FLT3 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Structure

The FLT3 antibody is a recombinant or monoclonal antibody targeting the extracellular domains of the FLT3 protein, a key therapeutic target in acute myeloid leukemia (AML) . The HRP (horseradish peroxidase) conjugation enables enzymatic detection in assays like ELISA, Western blotting, or immunohistochemistry (IHC). The antibody-enzyme complex retains specificity for FLT3 while leveraging HRP’s catalytic activity for signal amplification .

Conjugation Methods

HRP conjugation typically involves crosslinkers such as N-succinimidyl 3-(2-pyridyldithio)propionate (SPDP) or maleimide-based linkers, which covalently attach the enzyme to the antibody’s Fc region. This method minimizes steric hindrance while preserving antigen-binding affinity . Alternative approaches, such as elastin-like polypeptide (ELP) fusion proteins, enhance stability and pharmacokinetics, though these are more common in therapeutic ADCs .

Key Uses

  • Western Blotting: Detects FLT3 expression levels in lysates of AML cell lines (e.g., MV4-11, THP-1) .

  • ELISA: Quantifies FLT3 in patient sera or conditioned media .

  • Immunohistochemistry: Identifies FLT3-positive blasts in bone marrow biopsies .

  • Flow Cytometry: Combined with fluorescent secondary antibodies to assess FLT3 surface expression .

Validation Data

AssaySensitivitySpecificityCross-reactivity
Western Blotting50–100 ng/mL98%Human FLT3 only
ELISA10–50 ng/mL95%No cross-reactivity
Immunohistochemistry1:100–1:50090%Minimal vs. CD99

Therapeutic Implications

While not directly therapeutic, the HRP-conjugated FLT3 antibody informs the development of FLT3-targeted therapies:

  • Antibody-Drug Conjugates (ADCs): Studies on 20D9-ADC and FL-DM1 highlight FLT3 as a druggable target, with conjugation methods analogous to HRP-antibody systems .

  • Bispecific Antibodies: Dual targeting of FLT3 and CD3/CD99 enhances specificity and reduces off-target effects, as demonstrated in preclinical models .

Mechanism of Action

The antibody binds FLT3’s extracellular Ig-like domains, triggering receptor dimerization and downstream signaling pathways (e.g., AKT, MAPK, STAT5) . HRP conjugation preserves this binding while enabling enzymatic detection of phosphorylated tyrosine residues .

Limitations and Considerations

  • Specificity: Cross-reactivity with CD99 or normal hematopoietic stem cells requires optimization .

  • Stability: HRP-antibody complexes may degrade under high-temperature or prolonged storage conditions .

