FLT3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we are able to ship the products within 1-3 business days upon receipt of your order. Delivery time may vary depending on the method of purchase and location. For specific delivery timeframes, please consult your local distributor.
Synonyms
CD 135 antibody; CD135 antibody; CD135 antigen antibody; Fetal liver kinase 2 antibody; FL cytokine receptor antibody; Flk 2 antibody; Flk2 antibody; Flt 3 antibody; FLT-3 antibody; Flt3 antibody; FLT3_HUMAN antibody; FMS like tyrosine kinase 3 antibody; Fms related tyrosine kinase 3 antibody; Fms-like tyrosine kinase 3 antibody; Growth factor receptor tyrosine kinase type III antibody; Ly-72 antibody; OTTHUMP0000004234 antibody; Receptor type tyrosine protein kinase FLT3 antibody; Stem cell tyrosine kinase 1 antibody; Stk 1 antibody; STK-1 antibody; Stk1 antibody; Tyrosine protein kinase receptor FLT3 antibody; Tyrosine-protein kinase receptor FLT3 antibody
Target Names
Uniprot No.

Target Background

Function
FLT3 is a tyrosine-protein kinase that functions as a cell-surface receptor for the cytokine FLT3LG. It plays a crucial role in regulating differentiation, proliferation, and survival of hematopoietic progenitor cells and dendritic cells. Upon activation, FLT3 promotes the phosphorylation of SHC1 and AKT1, leading to activation of the downstream effector MTOR. It also stimulates RAS signaling and the phosphorylation of downstream kinases, including MAPK1/ERK2 and/or MAPK3/ERK1. Additionally, FLT3 promotes the phosphorylation of FES, FER, PTPN6/SHP, PTPN11/SHP-2, PLCG1, and STAT5A and/or STAT5B. While activation of wild-type FLT3 results in only marginal activation of STAT5A or STAT5B, mutations that induce constitutive kinase activity promote cell proliferation and resistance to apoptosis through the activation of multiple signaling pathways.
Gene References Into Functions
  1. Multivariate Cox's proportional hazards regression analyses indicated that high OCT4 mRNA expression was an independent predictor of shorter EFS and OS in AML patients. Notably, OCT4 correlated with the presence of CK, FLT3-ITD mutation, and poorer risk stratification, suggesting its potential as a biomarker for predicting unfavorable prognosis in AML patients. PMID: 29950146
  2. The findings demonstrate that DNMT3A mutations alone do not impact clinical outcomes of AML patients undergoing allogeneic HSCT. However, when these mutations occur alongside FLT3-ITD mutations, the OS was significantly reduced (5-year OS 0% for DNMT3A R882mut/FLT3-ITDpos patients vs. 62% DNMT3A R882wt/FLT3-ITDneg, p=0.025), and the relapse rate increased. PMID: 29786546
  3. RIPK3-dependent cell death and inflammasome activation in FLT3-internal-tandem-duplication-expressing leukemia-initiating cells PMID: 27517160
  4. The study findings suggest that FLT3 ITD mutations could be an indicator of poor prognosis in APL, and these patients may benefit from more intensive therapy according to current guidelines. PMID: 29251252
  5. Low FLT3 expression has been associated with Pancreatic ductal adenocarcinoma. PMID: 30275197
  6. DNMT3A R882 mutation plays a significant role in the prognosis and clinical outcomes of CN-AML patients, both in the presence and absence of NPM1 and FLT3 mutations. PMID: 29079128
  7. The FLT3 inhibitor AC220 significantly inhibited glutamine flux into the antioxidant factor glutathione, likely due to defective glutamine import. PMID: 28947392
  8. Mutations in the FLT3 gene have been associated with Acute Myeloid Leukemia. PMID: 29530994
  9. Acute myeloid leukemia harboring internal tandem duplication of FMS-like tyrosine kinase 3 (AML(FLT3-ITD)) is often associated with a poor prognosis. PMID: 29330746
  10. The influence of FLT3-ITD diversity on response to induction chemotherapy in patients with acute myeloid leukemia has been explored. PMID: 28034991
  11. The study findings indicated that FLT3 overexpression is a potential risk factor in leukemia. PMID: 29257272
  12. This study evaluated FLT3 and NPM1 mutations in adult Iranian patients with de novo cytogenetically normal acute myeloid leukemia and investigated their correlations with clinical and laboratory parameters. PMID: 28294102
  13. FLT3 and FLT3-ITD can directly bind and selectively phosphorylate p27kip1 on tyrosine residue 88 in acute myeloid leukemia. Inhibition of FLT3-ITD in cell lines led to a significant reduction in p27 tyrosine 88 phosphorylation, resulting in increased p27 levels and cell cycle arrest. PMID: 28522571
  14. The study demonstrated that FLT3 can be targeted by FLT3-CAR T cells for the treatment of FLT3(+) AML. FLT3-CAR T cells may provide a novel immunotherapeutic approach for AML patients. PMID: 28496177
  15. High BCRP mRNA expression calculated with Pfaffl's rule, along with FLT3-ITD, are independent poor risk factors in adult patients with AML and intermediate or normal karyotype. PMID: 28618074
  16. A novel juxtamembrane deletion mutation in FLT3 exhibits a dominant negative effect on the wild-type FLT3 receptor. PMID: 27346558
  17. While FLT3 cell-surface expression did not vary based on FLT3 mutational status, high FLT3 expression was strongly associated with KMT2A rearrangements. The study found that FLT3 cell surface expression did not have prognostic significance in pediatric Acute Myeloid Leukemia. PMID: 28108543
  18. DNA mutational analysis in FLT3 in acute myeloid leukemia. PMID: 27071442
  19. The data confirms MLL-PTD and, to a lesser extent, FLT3-ITD as common events in +11 AML. However, the high mutation frequencies of U2AF1 and genes involved in methylation (DNMT3A, IDH2) have not been previously reported in +11 AML. PMID: 27435003
