FGFR1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Structure

FGFR1 Recombinant Monoclonal Antibodies are immunoglobulin-based molecules produced via recombinant methods, targeting extracellular or intracellular domains of FGFR1. Key structural features include:

  • Antigen-binding regions: Complementarity-determining regions (CDRs) tailored to bind FGFR1 epitopes (e.g., D1 domain, heparin-binding domain adjacent sequences) .

  • Valency: Engineered as monovalent (scFv) or bivalent/multivalent (scFv-Fc, tetravalent) formats to modulate receptor clustering and internalization .

  • Species cross-reactivity: Some antibodies bind human, mouse, and macaque FGFR1 isoforms (e.g., 1Mb and IIIc) .

Mechanism of Action

These antibodies mediate FGFR1 signaling modulation through distinct pathways:

MechanismExample AntibodyEffect
Receptor antagonismIMB-R1 Blocks FGF binding to FGFR1 by masking the heparin-binding domain (HBD).
Receptor dimerizationscFvD2-Fc Induces FGFR1 clustering, triggering clathrin-mediated endocytosis (CME).
Degradation inductionTetravalent antibodies Enhances receptor internalization and lysosomal degradation.
Signal inhibitionAnti-D1 domain antibodies Prevents β-Klotho co-receptor binding, disrupting FGF23 signaling.

3.1. Target Epitopes and Binding

  • D1 domain: Antibodies like scFvD2-Fc bind FGFR1’s regulatory D1 domain, blocking β-Klotho interaction without affecting FGF1 binding .

  • HBD-adjacent sequence: IMB-R1 targets the SSSEEKETDNTKPNR peptide near the HBD, inhibiting FGF2-driven cell growth .

  • Splice variant specificity: Certain antibodies selectively bind FGFR1 isoforms (e.g., IIIc) overexpressed in cancers .

3.2. Functional Outcomes

  • Internalization efficiency:

    • Bivalent antibodies (e.g., scFvD2-Fc) induce 80–90% FGFR1 internalization via CME .

    • Tetravalent formats split internalization between CME and clathrin-independent pathways, improving degradation .

  • Receptor activation: Monovalent scFv formats fail to activate FGFR1 or induce degradation .

3.3. Preclinical Efficacy

  • Cancer cell lines:

    • IMB-R1 inhibits FGFR1-positive sarcoma cell growth (IC₅₀: 0.5–1 μg/mL) and induces apoptosis .

    • Anti-D1 antibodies reduce ERK1/2 phosphorylation by 70% in NIH3T3 cells .

  • In vivo models: Patent data suggest FGFR1 antibodies suppress tumor growth in xenograft models of NSCLC and breast cancer .

Clinical and Therapeutic Implications

  • Oncology: FGFR1 antibodies are explored in antibody-drug conjugates (ADCs) for cancers with FGFR1 amplifications (e.g., lung, breast) .

  • Autoimmune diseases: Early studies suggest utility in rheumatoid arthritis by modulating FGF23-driven inflammation .

  • Limitations: Off-target effects due to FGFR family homology (e.g., FGFR2/3) require isoform-specific engineering .

Table 1: Antibody-Induced FGFR1 Internalization

Antibody FormatInternalization EfficiencyPathwayDegradation
Monovalent scFv<10%NoneNo
Bivalent scFv-Fc80–90%Clathrin-mediatedYes
Tetravalent95%CME + Clathrin-independentYes

Table 2: FGFR1 mRNA Expression in Cancer Cells

Cell LineFGFR1 Expression (Fold vs. FGFR2/3)
MG63 (osteosarcoma)>100
MDA-MB468 (breast)>100
T47D (breast)10

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 the products within 1-3 business days after receiving your order. The delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
Basic fibroblast growth factor receptor 1 antibody; bFGF-R-1 antibody; BFGFR antibody; CD331 antibody; CEK antibody; FGFBR antibody; FGFR 1 antibody; FGFR-1 antibody; FGFR1 antibody; FGFR1/PLAG1 fusion antibody; FGFR1_HUMAN antibody; fibroblast growth factor receptor 1 antibody; FLG antibody; FLT-2 antibody; FLT2 antibody; Fms-like gene antibody; Fms-like tyrosine kinase 2 antibody; fms-related tyrosine kinase 2 antibody; HBGFR antibody; heparin-binding growth factor receptor antibody; HH2 antibody; HRTFDS antibody; hydroxyaryl-protein kinase antibody; KAL2 antibody; N-SAM antibody; OGD antibody; Proto-oncogene c-Fgr antibody
Target Names
Uniprot No.