  • Clinical Translation: Requires validation in patient samples and comparison with existing FLT3 inhibitors (e.g., midostaurin) .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery time estimates.
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. FLT3 promotes phosphorylation of SHC1 and AKT1, leading to activation of the downstream effector MTOR. It also promotes activation of RAS signaling and phosphorylation of downstream kinases, including MAPK1/ERK2 and/or MAPK3/ERK1. Additionally, FLT3 promotes phosphorylation of FES, FER, PTPN6/SHP, PTPN11/SHP-2, PLCG1, and STAT5A and/or STAT5B. Notably, activation of wild-type FLT3 induces only marginal activation of STAT5A or STAT5B. Mutations that cause constitutive kinase activity, however, promote cell proliferation and resistance to apoptosis through activation of multiple signaling pathways.
Gene References Into Functions
  1. Multivariate Cox's proportional hazards regression analyses revealed that high OCT4 mRNA expression was an independent predictor of shorter EFS and OS in AML patients. These findings suggest that OCT4 may serve as a valuable biomarker for predicting unfavorable prognosis in AML patients, particularly in cases with CK, FLT3-ITD mutation, and poorer risk stratification. PMID: 29950146
  2. This study demonstrated that DNMT3A mutations alone do not significantly impact the clinical outcomes of AML patients undergoing allogeneic HSCT. However, when accompanied by 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 results of this study indicated that FLT3 ITD mutations could serve as an indicator of poor prognosis in APL patients. These patients may require more intensive therapy according to current treatment guidelines. PMID: 29251252
  5. Low FLT3 expression has been associated with Pancreatic ductal adenocarcinoma. PMID: 30275197
  6. The DNMT3A R882 mutation plays a significant role in CN-AML patients' prognosis and clinical outcomes, both in the presence and absence of NPM1 and FLT3 mutations. PMID: 29079128
  7. The FLT3 inhibitor AC220 effectively inhibited glutamine flux into the antioxidant factor glutathione, primarily due to defective glutamine import. PMID: 28947392
  8. Mutations in the FLT3 gene have been linked to Acute Myeloid Leukemia. PMID: 29530994
  9. Acute myeloid leukemia harboring internal tandem duplication of FMS-like tyrosine kinase 3 (AML(FLT3-ITD)) is associated with poor prognosis. PMID: 29330746
  10. The influence of FLT3-ITD diversity on the response to induction chemotherapy in patients with acute myeloid leukemia has been investigated. PMID: 28034991
  11. This study indicated that overexpression of FLT3 represents 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 assessed their correlations with clinical and laboratory parameters. PMID: 28294102
  13. FLT3 and FLT3-ITD have been shown to directly bind and selectively phosphorylate p27kip1 on tyrosine residue 88 in acute myeloid leukemia. Inhibition of FLT3-ITD in cell lines significantly reduced p27 tyrosine 88 phosphorylation, leading to increased p27 levels and cell cycle arrest. PMID: 28522571
  14. This study demonstrated the potential of targeting FLT3 by FLT3-CAR T cells for the treatment of FLT3(+) AML. FLT3-CAR T cells may offer a promising new immunotherapeutic approach for AML patients. PMID: 28496177
  15. The study found that high expressions of BCRP mRNA calculated with Pfaffl's rule and FLT3-ITD are independent poor risk factors in adult patients with AML and intermediate or normal karyotype. PMID: 28618074
  16. A newly identified and recurrent FLT3 juxtamembrane deletion mutation has been shown to exhibit a dominant negative effect on the wild-type FLT3 receptor. PMID: 27346558
  17. While FLT3 cell-surface expression did not vary by FLT3 mutational status, this study found a strong association between high FLT3 expression and KMT2A rearrangements. Importantly, the study concluded that FLT3 cell surface expression did not hold prognostic significance in pediatric Acute Myeloid Leukemia. PMID: 28108543
  18. DNA mutational analysis in FLT3 in acute myeloid leukemia. PMID: 27071442
  19. Data from this study confirmed MLL-PTD and, to a lesser extent, FLT3-ITD as frequent events in +11 AML. Notably, 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 crucial 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 rarely observed using targeted genomic profiling. As of yet, FLT3 amplification does not appear to be an actionable target or a reliable biomarker for FLT3 inhibitor sensitivity. PMID: 27906677
  22. FLT3 has a role in cytarabine transport by SLC29A1 in pediatric acute leukemia. PMID: 27391351
  23. Data suggests a pathway involving MYSM1/miR-150/FLT3 that inhibits the proliferation of B1a cells. This pathway may play a role in the pathogenesis of systemic lupus erythematosus (SLE). PMID: 27590507
  24. This study confirmed that FLT3-ITD-location influences disease biology and leads to changes in global gene expression. In the model used, ITD-location altered 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, depending on 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 there is a place for escalated daunorubicin dosing 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 examines 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. This 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 case 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, not secondary, event in leukemogenesis. The oncogenic events of FLT3/ITD occur at a cell stage possessing CD123. PMID: 27465508
  35. Sorafenib may potentially enable cure of a proportion of very poor risk FLT3-internal tandem duplication-positive acute emyeloid leukemia relapsing after allogeneic stem cell transplantation. PMID: 29055209
  36. This study provided 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. These 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. This study developed a novel targeted therapeutic strategy using FLT3L-guided miR-150-based nanoparticles to 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 for 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 results in 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 toward illegitimate recombination. PMID: 28108507
  47. This study revealed a novel mechanism that regulates acute myeloid leukemia cell death by 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

How does FLT3 signaling function in normal cells versus leukemic cells?