  20. The cytokine Fms-like tyrosine kinase 3 ligand is a significant regulator of hematopoiesis. Its receptor, Flt3, is expressed on myeloid, lymphoid, and dendritic cell progenitors and is considered an important growth and differentiation factor for several hematopoietic lineages. [review] PMID: 28538663
  21. FLT3 amplification in solid cancers is infrequently observed using targeted genomic profiling. Currently, FLT3 amplification does not appear to be an actionable target or a suitable biomarker for FLT3 inhibitor sensitivity. PMID: 27906677
  22. FLT3 plays a role in cytarabine transport by SLC29A1 in pediatric acute leukemia. PMID: 27391351
  23. Data suggests a pathway MYSM1/miR-150/FLT3 that inhibits proliferation of B1a cells, potentially involved in the pathogenesis of systemic lupus erythematosus (SLE). PMID: 27590507
  24. These findings confirm that FLT3-ITD-location influences disease biology and leads to changes in global gene expression. In this model, ITD-location alters proliferative capacity and sensitivity to FLT3-TKI-treatment in vivo. PMID: 26487272
  25. A decision analysis comparing allo-HCT vs chemotherapy in first complete remission for patients with cytogenetically intermediate-risk acute myeloid leukemia, considering the presence or absence of FLT3-ITD, NPM1, and CEBPA mutations, showed that allo-HCT was a preferred postremission strategy in patients with FLT3-ITD, while chemotherapy was favored in patients with biallelic CEBPA mutations. PMID: 27040395
  26. ATM/G6PD-driven redox metabolism promotes FLT3 inhibitor resistance in acute myeloid leukemia, which can be successfully reversed. PMID: 27791036
  27. Data suggests that escalated daunorubicin dosing may be appropriate for fms-like tyrosine kinase 3 (FLT3)-ITD mutated cases. PMID: 27268085
  28. Integrin alphavbeta3 plays a role in enhancing beta-catenin signaling in acute myeloid leukemia harboring Fms-like tyrosine kinase-3 internal tandem duplication mutations. PMID: 27248172
  29. This review explores the role of the most common form of FMS-like tyrosine kinase 3 (FLT3) mutation (internal tandem duplication) in acute myeloid leukemia. PMID: 28470536
  30. The present cohort study demonstrated that FLT3-ITD and DNMT3A R882 double mutation predicts poor prognosis in Chinese AML patients receiving chemotherapy or allo-HSCT treatment. PMID: 28616699
  31. While transient responses to FLT3 inhibitors are often observed in cases of disease relapse, the most promising approach is the use of FLT3 inhibitors either in combination with induction chemotherapy or as consolidation/maintenance therapy after allogeneic hematopoietic cell transplantation. PMID: 27775694
  32. This review focuses on three key areas in acute myeloid leukemia (AML) developmental therapeutics: FLT3 inhibitors, IDH (IDH1 and IDH2) inhibitors, and drugs that may be particularly beneficial in secondary AML. PMID: 28561688
  33. Concomitant monitoring of WT1 and FLT3-ITD expression in FLT3-ITD acute myeloid leukemia patients. PMID: 28211167
  34. FLT3/ITD are present at the leukemic stem cell level and may be a primary, rather than secondary, event in leukemogenesis. The oncogenic events of FLT3/ITD occur at a cell stage expressing CD123. PMID: 27465508
  35. Sorafenib may enable the cure of a proportion of very poor risk FLT3-internal tandem duplication-positive acute myeloid leukemia relapsing after allogeneic stem cell transplantation. PMID: 29055209
  36. Results provide evidence that mutations in the tyrosine kinase domain in FLT3 were found in 7% of Pakistani patients with acute myeloid leukemia. PMID: 27735988
  37. FLT3/ITD increases aerobic glycolysis through AKT-mediated upregulation of mitochondrial hexokinase (HK2). Inhibition of glycolysis preferentially causes severe ATP depletion and massive cell death in FLT3/ITD leukemia cells. PMID: 28194038
  38. The results indicate that CD4 expression and older age are adverse prognostic factors in wild-type NPM1, FLT3-ITD-negative CN-AML. PMID: 28318150
  39. FLT3 mutation has been associated with Metaplastic Breast Cancer. PMID: 27568101
  40. Collectively, a novel targeted therapeutic strategy has been developed, using FLT3L-guided miR-150-based nanoparticles, to effectively treat FLT3-overexpressing AML with high efficacy and minimal side effects. PMID: 27280396
  41. Y842 is critical for FLT3-mediated RAS/ERK signaling and cellular transformation. PMID: 28271164
  42. The value of FLT3-ITD allelic ratio in AML in risk assessment and evaluating prognosis. PMID: 27416910
  43. DOCK2 is a potential therapeutic target for novel AML treatments, as it regulates the survival of leukemia cells with elevated FLT3 activity and sensitizes FLT3/ITD leukemic cells to conventional antileukemic agents. PMID: 27748370
  44. Gedatolisib significantly extended survival of mice in a sorafenib-resistant acute myeloid leukemia (AML) patient-derived xenograft model. This suggests that aberrant activation of the PI3K/mTOR pathway in FLT3-ITD-dependent AML leads to resistance to drugs targeting FLT3. PMID: 26999641
  45. HHEX could potentially replace RUNX1 in cooperating with FLT3-ITD to induce Acute myeloid leukemia (AML). PMID: 28213513
  46. Mutated FLT3-ITD and JAK2 augment reactive oxygen species production and homologous recombination, shifting the cellular milieu towards illegitimate recombination. PMID: 28108507
  47. These data reveal a novel mechanism regulating acute myeloid leukemia cell death through ceramide-dependent mitophagy in response to FLT3-ITD targeting. PMID: 27540013
  48. MSI2 and FLT3 are significantly co-regulated in human AML. PMID: 28107692
  49. Sorafenib-resistant leukemia cells with a FLT3/ITD mutation are sensitive to glycolytic inhibitors. PMID: 27132990
  50. Factors that did not influence the relapse risk included: age, graft type, graft source, type of FLT3 mutation, or conditioning intensity. PMID: 28052408

Show More

Hide All

Database Links

HGNC: 3765

OMIM: 136351

KEGG: hsa:2322

STRING: 9606.ENSP00000241453

UniGene: Hs.507590

Involvement In Disease
Leukemia, acute myelogenous (AML)
Protein Families
Protein kinase superfamily, Tyr protein kinase family, CSF-1/PDGF receptor subfamily
Subcellular Location
Membrane; Single-pass type I membrane protein. Endoplasmic reticulum lumen. Note=Constitutively activated mutant forms with internal tandem duplications are less efficiently transported to the cell surface and a significant proportion is retained in an immature form in the endoplasmic reticulum lumen. The activated kinase is rapidly targeted for degradation.
Tissue Specificity
Detected in bone marrow, in hematopoietic stem cells, in myeloid progenitor cells and in granulocyte/macrophage progenitor cells (at protein level). Detected in bone marrow, liver, thymus, spleen and lymph node, and at low levels in kidney and pancreas. H

Q&A

What is FLT3 and why is it significant in hematological research?

FLT3 (FMS-like tyrosine kinase 3) is a receptor-tyrosine kinase that plays a crucial role in hematopoiesis and is expressed on leukemic cells of both myeloid and lymphoid lineages . Its significance stems from its involvement in the development and progression of leukemia, particularly acute myeloid leukemia (AML) . FLT3 stimulates the proliferation of early hematopoietic cells through activation of downstream signaling cascades that affect cell fate decisions . The receptor is particularly important as a therapeutic target because it is expressed on almost all AML blasts at levels generally higher than on normal bone marrow hematopoietic stem and progenitor cells (HSPCs) . FLT3 overexpression has been correlated with poor prognosis and reduced survival rates in leukemia patients, making it both a prognostic biomarker and a promising therapeutic target .

How do FLT3 antibodies differ from small molecule FLT3 inhibitors in research applications?

FLT3 antibodies and small molecule inhibitors represent distinct approaches to targeting FLT3 with different research applications:

FLT3 Antibodies:

  • Target the extracellular domains of FLT3, with different antibodies binding to specific domains (e.g., 4G8 targeting domain 4, BV10 targeting domain 2)

  • Enable detection and quantification of FLT3 expression through techniques like immunohistochemistry, flow cytometry, and immunofluorescence

  • Can be engineered as bispecific antibodies (e.g., FLT3 x CD3) to redirect T cells against leukemia cells

  • Function through mechanisms like antibody-dependent cellular cytotoxicity (ADCC) or direct recruitment of immune effector cells

Small Molecule Inhibitors:

  • Target the intracellular kinase domain of FLT3, inhibiting downstream signaling

  • Typically identified through structure-based virtual screening or high-throughput screening approaches

  • More easily penetrate cells to reach intracellular targets

  • Often designed to specifically inhibit mutant forms of FLT3 (such as FLT3/ITD or FLT3/D835Mt)

While both approaches target FLT3, antibodies are more useful for detection, quantification, and immune-based therapies, whereas small molecule inhibitors are primarily focused on disrupting kinase activity and downstream signaling pathways .

What are the key structural domains of FLT3 relevant to antibody targeting?

FLT3 contains distinct extracellular domains that serve as targets for different antibodies, each with potential implications for research and therapeutic applications:

  • Domain 1 and 2: These domains form part of the extracellular portion of FLT3. The BV10 antibody specifically targets domain 2 . Targeting these domains may affect ligand binding.

  • Domain 3: Part of the immunoglobulin-like structure of the extracellular region.

  • Domain 4: Located more proximal to the cell membrane, this domain is targeted by the 4G8 antibody . Antibodies targeting this domain have shown promising results in bispecific antibody constructs.

  • Transmembrane domain: Connects the extracellular and intracellular portions.

  • Intracellular tyrosine kinase domain: The site of activating mutations like FLT3/ITD and FLT3/D835Mt that are associated with poor prognosis in AML . This domain is the target for small molecule inhibitors rather than antibodies.

Research has shown that antibodies targeting different domains exhibit varying effects on receptor function and downstream signaling . Domain-specific targeting can be strategically selected based on the desired experimental outcome or therapeutic approach. For instance, domain 4-targeting antibodies like 4G8 have demonstrated superior efficacy in certain bispecific antibody formats compared to antibodies targeting other domains .

How can flow cytometry be optimized for FLT3 protein quantification in research samples?

Optimizing flow cytometry for FLT3 quantification requires careful consideration of several parameters:

Cell Line Selection for Controls and Calibration:

  • Select appropriate positive control cell lines such as EOL-1, which demonstrates consistent high FLT3 expression

  • Establish a calibration curve using cell populations with varying FLT3 expression levels (20%-120% of positive control cells)

  • Include quality control (QC) samples to ensure reproducibility across experiments

Antibody Parameters:

  • Determine optimal antibody concentration through titration experiments

  • Select antibody clones with high specificity and minimal background (e.g., the rabbit polyclonal antibody used in ab238610)

  • Consider using directly conjugated antibodies to reduce protocol complexity

Protocol Optimization:

  • Standardize cell density (typically 1×10^6 cells per sample)

  • Optimize incubation time and temperature for antibody binding

  • Establish consistent fixation and permeabilization protocols if intracellular epitopes are targeted

Validation and Quality Control:

  • Validate results against established methods like Western blotting

  • Ensure intra- and inter-day precision (%CV) of <20%

  • Maintain accuracy (% recovery) within 80%-120% range

  • Confirm linearity of calibration curves (r² > 0.99)

This optimized approach provides a practical, reliable, and economical method for quantifying FLT3 protein levels in research and clinical samples, offering advantages over more labor-intensive techniques like Western blotting .

What are the recommended protocols for using FLT3 antibodies in immunohistochemistry and immunofluorescence studies?

Immunohistochemistry (IHC-P) Protocol:

  • Sample Preparation:

    • Fix tissue samples in 4% formaldehyde or paraformaldehyde

    • Process and embed in paraffin

    • Section to 4-6 μm thickness

  • Antigen Retrieval:

    • Deparaffinize and rehydrate sections

    • Perform heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)

    • Allow sections to cool to room temperature

  • Blocking and Primary Antibody:

    • Block endogenous peroxidase activity with 3% H₂O₂

    • Block non-specific binding with 10% normal serum

    • Apply FLT3 antibody at optimized dilution (e.g., 1/100 dilution for ab238610)

    • Incubate overnight at 4°C in a humidified chamber

  • Detection and Visualization:

    • Apply appropriate secondary antibody

    • Develop signal using DAB or other chromogen

    • Counterstain with hematoxylin

    • Dehydrate, clear, and mount

Immunocytochemistry/Immunofluorescence (ICC/IF) Protocol:

  • Cell Preparation:

    • Culture cells on chamber slides or coverslips

    • Fix cells with 4% formaldehyde for 10-15 minutes at room temperature

    • Permeabilize using 0.2% Triton X-100 for 10 minutes

  • Blocking and Primary Antibody:

    • Block with 10% normal serum from the species of secondary antibody

    • Apply FLT3 antibody at optimized dilution (e.g., 1/266 dilution for ab238610 in ICC/IF)

    • Incubate overnight at 4°C

  • Secondary Antibody and Visualization:

    • Apply fluorophore-conjugated secondary antibody (e.g., Alexa Fluor 488-conjugated Goat Anti-Rabbit IgG)

    • Counterstain nuclei with DAPI

    • Mount with anti-fade mounting medium

  • Controls and Validation:

    • Include positive controls (e.g., MCF7 cells for FLT3 expression)

    • Include negative controls (primary antibody omission)

    • Document imaging parameters for reproducibility

These protocols can be adapted based on specific research requirements and sample types. Optimization of antibody concentration, incubation times, and antigen retrieval methods may be necessary for different tissue types or cell lines .

How can researchers accurately quantify FLT3 transcript levels in experimental samples?

Accurate quantification of FLT3 transcript levels is essential for understanding its role in normal and leukemic hematopoiesis. The following methodological approach is recommended:

Sample Preparation:

  • Extract total RNA from target cells using a high-quality RNA isolation kit

  • Assess RNA integrity (RIN score >7) using bioanalyzer or gel electrophoresis

  • Treat samples with DNase to eliminate genomic DNA contamination

RT-qPCR Method:

  • Perform reverse transcription using oligo(dT) primers or random hexamers

  • Design primers spanning exon-exon junctions to avoid genomic DNA amplification

  • Include at least two reference genes (e.g., GAPDH, β-actin) for normalization

  • Establish a standard curve using serial dilutions of plasmid containing FLT3 cDNA or a well-characterized sample

  • Report results as absolute copy numbers per μg RNA for cross-study comparison

Quality Control Measures:

  • Determine PCR efficiency (should be between 90-110%)

  • Verify primer specificity through melt curve analysis and/or sequencing

  • Include no-template and no-RT controls

  • Run technical triplicates for each sample

  • Ensure threshold cycle values fall within the linear range of the standard curve

Data Interpretation:

  • Normal mononuclear cells typically show lower FLT3 expression than AML samples

  • FLT3 overexpression is defined as >200,000 copies/μg RNA in AML samples without FLT3/ITD

  • Compare expression levels with clinical parameters for prognostic significance

  • Consider concurrent gene mutations when interpreting FLT3 expression data

This methodology has been validated in clinical studies and has demonstrated prognostic value in AML patients, particularly in distinguishing a novel disease entity in AML without FLT3 mutations that may still benefit from FLT3 inhibitor therapy .

How does FLT3 expression correlate with genetic mutations in AML samples?

FLT3 expression demonstrates significant correlations with various genetic mutations in AML, providing important insights for research and clinical assessment:

FLT3/ITD (Internal Tandem Duplication):

  • AML samples with FLT3/ITD mutations typically exhibit higher FLT3 expression levels compared to wild-type samples

  • This correlation suggests a potential positive feedback loop where the mutation may upregulate receptor expression

FLT3/D835Mt (Activation Loop Mutations):

  • Similar to FLT3/ITD, samples with D835 mutations also show elevated FLT3 expression

  • This indicates that activating mutations across different regions of FLT3 may influence transcriptional regulation

MLL-TD (Mixed Lineage Leukemia-Tandem Duplication):

  • High FLT3 expression is associated with MLL-TD mutations

  • Interestingly, the relationship between high FLT3 expression and MLL-TD is independent of FLT3 mutations, suggesting separate regulatory mechanisms

p53 and N-RAS Mutations:

  • Unlike the above mutations, p53 and N-RAS mutations do not show a clear correlation with FLT3 expression levels

  • This distinction highlights the specificity of the relationship between FLT3 expression and certain genetic alterations

These correlations have significant implications for leukemia research, particularly in understanding disease mechanisms and developing targeted therapies. Researchers should consider these relationships when designing experiments and interpreting results, as the combinatorial effect of FLT3 expression levels and specific mutations may influence cellular behavior and therapeutic responses .

What methodological approaches are used to develop and evaluate bispecific FLT3 x CD3 antibodies?

