FLT3 is a receptor tyrosine kinase critical for hematopoiesis and is frequently mutated in acute myeloid leukemia (AML), particularly the internal tandem duplication (ITD) mutation, which confers poor prognosis . The FLT3 antibody, when conjugated with fluorescein isothiocyanate (FITC), enables fluorescent detection of FLT3 expression via flow cytometry or immunofluorescence, aiding in diagnostic and research workflows.
The SF1.340 antibody (Santa Cruz Biotechnology) is a mouse monoclonal IgG1 κ antibody that specifically binds to human FLT3. Its FITC-conjugated form is optimized for detecting FLT3 on hematopoietic cells, including leukemic blasts, in clinical and preclinical settings .
Epitope Specificity: The SF1.340 antibody targets the extracellular domain of FLT3, enabling detection of surface-expressed FLT3 on cells . This contrasts with therapeutic antibodies like 20D9h3, which target domain 2 for drug conjugation .
FITC Conjugation: FITC is covalently linked to the antibody’s lysine residues, preserving its binding affinity while enabling fluorescence detection. This conjugation enhances its utility in flow cytometry for quantifying FLT3 expression levels .
Cross-Reactivity: The antibody exhibits minimal binding to FLT3 paralogues (e.g., VEGFR, PDGFRα) or orthologues (e.g., cynomolgus monkey FLT3), ensuring specificity .
The FLT3-FITC antibody is widely used to assess FLT3 expression in AML patient samples. For example, studies employing this antibody have demonstrated:
AML Diagnosis: Elevated FLT3 expression correlates with ITD mutations, aiding in stratifying patients for FLT3-targeted therapies .
Leukemic Stem Cell (LSC) Detection: Dual-targeting strategies (e.g., FLT3 + CD99) using FITC-conjugated antibodies have shown enhanced specificity for LSCs in AML models .
In preclinical models, the antibody has been used to localize FLT3 expression in bone marrow biopsies and leukemic cell lines, providing insights into disease progression .
Prognostic Value: Quantitative FLT3 expression analysis via flow cytometry (using FLT3-FITC) predicts treatment outcomes in AML. Patients with high FLT3 expression often require more aggressive therapies .
Therapeutic Monitoring: The antibody facilitates monitoring of FLT3-targeted therapies, such as FLT3 inhibitors (e.g., midostaurin), by tracking receptor downregulation .
FLT3 (FMS-related tyrosine kinase receptor 3) is a class III receptor tyrosine kinase predominantly expressed on leukemic cells of the myeloid and lymphoid lineage. It has emerged as a significant therapeutic target in hematologic malignancies due to its frequent overexpression or constitutive activation in acute myeloid leukemia (AML) . FLT3 is often affected by internal tandem duplication (ITD) and tyrosine kinase domain (TKD) mutations, which contribute to leukemogenesis and are associated with poor clinical outcomes . The receptor's relatively specific expression pattern on leukemic cells makes it an attractive target for antibody-based therapies, allowing for selective targeting of malignant cells while potentially sparing normal tissues .
FITC (fluorescein isothiocyanate) conjugation provides a direct fluorescent labeling approach that enables visualization and quantification of FLT3 expression through flow cytometry, fluorescence microscopy, and other fluorescence-based detection methods. The conjugation process involves covalent binding of the fluorescent dye to the antibody without significantly altering binding properties when optimally performed. When working with FITC-conjugated FLT3 antibodies, researchers should be aware that:
Optimal excitation occurs at approximately 495 nm with emission at 519 nm
FITC conjugation allows direct detection without secondary antibodies
The conjugation ratio (number of FITC molecules per antibody) affects both signal intensity and potential interference with antigen binding
Based on research findings, FLT3 expression varies significantly across different cell types:
When designing experiments, it's critical to account for these expression differences when setting detection thresholds and interpreting results .
For reliable FLT3 detection using FITC-conjugated antibodies, follow these methodological guidelines:
Sample preparation:
Use freshly isolated cells when possible
For peripheral blood or bone marrow, perform red blood cell lysis using ammonium chloride-based buffers
Maintain cells at 4°C throughout processing to prevent receptor internalization
Staining protocol:
Use 5-10 μL of antibody per 10⁶ cells (adjust based on titration experiments)
Incubate for 30 minutes at 4°C in the dark
Wash cells twice with phosphate-buffered saline containing 2% fetal bovine serum
Fix cells with 1-2% paraformaldehyde if analysis will be delayed
Controls and validation:
Most commercially available FLT3 antibodies, including FITC-conjugated versions, bind to epitopes present in both wild-type and mutated FLT3, making direct distinction challenging. Alternative approaches include:
Complementary molecular testing (PCR) to identify FLT3-ITD or FLT3-TKD mutations
Analysis of downstream signaling molecules specific to constitutively activated FLT3
Differential binding patterns of epitope-specific antibodies
For example, antibodies like 4G8 (binding to domain 4) and BV10 (binding to domain 2) have different binding characteristics that can be leveraged for comparative analysis . Research has shown that antibodies binding to membrane-proximal domains may show different internalization kinetics between wild-type and mutated FLT3 .