In normal cells, FLT3 signaling is tightly regulated and only activated when the FLT3 ligand (FL) binds to the receptor, causing dimerization and transphosphorylation of tyrosine residues on the tyrosine kinase domain. These phosphorylated domains recruit adaptors containing SH2 or PTB domains and activate downstream signaling proteins including AKT, MAPK, STAT5, and SFK family members, leading to controlled anti-apoptosis, cell survival, and proliferation responses .

In leukemic cells, particularly those with FLT3-ITD mutations, the receptor exhibits constitutive autophosphorylation independent of ligand binding. Research has demonstrated that this aberrant signaling primarily affects the immature form of the receptor, which is potentially localized intracellularly . This constitutive activation leads to dysregulated downstream signaling that promotes factor-independent growth and survival of leukemic cells . Additionally, the balance between immature and mature forms of the receptor is disturbed in mutated FLT3, with weaker expression of the mature form observed in immunoblot analyses . These alterations in signaling contribute to the aggressive nature of FLT3-mutated leukemias and their resistance to conventional therapies.

What detection techniques are available for analyzing FLT3 expression in research samples?

Several validated detection methods are available for FLT3 analysis in research settings:

TechniqueApplicationsDetection RangeKey Advantages
Western BlotProtein expression and phosphorylation status130-160 kDa observed MWAllows assessment of both mature/immature forms
Immunohistochemistry (IHC-P)Tissue localizationN/ASpatial information in tissue context
Flow CytometryCell surface expressionN/ASingle-cell resolution
ELISAQuantitative protein measurementVariable based on standardsHigh throughput capability

For Western blotting applications, HRP-conjugated secondary antibodies are commonly used at a dilution of 1:10000 for visualization of FLT3, with recommended primary antibody dilutions ranging from 1:100 to 1:500 . The observation of multiple bands (130kDa/160kDa) is expected due to different glycosylation states of the receptor, with the lower band typically representing the immature, potentially intracellular form and the higher band representing the mature, cell surface form .

How can FLT3 antibodies be used to study receptor internalization and trafficking in leukemic cells?

FLT3 antibodies can provide valuable insights into receptor internalization and trafficking dynamics through several methodological approaches:

To study internalization kinetics, researchers can employ fluorescently-labeled FLT3 antibodies to track receptor movement after binding. This typically involves incubating cells with labeled antibodies at 4°C (to permit binding without internalization), followed by warming to 37°C to initiate internalization. Sequential imaging or flow cytometry analysis at defined time points allows quantification of internalization rates .

For investigating intracellular trafficking pathways, dual-labeling techniques combining FLT3 antibodies with markers for different cellular compartments (endosomes, lysosomes, etc.) enable visualization of receptor routing. Co-localization analyses can determine whether FLT3 follows recycling or degradative pathways after internalization .

These methodologies are particularly important when developing antibody-drug conjugates (ADCs) targeting FLT3, as efficient internalization is crucial for therapeutic efficacy. Research has demonstrated that both wild-type FLT3 and FLT3-ITD mutants can internalize bound antibodies, though potentially with different kinetics and routing, which has significant implications for drug delivery strategies .

What are the optimal conditions for preserving FLT3 epitope integrity during sample preparation for immunodetection?

Preserving FLT3 epitope integrity requires careful consideration of several methodological factors:

For protein extraction, a balanced approach is necessary. Harsh detergents like SDS may maximize protein yield but can denature epitopes, while milder detergents like NP-40 or Triton X-100 (0.5-1%) better preserve conformational epitopes. Buffer systems should maintain physiological pH (7.2-7.4) and include protease inhibitors to prevent degradation. For phospho-specific detection, phosphatase inhibitors (sodium orthovanadate, sodium fluoride) must be included .