Target Background

Function
Fibroblast growth factor receptor 1 (FGFR1) is a tyrosine-protein kinase that functions as a cell-surface receptor for fibroblast growth factors. It plays a crucial role in regulating embryonic development, cell proliferation, differentiation, and migration. FGFR1 is essential for normal mesoderm patterning, proper axial organization during embryonic development, normal skeletogenesis, and the normal development of the gonadotropin-releasing hormone (GnRH) neuronal system.

FGFR1 phosphorylates several downstream targets, including PLCG1, FRS2, GAB1, and SHB. Ligand binding to FGFR1 activates multiple signaling cascades. Activation of PLCG1 leads to the production of diacylglycerol and inositol 1,4,5-trisphosphate, crucial signaling molecules within cells. Phosphorylation of FRS2 initiates the recruitment of GRB2, GAB1, PIK3R1, and SOS1, ultimately mediating the activation of RAS, MAPK1/ERK2, MAPK3/ERK1, and the MAP kinase signaling pathway. It also activates the AKT1 signaling pathway. Additionally, FGFR1 promotes phosphorylation of SHC1, STAT1, and PTPN11/SHP2.

Within the nucleus, FGFR1 enhances the activity of RPS6KA1 and CREB1, contributing to the regulation of transcription. The signaling of FGFR1 is downregulated by IL17RD/SEF and by FGFR1 ubiquitination, internalization, and subsequent degradation.
Gene References Into Functions
  1. Myeloid/lymphoid neoplasms with FGFR1 rearrangement are a rare entity, with no distinct clinical phenotype. FGFR rearrangement confirmation by FISH should be performed in any hematological malignancy with 8p translocation. PMID: 29119847
  2. Amplifications of CCND1, C-MYC, and FGFR1 were observed in 34.28%, 28.57%, and 17.14% of the 35 samples (invasive ductal breast carcinoma). PMID: 30119151
  3. High FGFR1 expression is associated with Peritoneal Dissemination Via Epithelial-to-Mesenchymal Transition in Gastric Cancer. PMID: 29976636
  4. The present study aimed to evaluate the relationship between a common FGFR1 single nucleotide polymorphism (rs13317) with craniofacial morphology. PMID: 29872111
  5. Clinical outcomes of myeloid/lymphoid neoplasms with fibroblast growth factor receptor-1 (FGFR1) rearrangement PMID: 29486661
  6. This study suggests that genomic alterations involving the cell cycle (TP53, CCND1, CDKN2A), as well as FGFR1 amplifications and tumor genomic alterations burden are prognostic biomarkers of survival in head and neck squamous cell carcinoma. PMID: 29331751
  7. A novel heterozygous frameshift mutation c.299_300insCCGCAGACTCCGGCCTCTATGC (p.C101Rfs*17) associated with Kallmann syndrome PMID: 29658329
  8. FGFR3, along with its downstream regulatory PI3K/AKT kinases, may serve as potential biomarkers for the invasiveness and prognosis of laryngeal cancer. PMID: 29299828
  9. These experiments presented a new mechanism adopted by GDNF supporting glioma development and indicated a possible therapeutic potential via the inhibition of proN-cadherin/FGFR1 interaction. PMID: 29750313
  10. There was no significant difference in the expression of FGFR1 between different types of circulating tumor cells. PMID: 29764586
  11. Our data may facilitate the design of therapeutically relevant targeting molecules for selective treatment of FGFR1 overproducing cancers PMID: 29748524
  12. This study found infrequent BRAF alterations but enriched FGFR alterations in adults compared to that reported in pediatric pilocytic astrocytomas. Additionally, coexistent BRAF and FGFR alterations and a significant association of FGFR alterations with age and tumor location were noted. PMID: 27608415
  13. SNP rs17182023 was correlated with reduced breast cancer risk and was associated with FGFR1 protein expression. High FGFR1 protein expression was an independent risk factor of breast cancer and resulted in poor prognosis. PMID: 29996114
  14. Besides RET and HRAS, FGFR1 is only the third protooncogene found to be recurrently mutated in pheochromocytomas. PMID: 29159601
