6300, consisting of epirubicin-incorporating micelles conjugated with F(ab')2 fragments of anti-TF antibody. This approach achieved enhanced antitumor effects that were independent of tumor accumulation but dependent on selective intratumor localization and preferential internalization into high TF tumor cells .
Integration of TF Antibody, FITC conjugated into multiplexed imaging systems requires careful consideration of several methodological aspects:
Spectral unmixing protocols:
Implement linear unmixing algorithms to separate FITC signal from autofluorescence and other fluorophores
Acquire single-color controls for each fluorophore in the panel
Use reference spectra libraries to improve unmixing accuracy
Panel design for multiplexed immunofluorescence:
Position FITC in appropriate channel based on expected TF expression level
Combine with markers for tumor cells (cytokeratins, EpCAM), immune cells (CD45, CD3, CD8), and vasculature (CD31)
Include proliferation markers (Ki67) and functional markers (cleaved caspase-3)
Image acquisition optimization:
Standardize exposure times and laser power settings
Implement flat-field correction to account for illumination non-uniformity
Use appropriate z-stack sampling for volumetric analysis
Quantitative image analysis workflows:
Develop cell segmentation algorithms for accurate identification of TF-positive cells
Implement spatial analysis tools to quantify TF-expressing cell distribution
Apply machine learning approaches for pattern recognition and phenotype classification
These advanced multiplexed imaging approaches can help researchers better understand the relationship between TF expression, tumor progression, and response to therapy, particularly in challenging cancers like triple-negative breast cancer and pancreatic adenocarcinoma where TF is frequently overexpressed .