When preparing tissues for immunohistochemistry, optimal fixation involves 10% neutral buffered formalin for 24-48 hours, followed by paraffin embedding. Antigen retrieval methods vary by antibody but typically require heat-induced epitope retrieval in citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) .

For flow cytometry, gentle cell dissociation methods (using enzyme-free dissociation buffers when possible) help preserve surface epitopes. Fixation should use 2-4% paraformaldehyde rather than methanol-based fixatives that may disrupt membrane proteins. Additionally, keeping cells at 4°C during processing minimizes receptor internalization and epitope degradation .

Validation studies have shown that these optimized conditions significantly improve detection sensitivity, ensuring reliable analysis of both wild-type and mutant FLT3 variants in research applications .

How can researchers differentiate between wild-type FLT3 and FLT3-ITD mutations using antibody-based detection methods?

Differentiating between wild-type FLT3 and FLT3-ITD mutations using antibody-based methods requires specific analytical approaches:

Western blot analysis provides valuable information through examination of phosphorylation patterns. Wild-type FLT3 typically shows ligand-dependent phosphorylation, while FLT3-ITD exhibits constitutive phosphorylation, particularly of the lower molecular weight (immature) form . Additionally, the ratio between mature and immature receptor forms differs, with FLT3-ITD showing relatively stronger expression of the immature form .

For assessing functional differences, researchers can employ phospho-specific antibodies targeting key downstream signaling molecules. FLT3-ITD mutations show altered activation profiles of MAPK/Erk and Akt signaling pathways, with some mutations (like ITD2) inducing significant constitutive activation of Erk proteins even without ligand stimulation .

Immunofluorescence approaches can reveal differences in subcellular localization, as FLT3-ITD may show greater retention in intracellular compartments compared to wild-type FLT3, which predominantly localizes to the plasma membrane .

These antibody-based approaches complement molecular techniques like PCR and sequencing, providing functional information about the consequences of genetic alterations on protein expression, localization, and signaling activities.

What are the critical parameters for optimizing HRP-conjugated antibody detection of FLT3 in Western blotting?

Optimizing HRP-conjugated antibody detection for FLT3 requires careful consideration of several critical parameters:

Signal-to-noise optimization begins with proper blocking (3% nonfat dry milk in TBST has been validated for FLT3 detection) . Primary antibody concentration should be titrated within the recommended range (1:100-1:500 for FLT3), with overnight incubation at 4°C generally yielding better results than shorter incubations at room temperature . For HRP-conjugated secondary antibodies, dilutions around 1:10000 provide optimal balance between signal strength and background .

Membrane transfer conditions are particularly important for large proteins like FLT3 (113kDa calculated MW). Extended transfer times (90-120 minutes) at lower voltage or overnight transfers at 30V improve transfer efficiency of high molecular weight proteins. Using PVDF membranes (0.45μm pore size) rather than nitrocellulose enhances protein retention and signal intensity .

Detection sensitivity can be further improved through enhanced chemiluminescence (ECL) systems. For FLT3 detection, enhanced ECL kits have been successfully used with exposure times of approximately 60 seconds . Signal amplification systems may be beneficial for detecting low expression levels or phosphorylated forms of FLT3.

These optimized conditions allow reliable detection of both mature (160kDa) and immature (130kDa) forms of FLT3, enabling researchers to analyze expression patterns and processing differences between wild-type and mutant receptors .

What strategies can improve the specificity of HRP-conjugated FLT3 antibodies in complex sample types?

Enhancing specificity of HRP-conjugated FLT3 antibodies in complex samples involves several validated methodological approaches:

Pre-adsorption techniques can significantly reduce cross-reactivity in complex tissues. This involves pre-incubating the primary antibody with recombinant FLT3 protein at concentrations that saturate non-specific binding sites while maintaining specific epitope recognition. Studies have shown this approach particularly valuable for immunohistochemistry applications in tissues with high background .

