While "FAP3 Antibody" is not a widely recognized term in scientific literature, antibodies targeting Fibroblast Activation Protein (FAP) are well-documented and studied for their potential in cancer therapy and diagnostics . FAP, a type II integral serine protease, is highly expressed on cancer-associated fibroblasts (CAFs) in the tumor stroma, playing a crucial role in tumor growth, invasion, metastasis, and immunosuppression . Therefore, antibodies against FAP are being explored as a means to target CAFs and disrupt their pro-tumorigenic activities .
FAP antibodies are designed to specifically bind to the fibroblast activation protein . FAP is a membrane protease that is highly expressed by cancer-associated fibroblasts (CAFs) . It is involved in extracellular matrix remodeling, intracellular signaling, angiogenesis, epithelial-to-mesenchymal transition, and immunosuppression, all of which contribute to tumor development and metastasis .
Recombinant Monoclonal Antibodies: These are produced using recombinant DNA technology, ensuring high specificity and reproducibility . An example is the anti-FAP antibody [73.3], which is manufactured using AbAb’s Recombinant Platform with variable regions from the hybridoma 73.3 .
Single-Chain Variable Fragments (scFvs): These are engineered antibody fragments consisting of the variable regions of the heavy and light chains, linked by a short peptide . scFvs can be designed to inhibit FAP function and disrupt its role in tumor invasion and metastasis .
Humanized Monoclonal Antibodies: These are developed to reduce immunogenicity in humans, making them suitable for therapeutic applications .
FAP antibodies have a wide range of applications of interest in cancer research and therapy :
Targeted Drug Delivery: FAP antibodies can be used to deliver cytotoxic drugs or other therapeutic agents specifically to the tumor microenvironment, sparing healthy tissues .
CAR-T Cell Therapy: FAP-specific CAR-T cells (chimeric antigen receptor T-cells) can be engineered to target and kill FAP-expressing cells in the tumor stroma . These cells have shown promising results in preclinical models, significantly reducing tumor growth without significant toxicity .
Tumor Imaging: FAP antibodies labeled with imaging agents can be used to visualize and monitor tumors, as well as assess the effectiveness of cancer therapies .
Inhibitory Antibodies: Some FAP antibodies can inhibit the enzymatic activity of FAP, reducing its ability to promote tumor growth and metastasis . An inhibitory scFv antibody, named E3, has been shown to attenuate FAP cleavage activity .
Inhibition of FAP Function: An scFv antibody (E3) was identified that could attenuate 35% of FAP cleavage of a fluorescent substrate . A mutant E3 scFv with higher affinity and enhanced inhibitory effect on FAP enzyme activity was also developed .
Reduction of Fibronectin Fiber Orientation: The application of inhibitory anti-FAP scFvs significantly affected the formation of 3-dimensional FAP-positive cell matrix, reducing fibronectin fiber orientation .
Enhanced Anti-tumor Response: Anti-FAP-CAR T cells demonstrated enhanced antitumor response when combined with a tumor vaccine in mouse models .
Immunogenicity and Target Cell Lysis: A replication-defective adenovirus (Ad)-based vaccine expressing FAP induced robust FAP-specific B and T cell responses in mice, mediating specific target cell lysis .
Autoimmune Disorder Identification: Anti-FGFR3 antibodies (Abs) were found to identify a subgroup of patients with sensory neuropathy (SN) in whom an underlying autoimmune disorder is suspected .
Fibroblast activation protein (FAP), also known as seprase, is a cell surface, type II integral transmembrane serine protease encoded by the Fap gene. It exhibits gelatinase, endopeptidase, and potentially collagenase activity in vitro. FAP has significant research importance due to its selective expression pattern - it is highly expressed on mesenchymal cells during embryogenesis but becomes repressed shortly after birth .
FAP expression becomes up-regulated on activated fibroblasts in conditions associated with matrix remodeling, including:
Wound healing processes
Fibrotic tissue disorders
Rheumatoid arthritis and osteoarthritis
Liver disease
Inflammatory bowel diseases
This specific expression pattern makes FAP a valuable target for research into disease mechanisms and potential therapeutic interventions, particularly in cancer and fibrosis-related conditions.
FAP3 antibodies target a specific epitope region (731-740 aa) of the FAP protein that has demonstrated strong immunogenicity in research settings . The selection of FAP3 as a target region resulted from systematic epitope screening processes that evaluated multiple candidate peptides for their ability to generate robust antibody responses.
When compared to other FAP-targeting antibodies:
Epitope specificity: FAP3 antibodies recognize a specific C-terminal region of the protein, whereas other antibodies may target different epitopes like FAP#1 (186-195 aa) or FAP#2 (238-247 aa) .