  15. This research focuses on the treatment of patients with breast cancer and FGFR1 amplifications. PMID: 29223982
  16. This study presents the atomic structure of a 1:1:1 ternary complex consisting of the shed extracellular domain of alpha-klotho, the FGFR1c ligand-binding domain, and FGF23. In this complex, alpha-klotho simultaneously tethers FGFR1c by its D3 domain and FGF23 by its C-terminal tail, thereby implementing FGF23-FGFR1c proximity and conferring stability PMID: 29342138
  17. This study identified FGFR1, a promoter of glycolysis-related enzyme, as the target of miR-361 that promoted glycolysis and repressed oxidative phosphorylation in breast cancer cells. FGFR1 mediated the anti-glycolytic function of miR-361 by regulating the activity of PDHK1 and LDHA. PMID: 29132384
  18. FGFR1 and/or FGF3 gene amplification correlated with a lower pathologic complete response in patients with HER2(+) early breast cancer treated with neoadjuvant anti-HER2 therapy. PMID: 28381415
  19. Data demonstrated that FOXC1 binds to an Fgfr1 upstream regulatory region and that FOXC1 activates an Fgfr1 promoter element. Furthermore, elevated expression of Foxc1 led to increased Fgfr1-IIIc transcript promoting invasion after TGFbeta1-induced EMT. PMID: 28684636
  20. These results suggest that FGFR1 gene amplification is a frequent alteration in squamous cell carcinoma of the lung and appears not to be a negative but rather a favorable prognostic marker for women and particularly for patients with advanced disease PMID: 29270870
  21. These data suggest that the ERalpha pathway remains active in estrogen-deprived ER(+)/FGFR1-amplified breast cancers. Therefore, these tumors are endocrine resistant and should be candidates for treatment with combinations of ER and FGFR antagonists. PMID: 28751448
  22. Amplification of gene FGFR1 is associated with lung adenocarcinoma. PMID: 28381877
  23. Lysosomal sequestration, resulting in an organelle-specific and pH-dependent nintedanib fluorescence, was identified as an intrinsic resistance mechanism in FGFR-driven lung cancer cells. Accordingly, combination treatment of nintedanib with agents compromising lysosomal acidification (bafilomycin A1, chloroquine) exerted distinctly synergistic growth inhibitory effects PMID: 28882160
  24. The close proximity between AcSDKP and FGFR1 was essential for the suppression of TGFbeta/smad signaling and EndMT associated with MAP4K4 phosphorylation (P-MAP4K4) in endothelial cells. PMID: 28771231
  25. This study reports a highly specific internalizing antibody fragment that can serve as a therapeutic targeting agent for efficient delivery of cytotoxic drugs into FGFR1-positive lung cancer cells. PMID: 28483948
  26. Anlotinib inhibits the activation of VEGFR2, PDGFRbeta, and FGFR1, as well as their common downstream ERK signaling PMID: 29454091
  27. Missense mutations in COL6A1, COL11A2, FGFR1, and BMP2 genetically predispose patients to ossification of posterior longitudinal ligaments. PMID: 27246988
  28. High levels of FGFR1 are associated with non-small cell lung cancer. PMID: 28558758
  29. The results of this study designate nFGFR1 signaling as a potential common dysregulated mechanism in investigated patients and a potential therapeutic target in schizophrenia. PMID: 28094170
  30. Findings indicate the great variability of fibroblast growth factor receptor 1 (FGFR1) mutation phenotypes in idiopathic hypogonadotropic hypogonadism (IHH) or Kallmann syndrome (KS). PMID: 28008864
  31. These results show that FGFR1 polymorphism influences lower anterior face height, the distance from the upper lip to the nasal floor, and lip shape PMID: 28415752
  32. Fibrolamellar carcinomas show polysomy of chromosome 8 and the FGFR1 locus, with only modest mRNA expression and weak or absent expression at the protein level. FGFR2 rearrangement was not detected. PMID: 26259677
  33. Endothelin-A receptor-activated ABCB1 expression plays a role in nintedanib resistance in FGFR1-driven small cell lung cancer PMID: 27367030