Sequential immunoprecipitation strategies offer another approach to specificity enhancement. By first immunoprecipitating FLT3 from complex lysates using validated antibodies, then proceeding with standard Western blotting, researchers can substantially reduce interfering proteins. This method has been successfully employed to study FLT3 phosphorylation status in primary patient samples .

Signal validation through multiple detection approaches is essential for confirming specificity. Detection of FLT3 should be verified using antibodies recognizing different epitopes, and results should be correlated with gene expression data where possible. Additionally, using FLT3-negative cell lines as controls helps establish baseline signal and identify potential cross-reactivity .

For complex tissues, dual-labeling with cell-type specific markers (CD34, CD45, etc.) can help confirm that detected FLT3 signals originate from the expected cell populations, reducing misinterpretation of results .

How does the choice of HRP substrate affect detection sensitivity for FLT3 in different experimental contexts?

The selection of HRP substrate significantly impacts detection sensitivity for FLT3 across various experimental applications:

In Western blotting applications, luminol-based enhanced chemiluminescence (ECL) substrates have been successfully used for FLT3 detection with exposure times around 60 seconds . For detecting low FLT3 expression or subtle changes in phosphorylation status, femto-level sensitivity substrates can improve detection by 10-50 fold compared to standard ECL, though optimization of antibody concentrations is required to prevent signal saturation.

For immunohistochemistry applications, the choice between chromogenic and fluorescent substrates depends on research objectives. 3,3'-Diaminobenzidine (DAB) provides permanent staining with excellent morphological context but limited dynamic range. In contrast, tyramide signal amplification (TSA) systems can enhance sensitivity 10-200 fold, enabling detection of low-abundance FLT3 in tissue samples that might otherwise be considered negative .

In multiplex detection scenarios where simultaneous visualization of FLT3 with other markers is required, HRP substrates with distinct spectral properties or sequential detection using HRP-inactivation steps between antibody applications enable complex analytical approaches that maintain specificity.

Comparative studies have shown that optimal substrate selection should consider not only sensitivity requirements but also stability, signal duration, and compatibility with downstream analyses such as image quantification or tissue clearing techniques .

How do antibody-drug conjugates targeting FLT3 compare to tyrosine kinase inhibitors in research applications?

Antibody-drug conjugates (ADCs) and tyrosine kinase inhibitors (TKIs) targeting FLT3 represent distinct therapeutic approaches with different research applications:

Mechanism comparison: TKIs directly inhibit FLT3 kinase activity by competing with ATP binding, primarily affecting signaling pathways. In contrast, ADCs utilize FLT3 primarily as a delivery vehicle to internalize cytotoxic payloads into FLT3-expressing cells, causing direct cell killing through payload-mediated mechanisms . This fundamental difference makes ADCs potentially effective against cells with both wild-type and mutant FLT3, while TKIs primarily target cells dependent on constitutively active FLT3 signaling .

Target population research shows that TKIs are most effective against the 30-40% of AML patients with FLT3 mutations (particularly FLT3-ITD), while ADCs potentially target the broader 90% of AML patients with FLT3 expression regardless of mutation status . This distinction represents an important research avenue for addressing the unmet clinical need in FLT3-wild-type AML patients.

Resistance mechanism studies reveal different vulnerabilities. TKI resistance often develops through secondary FLT3 mutations affecting the kinase domain, altered drug efflux, or activation of parallel signaling pathways . ADC resistance typically involves different mechanisms: reduced target expression, impaired internalization, altered intracellular trafficking, or payload efflux . These different resistance mechanisms suggest potential for sequential or combination approaches in research models.

Experimental data shows that combining FLT3-targeting ADCs with FLT3 TKIs produces enhanced cytotoxic effects through synergistic mechanisms, as TKI treatment increases surface expression of FLT3 on FLT3-ITD positive AML cells, potentially enhancing ADC binding and efficacy .

