Fgf2 Antibody, Biotin conjugated

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
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributor.
Synonyms
Fgf2 antibody; Fgf-2Fibroblast growth factor 2 antibody; FGF-2 antibody; Basic fibroblast growth factor antibody; bFGF antibody; Heparin-binding growth factor 2 antibody; HBGF-2 antibody
Target Names
Uniprot No.

Target Background

Function
Fibroblast growth factor 2 (FGF2) functions as a ligand for fibroblast growth factor receptors 1, 2, 3, and 4 (FGFR1, FGFR2, FGFR3, and FGFR4). It also acts as an integrin ligand, essential for FGF2 signaling, binding to integrin ITGAV:ITGB3. FGF2 plays a crucial role in regulating cell survival, division, differentiation, and migration. It acts as a potent mitogen in vitro, inducing angiogenesis. Additionally, FGF2 mediates phosphorylation of ERK1/2, thereby promoting retinal lens fiber differentiation.
Gene References Into Functions
  1. Dual delivery of bFGF and NGF binding coacervate demonstrated neuroprotective effects by stimulating neuronal growth and proliferation. PMID: 29895019
  2. FGF2 safeguards against renal ischemia-reperfusion injury by attenuating mitochondrial damage and proinflammatory signaling. PMID: 28544332
  3. Research indicates that the fibroblast growth factor 2-ERK1/2 pathway participates in the pathophysiology of depressive-like behaviors. Manipulating the neurogenesis pathway presents a viable therapeutic approach for inflammation-associated depression. PMID: 28529071
  4. These findings offer insight into the mechanism by which bFGF ASODN effectively suppresses the proliferation and differentiation of neural stem cells. PMID: 28390174
  5. The current study demonstrates that OCT, alone or in combination with bFGF, accelerates nerve repair in large peripheral nerve defects in rats. PMID: 27529414
  6. Effect of basic fibroblast growth factor released from chitosan-fucoidan nanoparticles on neurite extension PMID: 23696519
  7. FGF2 is an alcohol-responsive gene, forming a positive regulatory feedback loop with alcohol. This loop facilitates alcohol consumption, highlighting FGF2 as a potential therapeutic target for alcohol addiction. PMID: 28821667
  8. Researchers attempted to identify PKGII-targeted proteins associated with the inhibition of FGF2-induced MAPK activation. PMID: 28057484
  9. A moderate level of FGF-2 expression was observed in cells within the connective tissue of healing wounds in the normoglycemic group on all evaluated days, differing from that observed in the diabetic group. PMID: 27188585
  10. Overexpression of BNIP3L in H9C2 cardiomyoblast cells reduced the cardioprotective effects of FGF-2 in hydrogen peroxide-induced necrosis and mitochondrial dysfunction. PMID: 28006775
  11. The induction of active beta-catenin and subsequent fibronectin turnover in response to bFGF were significantly increased in pulmonary fibroblasts from rats with COPD. The beta-Catenin/RhoA pathway results in ECM deposition in lung fibroblasts and myofibroblasts differentiation. PMID: 27734223
  12. FGF2 plays a significant role as a key trigger of Intramuscular adipose tissue formation in vivo. PMID: 26154243
  13. This study demonstrated that altered Cx43 expression modulates bFGF expression, which correlates with prolactinoma development. PMID: 27078698
  14. Basic fibroblast growth factor increased in spinal microglia during the development of allodynia after spinal nerve ligation. PMID: 26583471
  15. TGF-beta1 was upregulated with FGF-2 treatment, and alpha-SMA expression induced by FGF-2 was inhibited after the cell was transferred with TGF-beta1 siRNA. PMID: 26729053
  16. Apocynin attenuated cardiac injury in type 4 radiorenal syndrome rats by inhibiting NADPH oxidase-dependent oxidative stress-activated ERK1/2 pathway and subsequent FGF-2 upregulation. PMID: 26109504
  17. Astrocyte-secreted FGF2 mediated stress-hormone-induced neural stem cell proliferation. PMID: 23599891
  18. Basic fibroblast growth factor and neurotrophin-3, released from astrocytes by exposure to thyroid hormone, influence each other to enhance Na+ current density in cultured hippocampal neurons. PMID: 26009773
  19. GK-2 had no effect on the expression level of FGFb and NT4, however, it promoted an increase in the expression level of BDNF. PMID: 26571801
  20. Researchers investigated the effects of FGF-2 in dissociated postnatal retinal cell cultures and found that FGF-2 is a potent factor triggering ganglion cell differentiation. PMID: 25402196
  21. Regional differences in the FGF-2 expression pattern were observed. Both the first and second injections of cocaine, by themselves, upregulated FGF-2 mRNA in the medial prefrontal cortex and nucleus accumbens while downregulating it in the hippocampus. PMID: 25124315
  22. Fibroblast growth factor 2 plays roles in maintaining the undifferentiated state and in the proliferation of Endothelial progenitor cells, enabling EPCs to retain the potential to differentiate into Endothelial Cells. PMID: 24694617