The development and evaluation of bispecific FLT3 x CD3 antibodies involves a systematic multi-step process:

Antibody Design and Construction:

  • Selection of parental antibodies targeting FLT3 and CD3

    • Evaluation of different antibody clones targeting distinct epitopes (e.g., 4G8 targeting domain 4 and BV10 targeting domain 2 of FLT3)

    • Assessment of CD3 antibodies like UCHT1, OKT3, and BMA031 for optimal effector function

  • Format selection and molecular engineering

    • Comparison of different formats such as bispecific single chain (bssc) and Fabsc (resembling normal antibody structure)

    • Optimization of linker regions and domain orientation

In Vitro Characterization:

  • Biophysical assessment

    • Determination of binding affinity to both targets (e.g., 49 pM for FLT3 and 27 nM for CD3)

    • Evaluation of production yield and aggregation tendency

    • Analysis of thermal stability and solution behavior

  • Functional testing with cell lines

    • Assessment of T-cell activation (CD69, CD25 expression)

    • Measurement of cytokine production (IFN-γ, TNF-α)

    • Quantification of leukemia cell killing at various effector:target ratios

    • Determination of EC50 values for cytotoxicity

Ex Vivo Evaluation with Patient Samples:

  • Testing with primary AML samples

    • Incubation of patient-derived PBMCs containing both T cells and leukemic blasts with bispecific antibodies

    • Assessment of T-cell activation, proliferation, and blast reduction

    • Evaluation of antibody efficacy across samples with varying FLT3 expression levels

  • Determination of optimal dosing

    • Titration experiments to identify minimal effective concentration

    • Assessment of potential on-target, off-tumor effects on normal hematopoietic cells

In Vivo Studies:

  • Evaluation in animal models

    • Assessment of pharmacokinetics and half-life

    • Determination of efficacy in xenograft models

    • Evaluation of safety and toxicity profiles, particularly regarding effects on normal hematopoietic cells

This comprehensive methodological approach has led to the development of promising bispecific antibodies that demonstrate high potency against AML cells while offering advantages over traditional therapeutic approaches .

How can researchers distinguish between the effects of FLT3 overexpression versus FLT3 mutations in experimental models?

Distinguishing between the effects of FLT3 overexpression and FLT3 mutations requires careful experimental design and multiple complementary approaches:

Genetic Engineering Approaches:

  • Create isogenic cell lines that differ only in FLT3 status:

    • Wild-type FLT3 at normal expression levels (control)

    • Wild-type FLT3 with overexpression

    • Mutant FLT3 (ITD or D835) at normal expression levels

    • Mutant FLT3 with overexpression

  • Use inducible expression systems to control FLT3 levels:

    • Tetracycline-regulated promoters allow titration of FLT3 expression

    • Compare cellular responses at equivalent protein levels between wild-type and mutant FLT3

Biochemical and Functional Assessments:

  • Analyze receptor phosphorylation patterns:

    • Overexpressed wild-type FLT3 shows autophosphorylation similar to mutant FLT3

    • Use phospho-specific antibodies to identify differential phosphorylation sites

  • Evaluate downstream signaling activation:

    • Compare activation of STAT5, MAPK, and PI3K/AKT pathways

    • Assess kinetics of signaling (constitutive versus ligand-dependent)

  • Test sensitivity to FLT3 inhibitors:

    • Overexpressed wild-type FLT3 and mutant FLT3 may show different sensitivities to inhibitors

    • Determine IC50 values for various inhibitors against each FLT3 variant

Functional and Phenotypic Analyses:

  • Assess cellular transformation properties:

    • Colony formation in semi-solid media

    • Growth factor independence

    • Cell cycle distribution and apoptosis resistance

  • Evaluate gene expression signatures:

    • Perform RNA-seq to identify distinct transcriptional profiles

    • Compare with patient data to validate clinical relevance

  • Assess in vivo leukemogenic potential:

    • Xenograft models using engineered cell lines

    • Monitor disease progression, latency, and phenotype

Research has demonstrated that FLT3 overexpression without mutations can be an independent negative prognostic factor in AML patients, suggesting a distinct biological entity . Understanding the overlapping yet distinct effects of expression level versus mutational status is crucial for developing targeted therapeutic strategies and selecting appropriate experimental models for drug testing.

What are the current methodological challenges in developing FLT3 antibodies that can distinguish between wild-type and mutant FLT3?

Developing antibodies that selectively recognize mutant forms of FLT3 presents several methodological challenges that researchers must address:

Structural Constraints:

  • FLT3/ITD mutations occur in the juxtamembrane domain, which is intracellular and inaccessible to conventional antibodies

  • Activating point mutations like D835 alter protein conformation subtly without creating unique epitopes

  • Developing antibodies that can penetrate the cell membrane while maintaining specificity requires innovative approaches

Epitope Selection Strategies:

  • Utilize computational modeling and structural biology to identify conformational changes specific to mutant proteins

  • Design synthetic peptides spanning mutation sites for immunization

  • Employ phage display technology with negative selection against wild-type epitopes

Validation Challenges:

  • Ensure antibodies recognize natural mutant protein in its native conformation

  • Develop robust controls using isogenic cell lines with different FLT3 variants

  • Confirm specificity across multiple patient-derived samples with diverse FLT3 mutations

Alternative Approaches:

  • Proximity-based detection systems:

    • Develop antibody pairs that recognize distinct epitopes and produce signal only when in proximity

    • One antibody targets common FLT3 epitope while another detects mutation-induced conformational change

  • Intrabodies and nanobodies:

    • Engineer smaller antibody formats capable of intracellular targeting

    • Express genetically encoded antibody fragments fused to fluorescent proteins

  • Conformation-specific antibodies:

    • Target unique conformational epitopes created by activating mutations

    • Utilize hydrogen-deuterium exchange mass spectrometry to identify mutation-specific accessible regions

  • Aptamer-based approaches:

    • Develop nucleic acid aptamers with high specificity for mutant conformations

    • Create aptamer-antibody conjugates for enhanced specificity

These methodological challenges require interdisciplinary approaches combining structural biology, protein engineering, and advanced screening technologies. While direct antibody-based discrimination between wild-type and mutant FLT3 remains difficult, these alternative strategies may yield valuable research tools for studying FLT3 biology and developing targeted therapies .

How can researchers address potential on-target, off-tumor toxicity when using FLT3 antibodies in experimental therapeutics?