What experimental approaches can assess the efficacy of FLT3 antibody-mediated cytotoxicity in leukemic cells?

Evaluating FLT3 antibody-mediated cytotoxicity requires multiple complementary experimental approaches:

Cell viability assays provide fundamental efficacy data. Research with FLT3-targeting ADCs has effectively employed MTT/MTS assays to determine IC50 values, with studies demonstrating nanomolar potency (IC50 of 12.9 nM and 1.1 nM against THP-1 and MV-4-11 AML cells respectively for FL-DM1 conjugate) . Dose-response analyses across multiple time points (24, 48, 72 hours) help characterize the kinetics of cytotoxic effects.

Mechanistic assessments of cell death pathways are crucial for understanding therapeutic mechanisms. Apoptosis analyses using flow cytometry (Annexin V/PI staining) and Western blotting for apoptotic markers (cleaved caspase-3, PARP) have demonstrated that FLT3-targeting agents induce caspase-3-dependent apoptosis . Cell cycle analysis has revealed that certain conjugates, like FL-DM1, arrest cells at the G2/M phase, consistent with the microtubule-disrupting mechanism of the DM1 payload .

Selectivity assessment is critical for evaluating therapeutic potential. Comparative studies using cell lines with and without FLT3 expression provide important specificity data. Research using HCD-57 cells transformed with FLT3-ITD versus parental HCD-57 cells (lacking FLT3 expression) has demonstrated selective targeting by FLT3-directed therapeutics . Additionally, evaluating effects on normal hematopoietic cells (such as CD43-positive cells) helps assess potential off-target toxicity .

Ex vivo testing using primary patient samples provides clinically relevant efficacy data. Studies have shown that FLT3-targeting conjugates can induce significant apoptosis in primary FLT3-positive AML cells, providing critical translational evidence beyond cell line models .

How can researchers design bispecific antibodies incorporating FLT3 targeting for enhanced therapeutic potential?

Designing bispecific antibodies incorporating FLT3 targeting involves several critical research considerations:

Format selection significantly impacts antibody properties. Research comparing different bispecific formats found that the Fabsc format (resembling normal antibody structure) offered advantages over bispecific single chain (bssc) formats for FLT3-targeting bispecifics. The Fabsc format demonstrated superior target affinity, higher production yield, and reduced aggregate formation compared to bssc antibodies with identical specificities . These properties are crucial for therapeutic development and in vivo application.

Epitope selection requires careful investigation. When designing FLT3-directed bispecifics, researchers should target epitopes that: (1) are abundantly expressed on target cells; (2) trigger efficient internalization; and (3) are accessible in the tumor microenvironment. For FLT3 × CD3 bispecifics, the 4G8 antibody targeting FLT3 combined with the UCHT1 antibody targeting CD3 has demonstrated superior properties compared to other combinations .

Functional screening approaches are essential for selecting optimal constructs. T-cell activation assays measuring cytokine production (IFNγ, TNFα) and activation markers (CD25, CD69) in the presence of target cells help identify constructs that efficiently crosslink T cells with FLT3-expressing leukemic cells. Cytotoxicity assays using various effector-to-target ratios provide insights into killing efficiency .

Physiologically relevant testing should employ peripheral blood mononuclear cells from leukemia patients containing "physiologic" amounts of blasts to evaluate bispecific antibody activity under realistic conditions . This approach provides more translatable insights than artificial systems using cell lines or purified cell populations.

How should researchers interpret differences between mature and immature FLT3 forms in Western blot analyses?

The interpretation of mature and immature FLT3 forms in Western blot analyses provides important biological insights:

Molecular weight interpretation is fundamental to distinguishing FLT3 forms. The mature, fully glycosylated form typically appears at approximately 160kDa, while the immature, partially glycosylated form appears at approximately 130kDa . The calculated molecular weight of unmodified FLT3 is 113kDa, illustrating the significant contribution of post-translational modifications to observed molecular weight .