  23. Subsarcolemmal mitochondria are more responsive than interfibrillar mitochondria to FGF-2-triggered protection from calcium-induced permeability transition, via a Cx43 channel-mediated pathway. PMID: 24654232
  24. Cultivating cells under hypoxic conditions and with bFGF is optimal for maintaining high viability and proliferation capacity of mesenchymal stem cells. PMID: 25715620
  25. The learning impairment in IL-1beta-treated rats is accompanied by lower FGF-2 mRNA levels in the medial prefrontal cortex and ventral (not dorsal) hippocampus, while TIMP-1 was not affected. PMID: 25697011
  26. Astroglial cell maturation is enhanced by bFGF through induction of miR-134. PMID: 25482448
  27. bFGF-induced differentiation of dorsal root ganglia stem cells toward Schwann cells might be mediated by binding to fibroblast growth factor receptor-1 (FGFR-1) through activation of the MAPK/ERK signal pathway. PMID: 24072480
  28. FGF2 is a key player in the origin and growth of neuronal and glial cells through autocrine and paracrine signaling. PMID: 24707873
  29. Research implicates FGF2 as a modifier of epigenetic mechanisms associated with emotional responsiveness, highlighting H3K9me3 as a key player in regulating affective vulnerability. PMID: 25071177
  30. Enhanced protein kinase C levels, reduction of basic FGF expression, and increased apoptosis might be associated with the development of diabetes-induced myoatrophy. PMID: 24008114
  31. Researchers studied the change of FGF-2 and IGF-1 in serum and bone callus after fracture in diabetic rats, exploring the molecular biological mechanism of diabetic fracture healing. PMID: 24418087
  32. TGF-beta1 and FGF2 induce the epithelial-mesenchymal transition of Hertwig's epithelial root sheath through a MAPK/ERK-dependent signaling pathway. PMID: 24610459
  33. Following FGF2 treatment, however, bHR-bLR differences in CCK and FGF-R1 mRNA expression were eliminated due to decreased CCK mRNA levels. PMID: 24121132
  34. Retinal injury may enhance neurotrophic factor expression in mesenchymal stem cells, promoting the repair process. PMID: 24030359
  35. Matrix proteoglycans such as perlecan serve as functional docking platforms for FGF2 in chronic transplant dysfunction. PMID: 24035513
  36. Basic fibroblast growth factor contributes to a shift in the angioregulatory activity of retinal glial (Muller) cells. PMID: 23861940
  37. With an increase in the degree of pressure ulcers, the expression of VEGF and bFGF in pressure ulcer tissue is decreased. This reduction in angiogenesis may be a crucial factor in the formation of pressure ulcers. PMID: 23740668
  38. This study provides evidence that E and FGF2 exert a cooperative effect on lactotroph proliferation, primarily through signaling initiated at the plasma membrane. PMID: 23651845
  39. Psychological stress could delay periodontitis healing in rats, which may be partly mediated by downregulation of the expression of bFGF in the periodontal ligament. PMID: 23326020
  40. FGF-2 secreted by bone marrow-derived cells strongly increases early glial proliferation, which can potentially improve peripheral nervous system regeneration. PMID: 22793996
  41. FGF-2 induced the phosphorylation of Akt and its substrate, glycogen synthase kinase 3beta (GSK3beta) in addition to three MAP kinases in rat glioma cells. PMID: 22575563
  42. Spinal cord treatment of a lesion with sciatic nerve and sciatic nerve plus FGF-2 allows recovery of hind limb movements compared to control, manifested by significantly higher behavioral scores after surgery. PMID: 22555431
  43. Inhibiting bFGF alleviates bleomycin-induced pulmonary fibrosis in rats. PMID: 20684286
  44. bFGF promotes the proliferation and migration of endothelial progenitor cells, the effects of bFGF being implemented by activating ERK signaling through the expression of Pdgfrb. PMID: 22731705
  45. The epithelial and smooth muscle cell hyperplasia and increased Fgf-2 expression observed in this experimental model of obesity/insulin-resistance might explain the high frequency of benign prostatic hyperplasia in insulin-resistant men. PMID: 22661309
  46. bFGF gene expression is elevated following cerebral concussion, and it might play a significant role in cell degeneration and necrosis. PMID: 12857442
  47. Data suggest that the molecular mechanism of dihydrotestosterone induction of Pfkfb4 (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4) during spermatogenesis involves stimulating Sertoli cells to secrete FGF-2. PMID: 22811469
  48. FGF-2 gene expression is significantly elevated from day 1 to day 14; the increase in FGF-2 protein level is most evident at day 7; cells expressing FGF-2 are primarily endothelial cells following myocardial infarction. PMID: 20674996
  49. The expression of VEGF and bFGF is significantly increased after stromal cell transplantation therapy during the late phase of acute myocardial infarction. PMID: 21162206
  50. Astrocyte migration to injury sites may be a key factor in the repair mechanisms orchestrated by FGF2. PMID: 22189091

Show More

Hide All

Database Links
Protein Families
Heparin-binding growth factors family
Subcellular Location
Secreted. Nucleus.
Tissue Specificity
Found in all tissues examined.

Q&A

What is FGF2 and what are its primary biological functions?

FGF2 (Fibroblast Growth Factor 2), also known as basic fibroblast growth factor (bFGF) or heparin-binding growth factor 2 (HBGF-2), is a multifunctional growth factor with diverse biological activities. It acts as a ligand for multiple fibroblast growth factor receptors including FGFR1, FGFR2, FGFR3, and FGFR4. Beyond receptor binding, FGF2 functions as an integrin ligand, specifically binding to integrin ITGAV:ITGB3, which is required for FGF2 signaling. FGF2 plays crucial roles in regulating cell survival, cell division, cell differentiation, and cell migration. In vitro studies have demonstrated that it functions as a potent mitogen. Additionally, FGF2 can induce angiogenesis and mediates phosphorylation of ERK1/2, which promotes retinal lens fiber differentiation .

What are the key characteristics of biotin-conjugated FGF2 antibodies?

Biotin-conjugated FGF2 antibodies, such as Rabbit Polyclonal FGF2 antibody-biotin (ab84027), are immunological tools designed for specific detection of FGF2 in various research applications. These antibodies are typically generated against recombinant full-length human FGF2 protein and validated for applications including Western Blotting (WB) and sandwich ELISA (sELISA). The biotinylation process enables enhanced detection sensitivity through streptavidin-based detection systems, leveraging the high-affinity biotin-streptavidin interaction. This characteristic makes these antibodies particularly valuable for detecting even low levels of FGF2 expression in experimental samples .

What experimental applications are biotin-conjugated FGF2 antibodies validated for?

Based on the available information, biotin-conjugated FGF2 antibodies have been validated for Western Blotting (WB) and sandwich ELISA (sELISA) applications specifically with human samples . The biotin conjugation provides significant advantages in these applications by enabling signal amplification through streptavidin-based detection systems. For Western blotting, these antibodies can detect FGF2 protein in complex biological samples after separation by gel electrophoresis. In sandwich ELISA, the biotin conjugation facilitates sensitive quantitative detection of FGF2 in solution through high-affinity interactions with streptavidin-conjugated reporter molecules. These applications are particularly valuable for studying FGF2 expression and quantification in research contexts.

How can researchers assess FGF2-receptor interactions using biotin-conjugated systems?

To evaluate FGF2-receptor interactions using biotin-conjugated systems, researchers can employ several methodological approaches. Biolayer interferometry (BLI) represents an effective technique as demonstrated in published research. For this approach, biotinylated extracellular domains of FGFR1c fused to Fc fragments are immobilized on streptavidin biosensors, followed by exposure to wild-type FGF2 or FGF2 conjugates . This method allows real-time monitoring of binding kinetics and determination of dissociation constants (Kd).

Another approach involves phosphorylation assays to assess downstream signaling activation. Researchers can treat cells expressing FGFRs (e.g., NIH 3T3 cells) with FGF2 for 15 minutes and then analyze activation of signaling pathways through Western blotting with anti-phospho-ERK 1/2 antibodies and anti-phospho-FGFR1 antibodies . This approach verifies whether FGF2 variants maintain their ability to stimulate downstream signaling at levels comparable to wild-type FGF2.

For cell-based receptor binding studies, flow cytometry can be employed using biotin-conjugated FGF2, followed by streptavidin-fluorophore detection. This enables quantification of FGF2 binding to cell surface receptors and comparison across different cell lines with varying receptor expression levels.

What protocols can be optimized for Western blotting using biotin-conjugated FGF2 antibodies?

When optimizing Western blotting protocols with biotin-conjugated FGF2 antibodies, researchers should consider several critical parameters for optimal results:

  • Sample preparation: Use complete protease inhibitor cocktails during protein extraction to preserve FGF2 integrity, as it can be sensitive to proteolytic degradation.

  • Gel electrophoresis: 10-12% SDS-PAGE gels are typically suitable for resolving FGF2 (~18-24 kDa depending on isoform).

  • Membrane selection: PVDF membranes may provide better protein retention than nitrocellulose for FGF2 detection.

  • Blocking strategy: Use BSA-based blockers (3-5%) rather than milk-based blockers, as milk contains endogenous biotin that might interfere with detection.

  • Antibody incubation: Optimize both concentration (typically starting at 1:1000 dilution) and incubation time (4°C overnight often yields best results).

  • Detection system: Employ streptavidin conjugated to HRP, fluorophores, or other detection molecules. For enhanced sensitivity, consider amplification systems like avidin-biotin complex (ABC) method.

  • Controls: Include appropriate positive (recombinant FGF2) and negative controls to validate detection specificity.

What methodological considerations are important for sandwich ELISA using biotin-conjugated FGF2 antibodies?

When implementing sandwich ELISA with biotin-conjugated FGF2 antibodies, researchers should optimize several key aspects of the protocol:

  • Antibody selection: Choose a capture antibody that recognizes a different epitope than the biotin-conjugated detection antibody to prevent competitive binding.

  • Plate coating: Coat high-binding 96-well plates with capture antibody (typically 1-5 μg/mL) in carbonate/bicarbonate buffer (pH 9.6) overnight at 4°C.

  • Blocking strategy: Use protein-free blocking buffer to minimize background while avoiding biotin-containing blocking agents like milk.

  • Sample preparation: Employ sample diluents that minimize matrix effects while preserving FGF2 immunoreactivity.

  • Standard curve: Prepare a dilution series using recombinant FGF2 (typically 0-1000 pg/mL) to establish assay linearity and sensitivity.

  • Detection system: Apply the biotin-conjugated FGF2 antibody at optimized concentration followed by streptavidin-HRP conjugate. Develop with appropriate substrate and measure spectrophotometrically.

  • Validation: Include controls for non-specific binding and confirm antibody specificity through spike-and-recovery and parallelism assessments.

How can FGF2-based systems be utilized in developing targeted cancer therapeutics?

The published research reveals sophisticated applications of FGF2 in developing targeted cancer therapeutics, particularly through the creation of FGF2-drug conjugates. Researchers have developed novel protein-drug conjugates using FGF2 as a targeting molecule for cancers overexpressing FGFR1. One advanced approach involves conjugating the cytotoxic agent monomethyl auristatin E (MMAE) to FGF2, creating conjugates that specifically target and kill FGFR1-positive cancer cells .

These FGF2-MMAE conjugates effectively target FGFR1-expressing cells, undergo FGFR1-mediated endocytosis, and exhibit cytotoxicity that correlates with FGFR1 expression levels . Studies have demonstrated that increasing the number of MMAE molecules attached to FGF2 (from 1 to 3) enhanced cytotoxic potency, highlighting the importance of optimizing drug loading.

For improved pharmacokinetic properties, advanced conjugates incorporating hydrophilic PEG4 or PEG27 spacers between FGF2 and MMAE have been developed. The conjugate with the highest hydrodynamic size (42 kDa) demonstrated superior cytotoxicity and effectively inhibited tumor growth in a mouse model of human breast cancer . This approach represents a promising alternative to antibody-drug conjugates for targeting FGFR1-overexpressing tumors.

What methodologies exist for site-specific conjugation to FGF2?

Site-specific conjugation to FGF2 can be achieved through several advanced methodological approaches:

  • Unnatural amino acid incorporation: Researchers have successfully incorporated propargyllysine (PrK) into FGF2 using amber codon suppression technology. The alkyne moiety in PrK enables highly specific conjugation through Cu(I)-catalyzed azide-alkyne 1,3-dipolar cycloaddition (CuAAC) with azide-functionalized molecules .

  • Cysteine-directed conjugation: FGF2 contains four cysteines, two of which are highly exposed and amenable to conjugation. Researchers have created FGF2 variants with controlled reactivity by selectively mutating specific cysteines (C78S/C96S mutations) while preserving others for targeted modification .

  • Terminal extensions: N- and C-terminal extensions containing cysteine residues (such as KCK tags) have been engineered into FGF2 to provide additional conjugation sites without disrupting the protein's core structure and biological activity .

  • Reaction optimization: For challenging conjugations, particularly with hydrophobic molecules, detergent-based approaches have demonstrated improved efficiency, achieving nearly 100% yield in difficult cases .

These methodological advances enable the creation of defined, homogeneous FGF2 conjugates with preserved biological activity for various research and therapeutic applications.

How does FGF2 maintain biological activity after conjugation?

The impact of conjugation on FGF2's biological activity depends on the conjugation strategy and modification sites. According to published studies, carefully designed FGF2 conjugates maintain their ability to bind receptors and activate downstream signaling. Research has demonstrated that FGF2 variants with cysteine-to-serine substitutions and N/C-terminal extensions, as well as their conjugates with vcMMAE, stimulated downstream signaling at levels comparable to wild-type FGF2, as evidenced by phosphorylation of ERK 1/2 and FGFR1 .

Biolayer interferometry (BLI) analysis has confirmed that FGF2 conjugates retained binding to the recombinant extracellular domain of FGFR1c, demonstrating that the introduced modifications did not impair receptor recognition . Additionally, fluorescently labeled FGF2 conjugates demonstrated preserved cellular internalization via FGFR1-mediated endocytosis, indicating functional retention of this critical biological property.

The data suggests that FGF2 is more robust than some other growth factors (like FGF1) in maintaining stability after modification. FGF2's higher resistance to thermal unfolding, aggregation, and proteolysis allows it to better tolerate conjugation while preserving functionality . This intrinsic stability makes FGF2 particularly valuable as a targeting molecule for bioconjugation strategies in research and therapeutic applications.

What factors affect the cytotoxicity of FGF2-cytotoxic drug conjugates in FGFR1-expressing cells?

Several critical factors influence the cytotoxicity of FGF2-cytotoxic drug conjugates in FGFR1-expressing cells:

  • Drug-to-protein ratio (DPR): Research demonstrates that conjugates bearing three monomethyl auristatin E (MMAE) molecules exhibited higher cytotoxicity than those with fewer attached drug molecules, confirming a direct relationship between drug loading and in vitro cytotoxicity .

  • FGFR1 expression level: Cytotoxicity studies across multiple cell lines revealed a clear correlation between FGFR1 expression and sensitivity to FGF2-MMAE conjugates. Cells with high FGFR1 expression (U2OS-R1) showed significantly greater sensitivity than those with low or undetectable FGFR1 levels (U2OS), demonstrating the targeting specificity of these conjugates .

  • Internalization efficiency: Confocal microscopy studies confirmed that FGF2 conjugates were effectively internalized through FGFR1-mediated endocytosis. This internalization is essential for the release of the active cytotoxic agent within target cells .

  • Linker chemistry: The valine-citrulline (vc) linker used in FGF2-vcMMAE conjugates enables specific cleavage by intracellular cathepsins following endocytosis, releasing active MMAE inside the cell. Linker design must balance stability in circulation with efficient drug release in target cells .

  • Hydrodynamic size: Among conjugates studied, those with higher hydrodynamic size (42 kDa) exhibited optimal in vivo efficacy in a mouse model of human breast cancer, suggesting that size optimization can enhance therapeutic outcomes .

These factors provide important considerations for researchers designing and optimizing FGF2-based targeted therapeutics.

How can researchers optimize conjugation reactions for creating FGF2 conjugates?

Optimizing conjugation reactions for FGF2 conjugates requires systematic evaluation of multiple parameters:

  • Site-specific strategy selection:

    • For cysteine-directed conjugation using maleimide chemistry, buffer should be PBS pH 7.2-7.4 with 1-5 mM EDTA to prevent disulfide formation

    • For click chemistry (CuAAC) with unnatural amino acids like propargyllysine, consider Cu(I) catalyst concentration, Cu(I)-stabilizing ligands, and mild detergents

  • Reaction conditions optimization:

    • Molar ratio of conjugation reagent to protein (typically 5-20 fold molar excess)

    • Reaction temperature and time (often 4°C overnight balances efficiency with protein stability)

    • For hydrophobic molecules, addition of mild detergents (0.05-0.1% Tween-20 or Triton X-100) improves solubility and reaction efficiency

  • Purification strategy:

    • Size exclusion chromatography to remove unreacted small molecules

    • Additional chromatographic steps for higher purity if needed

  • Quantification methods:

    • Degree of conjugation should be quantified using methods such as mass spectrometry or spectrophotometric assays

    • Functional activity assays to ensure biological function is preserved

Published research demonstrates that optimizing these parameters can achieve nearly 100% conjugation efficiency even for challenging reactions with hydrophobic compounds like MMAE .

What analytical methods confirm successful conjugation to FGF2?

Researchers can employ several analytical techniques to verify successful conjugation to FGF2:

  • Mass spectrometry: MALDI-TOF or ESI-MS analysis reveals mass shifts corresponding to the addition of conjugated molecules. LC-MS/MS following proteolytic digestion can identify specific modified residues with high precision.

  • SDS-PAGE with appropriate detection: For biotin conjugates, samples can be analyzed by gel electrophoresis followed by detection with streptavidin-HRP or streptavidin-fluorophore conjugates. Band shifts may be observed with multiple additions.

  • Functional binding assays: Biolayer interferometry (BLI) studies have been successfully employed to confirm that FGF2 conjugates maintain binding to recombinant FGFR1c extracellular domains, verifying that the conjugation did not disrupt receptor recognition .

  • Cell-based assays: Confocal microscopy using fluorescently labeled FGF2 conjugates has demonstrated preserved cellular internalization via FGFR1-mediated endocytosis, confirming functional integrity .

  • Signaling pathway activation: Western blotting with anti-phospho-ERK 1/2 antibodies and anti-phospho-FGFR1 has been used to verify that FGF2 conjugates maintain the ability to stimulate downstream signaling pathways, confirming biological activity post-conjugation .

These complementary approaches provide comprehensive verification of both the chemical success of conjugation and the preservation of biological function.

How do FGF2-based therapeutic approaches compare to other targeted strategies?

FGF2-based therapeutic approaches offer several distinctive advantages and limitations compared to other targeted strategies:

Targeting StrategyAdvantagesLimitationsResearch Context
FGF2-drug conjugates- Natural ligand with high affinity for FGFRs
- Smaller size may improve tumor penetration
- More thermally stable than some antibodies
- Lower production costs than antibodies
- Shorter half-life than antibodies
- Potential off-target effects on multiple FGFR-expressing tissues
- Less established clinical track record
Particularly promising for FGFR1-overexpressing cancers including certain breast cancers
Antibody-drug conjugates- High specificity
- Extended half-life
- Established clinical precedent
- Potential for ADCC
- Large size limits tumor penetration
- Complex manufacturing
- Higher immunogenicity risk
- Higher production costs
Currently the dominant approach in targeted cancer therapeutics
Small molecule FGFR inhibitors- Oral bioavailability
- Good tissue penetration
- Lower manufacturing costs
- Limited selectivity between FGFR subtypes
- Rapid clearance
- Development of resistance
Used clinically for various FGFR-driven cancers

Research has demonstrated that FGF2-MMAE conjugates effectively target FGFR1-expressing cells and exhibit cytotoxicity that correlates with FGFR1 expression levels . The incorporation of hydrophilic PEG spacers has further improved pharmacokinetic properties, with conjugates of optimal hydrodynamic size (42 kDa) showing effective inhibition of tumor growth in mouse models of human breast cancer .

These findings position FGF2-based approaches as promising alternatives to antibody-drug conjugates, particularly for targeting cancers with FGFR1 overexpression, where they may offer advantages in manufacturing simplicity, cost, and tumor penetration.

What methodological considerations exist for evaluating FGF2 conjugate internalization?

Evaluating FGF2 conjugate internalization requires careful methodological design:

  • Fluorescent labeling strategies:

    • Published research has successfully employed DyLight 550-labeled FGF2 conjugates to track cellular uptake via confocal microscopy

    • Dual labeling approaches can simultaneously track both the FGF2 carrier and its conjugated cargo

  • Cell model selection:

    • Comparative analysis using cell lines with varying FGFR1 expression levels provides critical controls

    • Co-culture experiments with prestained cell populations (e.g., CellTrace Violet) allow direct comparison on the same experimental platform

  • Time-course analysis:

    • Tracking internalization kinetics at multiple timepoints provides information on uptake rates

    • Pulse-chase experimental designs can distinguish surface-bound from internalized conjugates

  • Colocalization studies:

    • Simultaneous visualization of FGF2 conjugates with endosomal/lysosomal markers confirms trafficking through expected compartments

    • Quantitative colocalization analysis provides objective measurement of internalization efficiency

  • Biochemical fractionation:

    • Cell surface biotinylation followed by streptavidin pull-down can biochemically separate surface-bound from internalized conjugates

    • Subcellular fractionation allows quantitative distribution analysis across cellular compartments

These methodological approaches collectively provide comprehensive assessment of FGF2 conjugate internalization, both qualitatively and quantitatively, enabling researchers to optimize conjugate design for efficient cellular delivery.

How should researchers interpret results from different FGF2-receptor interaction assays?

When researchers encounter results from different FGF2-receptor interaction assays, systematic interpretation should consider several key factors:

  • Methodological differences:

    • Cell-free binding assays (BLI, SPR) measure direct physical interactions under controlled conditions

    • Cell-based assays incorporate additional complexities including co-receptors, extracellular matrix, and cellular trafficking

    • Phosphorylation assays measure downstream signaling rather than direct binding

  • The critical role of co-factors:

    • Heparan sulfate proteoglycans (HSPGs) function as essential co-receptors stabilizing FGF2-FGFR binding

    • Presence or absence of these co-factors between assay systems may cause apparent discrepancies in affinity measurements

  • Experimental conditions:

    • Buffer composition, pH, temperature, and ion concentrations significantly impact FGF2-receptor interactions

    • Physiological calcium concentrations affect FGFR dimerization and subsequent signaling

  • Receptor isoform specificity:

    • FGF2 interacts differentially with FGFR1c, FGFR3c, and FGFR4

    • Splice variants within these receptor families show distinct binding characteristics

The most robust interpretation comes from integrating multiple complementary approaches. For example, published research has employed both direct binding studies (BLI) and functional signaling assays (phosphorylation of ERK1/2 and FGFR1) to comprehensively characterize FGF2 conjugates . This multi-method approach provides more complete understanding of both physical binding properties and functional consequences of FGF2-receptor interactions.

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.