Addressing on-target, off-tumor toxicity is crucial when developing FLT3-targeted experimental therapeutics, as FLT3 is expressed not only on leukemic cells but also on normal hematopoietic stem and progenitor cells (HSPCs) and dendritic cells (DCs). Several methodological approaches can be implemented:

Preclinical Safety Assessment:

  • In vitro toxicity evaluation:

    • Compare antibody binding and effects on leukemic cells versus normal HSPCs and DCs

    • Conduct dose-response studies to identify therapeutic windows

    • Establish minimum effective concentration against leukemic cells

  • Primate studies:

    • Evaluate hematological toxicity in cynomolgus monkeys as their FLT3 expression pattern closely resembles humans

    • Monitor for reversibility of any observed toxicity

    • Assess recovery kinetics of affected cell populations

Engineering Strategies:

  • Affinity modulation:

    • Fine-tune antibody affinity to preferentially target high-expressing leukemic cells over low-expressing normal cells

    • Develop mathematical models to predict differential binding based on receptor density

  • Conditional activation systems:

    • Design antibodies with masked binding sites that become activated only in the tumor microenvironment

    • Utilize tumor-specific proteases or pH-sensitive linkers to control antibody activity

  • Dosing schedule optimization:

    • Implement fractionated dosing regimens to minimize toxicity

    • Design intermittent treatment schedules that allow recovery of normal cells

Combination Approaches:

  • Adjunctive cytoprotective strategies:

    • Co-administer cytokines that support HSPC survival and recovery

    • Explore ex vivo stem cell preservation for potential rescue after therapy

  • Selective targeting enhancement:

    • Combine FLT3 antibodies with agents that upregulate FLT3 specifically on leukemic cells

    • Utilize secondary targeting moieties that recognize leukemia-specific markers

Research has shown that toxicity to normal hematopoietic cells can be reversible and potentially manageable in clinical settings, suggesting that careful optimization of these approaches may yield therapeutics with acceptable safety profiles . The comprehensive evaluation of potential toxicity to normal cells expressing FLT3 should be an integral part of the experimental design when developing FLT3-targeted therapies.

What methodological approaches are most effective for analyzing the relationship between FLT3 signaling and the bone marrow microenvironment?

Understanding the complex interplay between FLT3 signaling and the bone marrow microenvironment requires sophisticated methodological approaches that capture both cellular and molecular interactions:

Advanced Co-Culture Systems:

  • 3D organoid models:

    • Develop bone marrow organoids incorporating stromal cells, osteoblasts, and endothelial cells

    • Compare FLT3 signaling in leukemic cells within organoids versus traditional 2D culture

    • Evaluate therapeutic responses in this more physiologically relevant context

  • Patient-derived xenograft (PDX) co-cultures:

    • Establish co-cultures using primary leukemic cells and patient-matched stromal components

    • Analyze differential FLT3 signaling in cells adherent to stroma versus cells in suspension

    • Assess spatial heterogeneity of FLT3 activation within the culture system

Molecular Interaction Analysis:

  • Proximity ligation assays:

    • Detect and quantify interactions between FLT3 and microenvironmental factors

    • Map spatial distribution of FLT3 signaling complexes relative to stromal contacts

    • Identify novel binding partners in the context of the microenvironment

  • Phosphoproteomics with cellular resolution:

    • Combine phospho-flow cytometry with mass cytometry (CyTOF) to analyze FLT3 signaling

    • Profile signaling changes in response to specific microenvironmental factors

    • Identify alterations in signaling networks that contribute to therapeutic resistance

In Vivo Imaging and Analysis:

  • Intravital microscopy:

    • Visualize FLT3-expressing cells within their native microenvironment

    • Track cellular behavior and signaling dynamics in real-time

    • Assess the impact of therapeutic interventions on both leukemic cells and surrounding stroma

  • Spatial transcriptomics and proteomics:

    • Map gene and protein expression patterns within intact bone marrow specimens

    • Correlate FLT3 expression and activation with microenvironmental niches

    • Identify stromal signatures associated with enhanced FLT3 signaling

Functional Dissection Methods:

  • Conditional genetic systems:

    • Use inducible knockout or overexpression of FLT3 in specific cellular compartments

    • Analyze reciprocal signaling between leukemic and stromal cells

    • Determine the role of FLT3 in remodeling the microenvironment

  • Microfluidic devices:

    • Create defined gradients of growth factors and chemokines

    • Analyze FLT3-dependent migration and homing behaviors

    • Test combinatorial effects of multiple microenvironmental stimuli

These methodological approaches enable researchers to dissect the bidirectional communication between FLT3-expressing leukemic cells and the bone marrow microenvironment, revealing mechanisms of leukemogenesis, disease progression, and therapeutic resistance that cannot be identified through conventional culture systems.

How should researchers interpret discrepancies between FLT3 protein expression and transcript levels in experimental samples?

Discrepancies between FLT3 protein expression and transcript levels are common in experimental samples and require careful interpretation. Several methodological considerations can help researchers address these discrepancies:

Potential Biological Mechanisms:

  • Post-transcriptional regulation:

    • Evaluate the role of microRNAs targeting FLT3 mRNA

    • Assess mRNA stability through actinomycin D chase experiments

    • Analyze polysome profiling to determine translational efficiency

  • Post-translational modifications:

    • Investigate protein stability using cycloheximide chase assays

    • Examine ubiquitination status of FLT3 protein

    • Assess the impact of proteasome inhibitors on FLT3 protein levels

  • Receptor trafficking and localization:

    • Distinguish between total and surface FLT3 expression using permeabilized vs. non-permeabilized flow cytometry

    • Evaluate subcellular localization using fractionation or imaging techniques

    • Assess internalization and recycling rates of the receptor

Technical Considerations:

  • Methodological limitations:

    • RT-qPCR may detect transcripts regardless of their translation status

    • Antibodies may have different affinities for various FLT3 conformations or modified forms

    • Flow cytometry detects primarily surface expression while Western blotting captures total protein

  • Sample processing effects:

    • Compare fresh versus frozen/fixed samples for potential differences

    • Standardize time from sample collection to analysis

    • Evaluate the impact of different preservation methods on protein detection

  • Assay dynamic ranges:

    • Ensure measurements fall within the linear range of both protein and transcript assays

    • Consider the possibility of signal saturation in highly expressing samples

    • Use appropriate dilution series to accurately quantify expression levels

Interpretation Framework:

  • Integrated analysis approach:

    • Correlate discrepancies with clinical or experimental outcomes

    • Consider the functional significance of protein versus transcript levels

    • Determine which measurement better predicts cellular behavior or therapeutic response

  • Context-specific evaluation:

    • Assess whether discrepancies are consistent across similar samples or unique to specific conditions

    • Compare with known regulatory patterns in different cell types or disease states

    • Develop mathematical models to account for the relationship between transcript and protein levels

When interpreting such discrepancies, researchers should recognize that each measurement provides distinct biological insights, and the integration of multiple approaches often yields the most comprehensive understanding of FLT3 biology in experimental systems .

What are the common technical challenges in using FLT3 antibodies for immunoprecipitation studies and how can they be addressed?

Immunoprecipitation (IP) studies with FLT3 antibodies present several technical challenges that researchers should anticipate and address:

Challenge 1: Maintaining Receptor Integrity

  • Problem: FLT3 is a large transmembrane protein (approximately 160 kDa) prone to degradation during sample processing.

  • Solutions:

    • Use fresh samples whenever possible

    • Incorporate multiple protease inhibitors targeting different classes of proteases

    • Maintain samples at 4°C throughout processing

    • Consider using shorter lysis times to minimize degradation

    • Add phosphatase inhibitors to preserve phosphorylation status

Challenge 2: Efficient Extraction from Membranes

  • Problem: As a transmembrane protein, FLT3 can be difficult to solubilize while maintaining native conformation.

  • Solutions:

    • Optimize detergent selection (compare NP-40, Triton X-100, CHAPS, or digitonin)

    • Use mild detergent concentrations (0.5-1%) to preserve protein-protein interactions

    • Consider membrane fractionation before solubilization

    • Implement gentle homogenization methods to avoid protein denaturation

    • Test different buffer compositions to enhance extraction efficiency

Challenge 3: Non-specific Binding

  • Problem: High background and false positives due to non-specific interactions.

  • Solutions:

    • Pre-clear lysates with protein A/G beads before adding the FLT3 antibody

    • Include appropriate isotype controls

    • Optimize antibody concentration through titration experiments

    • Use more stringent washing conditions for high-specificity applications

    • Consider crosslinking antibodies to beads to prevent heavy chain interference in Western blots

Challenge 4: Low IP Efficiency

  • Problem: Poor recovery of FLT3 during immunoprecipitation.

  • Solutions:

    • Evaluate multiple antibody clones targeting different epitopes

    • Optimize antibody-to-lysate ratios

    • Extend incubation time (overnight at 4°C) to enhance antigen capture

    • Test different types of beads (magnetic vs. agarose) for better performance

    • Consider using directly conjugated antibodies to eliminate secondary capture steps

Challenge 5: Co-IP of Interacting Partners

  • Problem: Difficulty in maintaining protein-protein interactions during IP.

  • Solutions:

    • Use chemical crosslinking to stabilize transient interactions

    • Adjust salt concentration in buffers (typically 150 mM NaCl for maintaining interactions)

    • Optimize detergent type and concentration to preserve complexes

    • Consider proximity-based labeling techniques (BioID, APEX) as complementary approaches

    • Validate interactions through reciprocal IP experiments

By systematically addressing these technical challenges, researchers can significantly improve the quality and reliability of immunoprecipitation studies involving FLT3, enabling more accurate characterization of its interactions, modifications, and signaling properties in normal and leukemic cells.

How can researchers accurately assess FLT3 antibody specificity and validate results across different experimental systems?

Rigorous validation of FLT3 antibody specificity across experimental systems is essential for generating reliable and reproducible research data. A comprehensive validation strategy should include:

Genetic Controls for Specificity Assessment:

  • Knockout/knockdown validation:

    • Test antibodies on FLT3 knockout cell lines created via CRISPR-Cas9

    • Compare signals between wild-type and FLT3-depleted samples using siRNA or shRNA

    • Include gradients of knockdown to assess signal correlation with expression level

  • Overexpression systems:

    • Evaluate antibody performance in cells with controlled FLT3 expression

    • Use inducible expression systems to create titration curves

    • Test antibody specificity against related receptor tyrosine kinases (e.g., c-KIT, PDGFR)

Multi-method Concordance Analysis:

  • Orthogonal detection techniques:

    • Compare results across different methodologies (flow cytometry, Western blotting, immunofluorescence)

    • Assess correlation between protein detection and mRNA expression

    • Use mass spectrometry to confirm the identity of immunoprecipitated proteins

  • Epitope mapping:

    • Determine the specific binding region using truncated protein variants

    • Perform peptide competition assays to confirm epitope specificity

    • Evaluate cross-reactivity with species homologs based on epitope conservation

Cross-Platform Standardization:

  • Reference standards:

    • Establish well-characterized positive control samples (e.g., EOL-1 cell line for FLT3 expression)

    • Create calibration curves using samples with defined FLT3 levels

    • Include consistent controls across all experiments

  • Reporting standards:

    • Document detailed antibody information (clone, supplier, lot number, concentration)

    • Specify exact experimental conditions (incubation time, temperature, buffer composition)

    • Report both positive and negative validation results

Application-Specific Validation:

  • For flow cytometry:

    • Perform fluorescence-minus-one (FMO) controls

    • Evaluate non-specific binding with isotype controls

    • Establish gating strategies based on known positive and negative populations

  • For immunohistochemistry/immunofluorescence:

    • Include tissue with known FLT3 expression patterns as positive controls

    • Perform antigen competition assays

    • Test multiple fixation and antigen retrieval protocols

  • For therapeutic applications:

    • Assess binding to primary patient samples with variable FLT3 expression

    • Evaluate potential cross-reactivity with normal tissues

    • Test functionality across different experimental models

Implementing this comprehensive validation strategy ensures that experimental findings are truly attributable to FLT3 and not artifacts of antibody cross-reactivity or technical variables. This is particularly important given the critical role of FLT3 as both a research target and therapeutic opportunity in leukemia .