Phosphorylation pattern analysis provides critical functional information. Research has demonstrated that in FLT3-ITD mutations, constitutive autophosphorylation primarily affects the immature (lower band) form, which likely corresponds to intracellularly retained receptor . The mature form may retain more normal ligand-responsive phosphorylation patterns. These differences should be carefully assessed using phospho-specific antibodies along with total FLT3 detection.

Cell compartment correlations help interpret biological significance. The immature form predominantly localizes to intracellular compartments (endoplasmic reticulum, Golgi), while the mature form represents surface-expressed receptor. Changes in the ratio may indicate altered trafficking, which has implications for therapeutic targeting, particularly for agents requiring cell surface binding .

What analytical approaches can identify true FLT3 signaling effects versus experimental artifacts?

Distinguishing genuine FLT3 signaling from artifacts requires rigorous analytical approaches:

Temporal signaling analysis provides critical validation. True FLT3-mediated signaling follows characteristic temporal patterns after ligand stimulation or in constitutively active mutants. Researchers should examine multiple time points (5, 10, 30, 60 minutes post-stimulation) to confirm expected phosphorylation kinetics of both FLT3 and downstream mediators . Artifacts typically show random or inconsistent temporal patterns.

Pathway consistency verification strengthens signaling interpretations. Legitimate FLT3 activation engages multiple downstream pathways simultaneously, including MAPK/Erk, PI3K/Akt, and STAT5 . Researchers should confirm coordinated activation across these pathways, with careful attention to phosphorylation sites specifically linked to FLT3 signaling. Discordant pathway activation may indicate non-specific effects or cross-talk from other receptors.

Pharmacological validation employs selective inhibitors to confirm signaling specificity. FLT3 tyrosine kinase inhibitors should block both receptor autophosphorylation and downstream signaling in wild-type FLT3 following ligand stimulation. In FLT3-ITD mutants, inhibitor sensitivity may differ between the mature and immature forms, providing additional insights into signaling mechanisms .

Genetic controls provide definitive validation. Experiments should include cell lines lacking FLT3 expression, FLT3 knockdown/knockout models, and cells expressing kinase-dead FLT3 mutants to distinguish FLT3-dependent and independent effects . The HCD-57 cell line model system, with and without FLT3-ITD transformation, offers a valuable comparative system for validating FLT3-specific effects .

How can researchers reconcile contradictory findings between antibody-based detection and functional FLT3 assays?

Resolving discrepancies between antibody detection and functional FLT3 assays requires systematic investigative approaches:

Epitope mapping analysis can identify potential mechanistic explanations. Different antibodies recognize distinct FLT3 epitopes, which may be differentially affected by conformational changes, post-translational modifications, or protein interactions. When detection and functional data conflict, researchers should determine whether the antibody epitope lies within functional domains (ligand-binding regions, kinase domain) or might be masked during activation . Comparing results using multiple antibodies targeting different epitopes can resolve this issue.

Subcellular localization assessment addresses compartmentalization effects. Since FLT3 signaling capability differs between cellular compartments, researchers should correlate detection data with compartmentalization information. The observed discrepancy may reflect detection of receptor pools with different functional states - surface-localized FLT3 may be functionally active while intracellular pools (detected in whole-cell lysates) may remain inactive .

Post-translational modification analysis helps resolve apparent contradictions. Glycosylation, phosphorylation, and ubiquitination can affect both antibody detection and receptor function. Enzymatic deglycosylation experiments, phosphatase treatments, and proteasome inhibition can determine whether modifications explain discrepant results . For instance, certain antibodies may preferentially detect unphosphorylated FLT3, missing activated forms.

Methodological sensitivity calibration addresses detection threshold issues. Functional assays often detect effects at receptor expression levels below antibody detection thresholds. Quantitative flow cytometry determining absolute receptor numbers per cell compared with functional dose-response curves can establish whether sensitivity differences explain contradictory findings . This approach is particularly valuable when analyzing primary patient samples with potentially low or heterogeneous FLT3 expression.

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.