Bispecific FLT3 antibodies represent an advanced approach for targeted leukemia therapy. These antibodies simultaneously bind to:
FLT3 on leukemic cells (targeting component)
Immune effector molecules like CD3 on T-cells (effector component)
The mechanism involves:
Formation of an immunological synapse between leukemic cells and effector T-cells
T-cell activation independent of antigen recognition or MHC restriction
Induction of cytotoxic activity against FLT3-positive cells
For example, the 4G8 X UCHT1 Fabsc-antibody format shows superior properties compared to traditional bispecific single chain (bssc) formats, including:
Higher affinity to the target antigen FLT3
Better production yield by transfected cells
Reduced aggregation tendency
Effective T-cell activation and efficient killing of leukemic blasts in primary PBMC cultures of AML patients
FLT3-targeting ADCs provide several advantages over conventional therapeutic approaches:
Enhanced potency through delivery of cytotoxic payloads directly to FLT3-expressing cells
Activity independent of FLT3 mutation status, overcoming resistance mechanisms
Reduced off-target effects compared to traditional chemotherapy
Recent development of the 20D9-ADC utilizing P5 conjugation technology has demonstrated:
Potent cytotoxicity against cells expressing both wild-type FLT3 and FLT3-ITD
Significant tumor reduction and durable complete remission in AML xenograft models
Minimal hematotoxicity at therapeutically relevant concentrations
Strong synergistic effects when combined with tyrosine kinase inhibitors like midostaurin
These properties make ADCs particularly promising for patients with poor prognosis despite treatment with FLT3 inhibitors .
The Fabsc (Fab-single chain) format represents an optimization over the bispecific single chain (bssc) format with several measurable advantages:
| Parameter | Fabsc Format | bssc Format | Advantage |
|---|---|---|---|
| FLT3 binding affinity | Higher | Moderately reduced | Better target engagement |
| Production yield | Superior | Lower | Improved manufacturing |
| Aggregation tendency | <10% | Higher | Better stability and safety |
| Serum half-life | ~3 hours (after 1 day) | ~3 hours (after 1 day) | Similar pharmacokinetics |
| T-cell activation | Effective | Effective | Comparable efficacy |
| Target cell killing | Comparable | Comparable | Similar cytotoxicity |
| Cytokine release | Lower IL-2 release | Higher IL-2 release | Potentially improved safety profile |
The Fabsc format's superior properties are attributed to its more physiologic antibody structure that resembles normal antibody architecture more closely than the bssc format .
Several factors affect FLT3 detection variability in clinical specimens:
Heterogeneous FLT3 expression levels:
AML patients show FLT3 expression ranging from 400 to 3,300 molecules per cell
Some samples may have expression below detection thresholds
Technical considerations:
Sample handling and processing time affect receptor integrity
Antibody clone selection impacts epitope accessibility
Flow cytometer settings and compensation parameters influence detection sensitivity
Biological factors:
Non-specific binding can lead to false positive results and misinterpretation of data. To address this issue:
Identification methods:
Use isotype-matched control antibodies conjugated to the same fluorochrome
Perform blocking experiments with excess unconjugated antibody
Test antibody binding on known FLT3-negative cell lines
Minimization strategies:
Optimize antibody concentrations through titration experiments
Include Fc receptor blocking reagents in staining buffer
Implement stringent gating strategies based on fluorescence minus one (FMO) controls
Use alternative clones targeting different epitopes for confirmation
Research has identified that monocytes expressing trace amounts of FLT3 (300-600 molecules per cell) can cause off-target activation of PBMC cultures, which can be specifically blocked by depletion of CD14+ cells or excess parental FLT3 antibody .
When assessing FLT3 antibodies for therapeutic applications, these controls are critical:
Target specificity controls:
Antibodies targeting unrelated antigens (e.g., 9.2.27 antibody targeting CSPG4)
Excess unconjugated FLT3 antibodies to block specific binding
Functional validation controls:
Testing on FLT3-negative cells to confirm specificity
Comparison with established therapeutic antibodies
Assessment in the presence and absence of target cells to determine off-target activation
Patient-derived controls:
Studies have demonstrated that rigorous controls can identify important characteristics, such as the specific blockade of PBMC activation after depletion of monocytes with magnetic beads carrying CD14 and CD33, confirming the specificity of therapeutic antibodies .
Combination strategies involving FLT3 antibodies show promising results:
FLT3 antibodies with tyrosine kinase inhibitors:
Bispecific antibodies with immune checkpoint inhibitors:
Potential to overcome immune suppression in the tumor microenvironment
May enhance T-cell mediated killing of leukemic cells
ADCs with conventional chemotherapy:
Sequential or concurrent administration strategies
Potential for dose reduction of conventional agents
These approaches represent the frontier of FLT3-directed immunotherapy research .
Recent technological advances in FLT3 antibody development include:
Novel conjugation technologies:
Advanced antibody formats:
Emerging combination approaches:
These innovations aim to overcome limitations of current therapies and improve outcomes for patients with FLT3-positive malignancies.