Functional characteristics: In experimental studies, antibodies against the FAP3 epitope demonstrated:
Cross-reactivity profile: FAP3 antibodies show minimal cross-reactivity with the structurally similar DPPIV protein, which shares 51% sequence identity with FAP, making them more selective for research applications .
Researchers can employ multiple methodological approaches to detect FAP expression using anti-FAP antibodies:
Sample preparation: Tissue homogenates or cell lysates typically require standard protein extraction protocols
Antibody concentration: Generally used at 1:500-1:1000 dilution depending on expression levels
Expected molecular weight: ~95 kDa for monomeric FAP; ~170 kDa for homodimeric active FAP
Antigen retrieval: Microwave method using 10 mmol/L citrate buffer (pH 6.0), high temperature for 5 minutes followed by medium temperature for 15 minutes
Visualization systems: Both DAB-based chromogenic and fluorescent secondary detection systems are compatible
Cellular localization: Primarily membrane-localized with some cytoplasmic distribution
Cell preparation: Standard protocols for surface staining (non-permeabilized)
Controls: FAP-null cells as negative control; FAP-expressing 3T3 cells as positive control
Applications: Particularly useful for identifying FAP+ cells in lung tissue and tumor specimens
Starting material: Cell or tissue lysates (lung homogenate has been successfully used)
Elution conditions: Standard IP protocols are applicable
Verification: Follow-up Western blot analysis to confirm specificity
FAP3 antibodies have demonstrated significant potential in therapeutic cancer research through several specialized applications:
The single-chain variable fragment (scFv) from anti-FAP antibodies can be incorporated into second-generation retroviral CARs containing CD8 stalk, human CD3ζ and 4-1BB domains
FAP-CAR-T cells generated through this approach demonstrate specific killing of FAP-expressing target cells
In established tumor models, treatment with these FAP-CAR-T cells significantly reduced tumor growth by 35-50%
Combination therapy with tumor vaccines showed enhanced antitumor responses beyond either treatment alone
FAP3 peptide (731-740 aa) conjugated to keyhole limpet hemocyanin (KLH) carrier protein
Co-administration with cytosine-phosphate-guanine (CpG) K3 adjuvant to promote Th1-directed immune responses
Vaccination schedule: Initial administration at 10 weeks of age followed by a booster at 12 weeks
This approach induces both IgG1 and IgG2 antibodies capable of mediating both ADCC and CDC, critical for effective elimination of FAP-expressing cells
FAP3 antibodies can potentially reduce the immunosuppressive influence of cancer-associated fibroblasts (CAFs)
Experimental designs should include assessment of tumor-infiltrating lymphocytes and inflammatory cytokine profiles following treatment
No significant systemic toxicity has been observed in preclinical models despite successful targeting of FAP-positive cells
When utilizing FAP antibodies in cardiac fibrosis research, investigators should consider these key methodological approaches:
Angiotensin II and phenylephrine (AngII/PE) continuous administration model:
Histological analysis: Masson's trichrome staining for quantification of fibrotic areas
Immunohistochemical evaluation: FAP staining to identify myofibroblast accumulation
Quantitative assessment metrics: Percentage of fibrotic area (control: 8.62±4.79% vs. FAP-vaccinated: 3.45±1.11%) and FAP-positive cell density (control: 7327±1741 vs. FAP-vaccinated: 4077±1746 cells/mm²)
IgG deposits in cardiac tissues should be assessed via immunohistochemistry
Evaluation of macrophage infiltration using F4/80 staining
These measurements help correlate therapeutic effect with antibody-mediated mechanisms
Timing is critical: While chronic FAP-positive cell elimination is beneficial in fibrosis models, transient activation of myofibroblasts plays an important role in acute injury repair
Experimental designs should include both acute (myocardial infarction) and chronic stress models to fully characterize antibody effects
Inflammatory markers should be monitored to detect potential antibody-dependent cell cytotoxicity in off-target tissues
When encountering specificity challenges with FAP antibodies, researchers should implement the following troubleshooting methodology:
DPPIV cross-reactivity testing: Due to 51% sequence homology between FAP and DPPIV, antibodies should be validated against both proteins
Western blot analysis: Compare bands from FAP-positive, FAP-negative, and DPPIV-positive samples
Cross-adsorption experiments: Pre-incubation with recombinant DPPIV protein to identify non-specific binding
Positive controls: FAP-expressing 3T3 cells or transfected cell lines with verified FAP expression
Negative controls: FAP-null cells or tissues
Isotype controls: Matched isotype antibodies with unknown specificity (e.g., Ab00178-23.0 for staining controls)
Validation Method | Expected Results | Troubleshooting Steps |
---|---|---|