What methodological approaches show promise for developing next-generation FLT3 antibodies with enhanced specificity or functionality?

Several innovative approaches are advancing the development of next-generation FLT3 antibodies with improved properties:

Structural Biology-Guided Design:

  • Cryo-EM and X-ray crystallography:

    • Utilize high-resolution structural data to identify unique epitopes

    • Design antibodies targeting specific conformational states of FLT3

    • Engineering antibodies that lock FLT3 in inactive conformations

  • Molecular dynamics simulations:

    • Predict antibody-antigen interactions and binding kinetics

    • Optimize binding interfaces through computational modeling

    • Identify allosteric sites that could modulate receptor function

Advanced Antibody Engineering Platforms:

  • AI-assisted antibody optimization:

    • Apply machine learning algorithms to predict optimal complementarity-determining regions (CDRs)

    • Use computational approaches to enhance stability and reduce immunogenicity

    • Design antibodies with predetermined binding and functional properties

  • Novel antibody formats:

    • Develop smaller antibody fragments with improved tissue penetration

    • Create multispecific antibodies targeting FLT3 along with other leukemia-associated antigens

    • Engineer antibodies with switchable binding domains for controlled activity

Functional Enhancement Strategies:

  • Antibody-drug conjugates (ADCs):

    • Conjugate FLT3 antibodies with novel payloads (e.g., PROTACs, immune modulators)

    • Utilize cleavable linkers responsive to the leukemic microenvironment

    • Optimize drug-to-antibody ratios for maximal efficacy and minimal toxicity

  • Engineered effector functions:

    • Modify Fc regions to enhance ADCC or complement-dependent cytotoxicity

    • Create bispecific T-cell engagers with optimized geometry and binding kinetics

    • Develop antibodies that recruit specific immune cell subsets for enhanced anti-tumor activity

Selective Targeting Approaches:

  • Conformation-specific antibodies:

    • Develop antibodies that preferentially bind activated FLT3 conformations

    • Create antibodies selective for mutant forms of FLT3

    • Design antibodies detecting specific post-translational modifications

  • Conditional activation systems:

    • Develop protease-activated antibodies that function only in the tumor microenvironment

    • Create pH-sensitive antibodies that bind preferentially in acidic tumor environments

    • Design antibodies with masking domains removable by tumor-associated enzymes

These methodological advances are driving the development of FLT3 antibodies with unprecedented specificity, potency, and functional versatility. Next-generation antibodies may offer improved therapeutic windows, reduced off-tumor toxicity, and enhanced efficacy against heterogeneous leukemic populations .

How might integrated multi-omic approaches enhance our understanding of FLT3 biology in normal and leukemic hematopoiesis?

Integrated multi-omic approaches offer powerful methodologies to comprehensively characterize FLT3 biology across normal and leukemic contexts:

Multi-layered Data Generation:

  • Genomic profiling:

    • Whole genome/exome sequencing to identify FLT3 mutations and co-occurring genetic alterations

    • Analysis of copy number variations affecting FLT3 expression

    • Investigation of regulatory region polymorphisms influencing transcription

  • Transcriptomic analysis:

    • RNA-seq to quantify transcript levels and identify splice variants

    • Single-cell RNA-seq to detect cellular heterogeneity in FLT3 expression

    • Nascent RNA analysis to assess transcriptional dynamics

  • Proteomic and post-translational modification mapping:

    • Mass spectrometry-based quantification of FLT3 protein levels

    • Phosphoproteomics to map signaling networks downstream of FLT3

    • Analysis of glycosylation patterns affecting receptor maturation and function

  • Epigenomic characterization:

    • ChIP-seq to identify transcription factors regulating FLT3 expression

    • ATAC-seq to assess chromatin accessibility at the FLT3 locus

    • DNA methylation analysis to detect epigenetic dysregulation

Computational Integration Strategies:

  • Network-based approaches:

    • Construct protein-protein interaction networks centered on FLT3

    • Identify signaling modules altered in leukemic versus normal cells

    • Apply causal reasoning algorithms to infer regulatory relationships

  • Machine learning methods:

    • Develop predictive models of therapeutic response based on multi-omic signatures

    • Identify patterns associated with disease progression or treatment resistance

    • Cluster patients based on integrated profiles for personalized treatment approaches

  • Temporal dynamics analysis:

    • Model changes in FLT3 signaling during differentiation and leukemic transformation

    • Track cellular responses to FLT3 inhibition across multiple molecular levels

    • Identify feedback mechanisms and compensatory pathways

Functional Validation Approaches:

  • CRISPR-based screening:

    • Conduct genome-wide knockout screens to identify synthetic lethal interactions with FLT3

    • Perform epigenome editing to manipulate FLT3 expression

    • Use base editing to introduce specific mutations for functional characterization

  • Pathway perturbation:

    • Systematically inhibit nodes in FLT3 signaling networks

    • Assess combinatorial effects of targeting multiple pathways

    • Identify optimal intervention points for therapeutic development

This integrated approach would enable researchers to:

  • Identify novel regulatory mechanisms controlling FLT3 expression

  • Discover previously unrecognized signaling nodes downstream of FLT3 activation

  • Develop more precise prognostic markers based on multi-omic signatures

  • Design rational combination therapies targeting complementary pathways

  • Understand mechanisms of resistance to FLT3-directed therapies

By implementing these multi-omic approaches, researchers can develop a comprehensive systems biology view of FLT3 function in health and disease, potentially revealing new therapeutic opportunities and biomarkers for personalized medicine approaches in leukemia .

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 2025 TheBiotek. All Rights Reserved.