  34. Loss of FGFR1 generates a gene signature that is reverse correlated with FGFR1 gene amplification and/or upregulation in human breast cancer. Our results suggest that FGFR1 signaling is a key pathway driving breast cancer lung metastasis and that targeting FGFR1 in breast cancer is an exciting approach to inhibit metastasis. PMID: 28433771
  35. Combination treatment with AKT and FGFR kinase inhibitors have additive effects on malignant phenotypes in vitro and in vivo by inhibiting multiple signaling pathways and mitigating the compensatory upregulation of FGFR signaling induced by AKT kinase inhibition. PMID: 28008155
  36. FGFR1/MAPK may be important for brachyury activation in lung cancer, and this pathway may be an appealing therapeutic target for a subset of brachyury-driven lung cancer. PMID: 27893433
  37. FGFR1 alteration mainly represented by FGFR1-ITD is a frequent event in dysembryoplastic neuroepithelial tumors. Digital droplet PCRtrade mark is an easy and alternative method than whole-genome sequencing to detect FGFR1-ITD in Formalin-fixed paraffin-embedded brain tumors, in routine practice. PMID: 27791984
  38. This study reports dramatic upregulation of fibroblast growth factor receptor 1 (FGFR1) and its cognate ligand FGF2 in both acquired and inherently resistant breast cancer cells. PMID: 27825137
  39. This study reveals a stringent association between FGFR and the downstream effector c-Myc in FGFR-dependent cancers, and suggests the potential therapeutic value of c-Myc in FGFR-targeted cancer therapy. PMID: 27401245
  40. Elevated FGFR3 and FGFR1 protein expression is common in aggressive ependymomas but likely not driven by genetic alterations. Further studies are warranted to evaluate whether ependymoma patients with high FGFR3 and/or FGFR1 expression could benefit from treatment with FGFR inhibitor-based therapeutic approaches currently under evaluation in clinical trials PMID: 28468611
  41. These data identify FGFR1 as a driver gene in multiple soft-tissue sarcoma subtypes and support FGFR1 inhibition, guided by patient selection according to the FGFR1 expression and monitoring of MAPK-ERK1/2 signaling, as a therapeutic option in this challenging group of diseases PMID: 27535980
  42. Our results demonstrated that the AcSDKP-FGFR1 signaling pathway is critical for maintaining mitochondrial dynamics by control of miR let-7b-5p in endothelial cells. PMID: 29269295
  43. Increased FGFR1 CN was observed in two racial groups not previously reported: African Americans and Native Americans. However, FGFR1 amplification is not prognostic in laryngeal squamous cell carcinomas PMID: 29351293
  44. This brief communication reports on a patient with an exceedingly rare "8p11 (eight-p-eleven) myeloproliferative syndrome" (EMS) with CEP110-FGFR1 rearrangement who responded to treatment with the multi-tyrosine kinase inhibitor (TKI) dasatinib PMID: 28242791
  45. This research aims to identify mutually exclusive activating hotspot mutations in FGFR1 and related PI-3K/RAS signaling genes in malignant phyllodes tumors, which are implicated in tumor pathogenesis and/or progression. PMID: 27255162
  46. We report FGFR1 as being frequently overexpressed in HNSCC and as a candidate prognostic biomarker in HPV-negative HNSCC. PMID: 26936917
  47. Head and neck cancers are recurrently affected by FGFR1 amplification, with a predominance in cancers of the oral cavity. PMID: 29022097
  48. High FGFR1 expression is associated with non-small cell lung cancer. PMID: 26936993
  49. This study presents a rare case of a 46,XY patient with CHD associated with ambiguous genitalia consisting of a clitoris-like phallus and a bifid scrotum. Exome sequencing revealed novel homozygous mutations in the FGFR1 and STARD3 genes that may be associated with the phenotype. PMID: 27055092
  50. PDGFRalpha levels are regulated by SMARCB1 expression, and assessment of clinical specimens documents the expression of both PDGFRalpha and FGFR1 in rhabdoid tumor patients. PMID: 27783942

Show More

Hide All

Database Links

HGNC: 3688

OMIM: 101600

KEGG: hsa:2260

STRING: 9606.ENSP00000393312

UniGene: Hs.264887

Involvement In Disease
Pfeiffer syndrome (PS); Hypogonadotropic hypogonadism 2 with or without anosmia (HH2); Osteoglophonic dysplasia (OGD); Hartsfield syndrome (HRTFDS); Trigonocephaly 1 (TRIGNO1); Encephalocraniocutaneous lipomatosis (ECCL); Jackson-Weiss syndrome (JWS)
Protein Families
Protein kinase superfamily, Tyr protein kinase family, Fibroblast growth factor receptor subfamily
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Nucleus. Cytoplasm, cytosol. Cytoplasmic vesicle. Note=After ligand binding, both receptor and ligand are rapidly internalized. Can translocate to the nucleus after internalization, or by translocation from the endoplasmic reticulum or Golgi apparatus to the cytosol, and from there to the nucleus.
Tissue Specificity
Detected in astrocytoma, neuroblastoma and adrenal cortex cell lines. Some isoforms are detected in foreskin fibroblast cell lines, however isoform 17, isoform 18 and isoform 19 are not detected in these cells.

Q&A

What is FGFR1 and why are monoclonal antibodies against it important for research?

Fibroblast Growth Factor Receptor 1 (FGFR1) is a transmembrane receptor that regulates essential cellular processes including division, motility, metabolism, and cell death. FGFR1 overexpression is observed in numerous tumors, making it an attractive target for cancer treatment strategies . Monoclonal antibodies against FGFR1 serve as crucial tools for studying receptor biology, examining expression patterns in normal versus diseased tissues, and developing targeted therapeutics. These antibodies enable researchers to visualize receptor localization, quantify expression levels, explore signaling mechanisms, and potentially develop therapeutic approaches that exploit receptor dynamics without activating downstream oncogenic pathways .

What are the major structural domains of FGFR1 and how do antibodies target them?

FGFR1 contains several structural domains with the extracellular portion consisting of three immunoglobulin-like domains (D1, D2, and D3) in α isoforms or two domains in β isoforms . Most FGFR1 antibodies are designed to target specific domains, with different functional consequences depending on the binding site. For example, antibodies binding to the D1 domain (like scFvD2, scFvC1, and scFvE2) do not interfere with FGF1 binding but can inhibit interactions with co-receptors like β-Klotho . The D2-D3 domains constitute the primary FGF binding region, and antibodies targeting these regions may directly affect ligand binding. The membrane-proximal Ig-like domain can be encoded by alternative exons yielding IIIb or IIIc isoforms , which can be specifically targeted by antibodies like the Human FGFR1 (IIIb) Antibody (clone #133111) .

How do different antibody formats affect FGFR1 receptor dynamics?

The valency of antibodies significantly impacts FGFR1 receptor dynamics. Research demonstrates that:

  • Monovalent antibody fragments (scFv format) bind FGFR1 but typically do not induce receptor dimerization, internalization, or downstream signaling

  • Bivalent antibody fragments (scFv-Fc format) can induce receptor dimerization and trigger clathrin-mediated endocytosis (CME) without activating downstream signaling

  • Tetravalent antibody formats enhance receptor clustering, dramatically improve internalization efficiency, and engage both CME and clathrin-independent endocytosis (CIE) pathways

This valency-dependent behavior provides researchers with tools to selectively modulate FGFR1 clustering, internalization, and degradation without activating the receptor, which has significant implications for targeted drug delivery approaches .

What are the optimal experimental conditions for using FGFR1 antibodies in neutralization assays?

For neutralization assays using FGFR1 antibodies, specific experimental conditions must be carefully established. For instance, when using Human FGFR1 (IIIb) Antibody (clone #133111), the typical working concentration (ND50) is 0.075-0.3 μg/mL in the presence of 6 ng/mL Recombinant Human FGF R1 alpha (IIIb) Fc Chimera, 0.3 ng/mL Recombinant Human FGF acidic, and 10 μg/mL heparin . The inclusion of heparin is crucial as it acts as a necessary cofactor for FGF binding to the receptor, which induces the proliferation that the antibody neutralizes . Researchers should determine optimal dilutions for each specific application through titration experiments. When designing neutralization assays, control experiments should include FGF1 alone to establish baseline activation levels and non-targeting antibodies to confirm specificity of inhibition.

How should researchers validate FGFR1 antibody specificity across FGFR family members?

Validating FGFR1 antibody specificity requires comprehensive cross-reactivity testing against all FGFR family members. Surface Plasmon Resonance (SPR) represents an effective methodology for this purpose. Researchers can immobilize FGFR1-Fc, FGFR2-Fc, FGFR3-Fc, and FGFR4-Fc proteins on CM5 sensors, then independently inject the antibody of interest at a standardized concentration (e.g., 1 μM) over all sensors at controlled flow rates (e.g., 30 μL/min) . By monitoring the association and dissociation phases (typically for 240 seconds each), researchers can quantitatively assess binding specificity. Positive control proteins like FGF1 that bind all FGFRs should be included, and measurements should be conducted in standardized buffers (such as PBS-PN: PBS with 0.005% surfactant P20, 0.02% NaN3; pH 7.2) . Cross-reactivity profiles can then be quantitatively compared using data analysis software like BIAevaluation 4.1.

What techniques effectively identify the epitope binding sites of FGFR1 antibodies?

To identify epitope binding sites of FGFR1 antibodies, researchers should employ multiple complementary techniques:

  • Pull-down assays with truncated receptors: Generate recombinant proteins representing full extracellular domains (D1-D2-D3) and truncated forms lacking specific domains (e.g., D2-D3 without D1). Testing antibody binding against these constructs can identify domain-specific interactions .

  • Direct binding to isolated domains: Produce recombinant individual domains (e.g., GST-tagged D1) and assess antibody binding. This confirms direct interaction with specific domains .

  • Surface Plasmon Resonance (SPR) epitope binning: Immobilize FGFR1 on a sensor chip and test antibodies in pairwise combinations to determine whether they compete for the same binding site .

  • Bio-layer interferometry (BLI): Immobilize FGFR1 on AR2G biosensors and analyze how antibody binding affects interactions with other binding partners like β-Klotho to further characterize functional epitopes .

How can researchers quantitatively assess FGFR1 internalization and degradation triggered by antibodies?

Quantitative assessment of FGFR1 internalization and degradation requires multiple experimental approaches:

  • Cycloheximide chase assays: Treat cells with cycloheximide (10 μg/ml) to inhibit protein synthesis, then add antibodies or FGF1 and monitor FGFR1 levels by Western blotting at different time points (e.g., up to 180 minutes). This approach allows for tracking receptor degradation kinetics without the confounding effect of new receptor synthesis .

  • Immunofluorescence microscopy: Fluorescently label antibodies and track their internalization and colocalization with endocytic markers. Comparing monovalent (scFv) versus bivalent (scFv-Fc) formats can reveal mechanistic differences in internalization pathways .

  • Quantitative Western blotting: Measure FGFR1 levels after antibody treatment, normalizing phosphorylated receptor signal to total receptor protein. For comparative studies, set the positive control (e.g., FGF1 stimulation) to 100% and express all other conditions relative to this baseline .

  • Proximity ligation assays: For studying receptor-antibody interactions in situ, techniques like the Duolink In Situ Assay can be used to visualize and quantify molecular proximities at the single-molecule level .

How do antibody-induced and ligand-induced FGFR1 internalization mechanisms differ?

Antibody-induced and ligand-induced FGFR1 internalization proceed through distinct mechanisms with important biological consequences:

Ligand-induced internalization (e.g., by FGF1) typically involves:

  • Binding to D2-D3 domains of FGFR1

  • Receptor dimerization and transphosphorylation

  • Activation of downstream signaling cascades

  • Clathrin-mediated endocytosis (CME)

  • Time-dependent receptor degradation in the presence of protein synthesis inhibitors

In contrast, antibody-induced internalization shows valency-dependent effects:

  • Monovalent antibodies (scFv format) generally do not induce significant FGFR1 internalization or degradation

  • Bivalent antibodies (scFv-Fc format) trigger receptor dimerization and CME without receptor activation

  • Tetravalent antibodies split internalization between CME and clathrin-independent endocytosis (CIE), dramatically improving internalization efficiency and receptor degradation

Critically, research demonstrates that both CME and CIE of FGFR1 triggered by antibodies do not require receptor activation, making them promising for targeted drug delivery applications that avoid activating oncogenic signaling pathways .

What molecular mechanisms explain how bivalent antibodies can induce FGFR1 dimerization without activating the receptor?

Bivalent antibodies can induce FGFR1 dimerization without activating the receptor through several molecular mechanisms:

  • Domain-specific binding: Antibodies that bind to D1 domain (rather than the ligand-binding D2-D3 region) can induce dimerization without mimicking the conformational changes triggered by natural ligands. Studies with scFvD2-Fc, scFvC1-Fc, and scFvE2-Fc demonstrate that these D1-binding antibodies can dimerize FGFR1 without inducing receptor phosphorylation or downstream ERK1/2 activation .

  • Non-productive dimerization geometry: The spatial arrangement of receptors in antibody-induced dimers differs from the precise orientation required for kinase domain transphosphorylation in ligand-activated dimers.

  • Receptor conformation: Bivalent antibodies may not induce the same conformational changes in the receptor that occur upon ligand binding, particularly in the intracellular kinase domains.

  • Compatibility with ligand binding: Remarkably, D1-binding antibodies do not interfere with FGF1-FGFR1 interaction, as demonstrated by chemical crosslinking and formation of ternary complexes containing FGF1, FGFR1, and antibody fragments . This suggests that antibody-induced dimerization does not recapitulate the active conformation of the receptor.

How does FGFR1 clustering affect the choice between clathrin-mediated and clathrin-independent endocytosis pathways?

The oligomeric state of FGFR1 in the plasma membrane dictates the endocytic pathway choice. Research using engineered antibodies of different valency demonstrates that:

  • Bivalent antibodies predominantly trigger clathrin-mediated endocytosis (CME) of FGFR1

  • Tetravalent antibodies induce a split between two distinct endocytic pathways:

    • Clathrin-mediated endocytosis (CME)

    • Clathrin-independent endocytosis (CIE) that requires dynamin-2

This switch in endocytic mechanism correlates with significantly improved efficiency of FGFR1 internalization and receptor degradation. The formation of higher-order receptor clusters by tetravalent antibodies likely creates membrane curvature patterns or receptor densities that engage additional endocytic machinery beyond the clathrin pathway. This phenomenon has important implications for developing antibody-based therapeutics with enhanced internalization properties for targeted drug delivery applications .

How do FGFR1 antibodies impact interactions with co-receptors and signaling modulators?

FGFR1 antibodies can significantly impact interactions with co-receptors even when they don't directly interfere with ligand binding. Studies using bio-layer interferometry (BLI) with the Octet RED K2 system demonstrate that antibody fragments binding to the D1 domain of FGFR1 inhibit interaction with the co-receptor β-Klotho . The experimental approach involved:

  • Chemical immobilization of FGFR1 D1-D3-Fc (10 μg/ml) on AR2G biosensors

  • Analysis of FGFR1 interaction with β-Klotho (11 μg/ml)

  • Pre-incubation of immobilized FGFR1 with excess scFv proteins (30 μg/ml)

  • Measurement of β-Klotho binding in the presence of scFv proteins

This finding suggests that even though D1-binding antibodies don't interfere with FGF1 binding, they can modulate FGFR1 function by preventing formation of receptor complexes with co-receptors that regulate specific signaling pathways. This mechanism provides an additional layer of control for researchers developing therapeutic antibodies that can selectively inhibit specific FGFR1-dependent signaling pathways without globally blocking all receptor functions .

What controls are essential when evaluating FGFR1 antibody effects on receptor activation and signaling?

When evaluating FGFR1 antibody effects on receptor activation and signaling, several essential controls must be included:

  • Positive activation control: Include FGF1 (typically 20 ng/ml with 20 U/ml heparin) treatment for 15 minutes to establish baseline receptor activation. This serves as a positive control for phosphorylation of FGFR1 and downstream effectors like ERK1/2 .

  • Dose-response analysis: Test increasing concentrations of antibodies to detect potential dose-dependent effects on receptor activation.

  • Antibody format controls: Compare monovalent (scFv) versus bivalent (scFv-Fc) formats of the same antibody to distinguish valency-dependent effects from epitope-specific effects .

  • Time-course analysis: Monitor signaling events at multiple time points (e.g., 15 min, 30 min, 1 hr, 2 hr) to distinguish between transient and sustained signaling effects.

  • Pathway-specific readouts: Beyond FGFR1 autophosphorylation, measure multiple downstream signaling molecules (e.g., ERK1/2, AKT, PLCγ) to characterize pathway-specific effects.

  • Competition experiments: Assess whether antibodies compete with FGF1 by pre-incubating cells with antibodies before FGF1 stimulation, measuring signaling outputs by Western blotting .

  • Protein synthesis inhibition: Include cycloheximide treatment when studying receptor degradation to prevent confounding effects from newly synthesized receptor .

How can researchers address variability in FGFR1 antibody performance across different cell lines?

Addressing variability in FGFR1 antibody performance across cell lines requires systematic characterization and optimization:

  • Receptor expression profiling: Quantify FGFR1 expression levels across cell lines using Western blot and flow cytometry. Compare both mRNA transcript levels and protein levels, as demonstrated in studies showing differential FGFR expression between normal and cancer cells .

  • Isoform characterization: Determine which FGFR1 isoforms (α versus β; IIIb versus IIIc) are expressed in each cell line, as antibodies may have isoform-specific binding properties .

  • Co-receptor expression: Analyze expression of FGFR1 co-receptors (e.g., β-Klotho, heparan sulfate proteoglycans) that may influence antibody binding or effects .

  • Pilot experiments with model cell lines: Establish baseline conditions using cell lines with defined FGFR1 properties. For example, studies have used both engineered lines with FGFR1 overexpression (U2OSR1) and lines with endogenous expression (NIH3T3) to validate antibody effects .

  • Cell type-specific optimization: Adjust antibody concentrations and experimental conditions for each cell line. For internalization studies, compare endocytic pathway components across cell types.

  • Standardized quantification: Use normalized quantification methods (e.g., setting FGF1 response to 100%) to make comparable measurements across cell lines .

What approaches can resolve contradictory results between functional and binding assays with FGFR1 antibodies?

When faced with contradictory results between functional and binding assays using FGFR1 antibodies, researchers should consider the following approaches:

  • Epitope characterization: Thoroughly map antibody binding sites using truncated receptor constructs (D1-D2-D3, D2-D3, isolated D1) . Antibodies binding to different domains may show strong binding signals but divergent functional effects.

  • Valency assessment: Compare monovalent versus bivalent formats of the same antibody. Research demonstrates that while both formats may show similar binding, they can have dramatically different effects on receptor internalization and degradation .

  • Temporal analysis: Binding assays (e.g., SPR) typically measure initial interaction, while functional assays assess downstream effects that occur over longer timeframes. Perform time-course experiments for both binding and functional readouts.

  • Multiparameter analysis: Combine multiple assay types - for example, use chemical crosslinking to detect ternary complexes containing FGF1, FGFR1, and antibody fragments to reconcile seemingly contradictory binding versus functional data .

  • Proximity-based assays: Implement methods like Duolink In Situ Assay to directly visualize molecular interactions in cellular contexts , which may reveal spatial relationships not apparent in biochemical assays.

  • Conformational considerations: Assess whether the antibody recognizes native receptor conformations. Comparing binding to cell-surface receptors versus recombinant proteins can identify conformation-dependent effects.

What methodological approaches best characterize the kinetics of antibody-FGFR1 interactions?

Characterizing the kinetics of antibody-FGFR1 interactions requires sophisticated biophysical techniques with careful experimental design:

  • Surface Plasmon Resonance (SPR):

    • Immobilize FGFR1-Fc at controlled density (e.g., 1000 RU) on CM4 sensors

    • Apply various concentrations of antibodies (typically 0.625-20 nM range)

    • Measure association for 120 seconds and dissociation for 180 seconds

    • Use a controlled flow rate (30 μL/min) in standardized buffer (PBS with 0.05% Tween 20, 0.02% NaN3, pH 7.2)

    • Regenerate chip surface with 10 mM glycine, pH 1.5 between measurements

    • Analyze data with specialized software to determine kinetic constants (kon, koff) and equilibrium dissociation constant (KD)

  • Bio-layer Interferometry (BLI):

    • Chemically immobilize FGFR1 on AR2G biosensors

    • Measure real-time binding kinetics of antibodies at various concentrations

    • Analyze association and dissociation phases to determine binding parameters

  • Isothermal Titration Calorimetry (ITC):

    • Measure thermodynamic parameters (ΔH, ΔS) along with binding affinity

    • Provides stoichiometry information critical for understanding complex binding modes

  • Comparative analysis across formats:

    • Compare binding parameters between scFv, scFv-Fc (bivalent), and more complex antibody formats

    • Correlate kinetic parameters with functional outcomes in cellular assays

    • Extract avidity effects by comparing monovalent versus multivalent formats

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 2024 Thebiotek. All Rights Reserved.