Western Blot | ~95 kDa band for monomeric FAP; ~170 kDa for homodimer | If multiple bands present, evaluate sample preparation, reducing conditions, and glycosylation status |
Immunofluorescence | Membrane and cytoplasmic staining pattern | Background staining may indicate insufficient blocking or non-specific binding |
ELISA | Specific binding to FAP-BSA and rmFAP | Cross-adsorption with related proteins to confirm specificity |
Flow Cytometry | Clear separation between positive and negative populations | Titrate antibody concentration; include FMO controls |
Epitope retrieval optimization: Adjust pH, buffer composition, and heating parameters
Antibody concentration titration: Test multiple dilutions to find optimal signal-to-noise ratio
Buffer composition adjustments: Modify blocking reagents and washing buffers to reduce non-specific binding
FAP antibodies provide powerful tools for investigating the complex tumor microenvironment through several methodological approaches:
Multiplexed immunohistochemistry/immunofluorescence: Co-staining with FAP antibodies and other CAF markers (α-SMA, PDGFRβ) allows identification of distinct CAF subpopulations
Spatial distribution analysis: Correlate FAP+ cell localization with tumor invasive front, hypoxic regions, or vascular structures
Digital pathology approaches: Quantitative analysis of FAP+ cell density in different tumor regions provides insights into heterogeneity of the stromal response
Flow cytometry-based isolation: FAP antibodies enable isolation of viable CAFs for ex vivo functional studies
Secretome analysis: Compare protein secretion profiles between FAP+ and FAP- stromal cells
Co-culture systems: Evaluate how FAP+ CAFs influence cancer cell growth, migration, and therapy resistance
Intravital microscopy: Fluorescently labeled FAP antibodies can track CAF dynamics in real-time in appropriate window chamber models
Imaging mass cytometry: Combine FAP antibodies with metal-tagged antibodies against numerous other markers for comprehensive tumor microenvironment mapping
These approaches reveal temporal and spatial relationships between FAP+ cells and other components of the tumor microenvironment
Correlation studies between FAP+ cell density and clinical outcomes in patient samples
Assessment of FAP expression changes in response to standard therapies
Evaluation of heterotypic signaling between FAP+ CAFs and immune cell populations within the tumor microenvironment
Researchers developing FAP antibody-based targeted therapies should consider these critical methodological aspects:
Full IgG antibodies: Maintain effector functions (ADCC, CDC) but have limited tumor penetration
Antibody fragments (Fab, scFv): Improved tissue penetration but shorter half-life
Bispecific formats: Can engage immune effector cells while binding FAP+ targets
Selection should be based on intended mechanism of action and therapeutic context
Comprehensive tissue cross-reactivity studies: Although FAP expression is largely restricted to activated fibroblasts in adults, thorough screening across normal tissues is essential
Assessment in injury models: As FAP is upregulated during wound healing, potential interference with normal tissue repair should be evaluated
Safety data from preclinical studies suggests minimal systemic or organ-specific inflammation despite successful targeting of FAP+ cells
Antibody-drug conjugates (ADCs): Selection of linker chemistry and cytotoxic payload must balance stability, potency, and bystander effect
Radioimmunotherapy: Half-life matching between antibody and radioisotope is critical for optimal therapeutic index
Immunomodulatory approaches: FAP-targeting vaccines with appropriate adjuvants (e.g., CpG K3) can induce endogenous antibody responses with effective ADCC and CDC capabilities
FAP-targeted approaches have shown enhanced efficacy when combined with:
Tumor vaccines
Immune checkpoint inhibitors
Conventional chemotherapy
Experimental designs should include careful sequencing and timing of combination treatments to optimize therapeutic synergy
The performance of FAP antibodies varies across tissue types and pathological conditions, with important methodological considerations:
Tissue Type | FAP Expression Pattern | Methodological Considerations |
---|---|---|
Cardiac Tissue | Localized to fibrotic areas; increased in pressure overload | Autofluorescence may interfere with immunofluorescence; consider chromogenic detection |
Lung Tissue | Present in fibroblastic foci in fibrosis; also on specific macrophage populations | Background staining can be problematic; careful titration required |
Tumor Stroma | Heterogeneous expression on CAFs; spatial variation within tumors | Multiplexed staining recommended to distinguish FAP+ cell subpopulations |
Liver | Activated hepatic stellate cells in fibrosis express FAP | Endogenous peroxidase activity must be thoroughly blocked |
Skin | Dermal fibroblasts during wound healing | Epitope retrieval protocols may need optimization |
Cancer Detection:
Fibrosis Assessment:
Inflammatory Conditions: