Recombinant Human Fibroblast growth factor receptor 3 (FGFR3), partial (Active)

Shipped with Ice Packs
In Stock

Description

Functional Role in Signaling Pathways

FGFR3 regulates critical cellular processes through ligand-dependent and -independent activation:

Key Signaling Pathways:

  • MAPK/ERK: Phosphorylates FRS2, recruiting GRB2/SOS1 to activate RAS-MAPK cascades .

  • PI3K-AKT: Interacts with p85 regulatory subunits of PI3K, modulating cell survival and proliferation .

  • PLCγ: Generates secondary messengers (e.g., inositol trisphosphate) via phospholipase C activation .

Disease Associations:

  • Cancer: Constitutive activation (e.g., K650E mutation) drives multiple myeloma and bladder cancer .

  • Skeletal Disorders: Gain-of-function mutations cause achondroplasia and thanatophoric dysplasia .

3.1. Inhibitor Screening

  • Peptide P3: Suppresses FGFR3 kinase activity (IC₅₀ ~10 µM) and blocks ERK phosphorylation in chondrocytes .

  • Pheophorbide A: Reduces FGFR3 half-life and downstream signaling in myeloma cells, validated in FGFR3 ACH mouse models .

3.2. Mechanistic Insights

Study FocusKey FindingSource
Autophagy RegulationFGFR3 activation decreases ATG12–ATG5 conjugate levels, inhibiting autophagy in chondrocytes .
RSK2 InteractionFGFR3 phosphorylates RSK2 at Ser386, promoting hematopoietic transformation .
PI3K CrosstalkDirect interaction with p85α at Y760 modulates ERK signaling in myeloma .

Production and Validation

Expression Systems:

  • HEK293: Preferred for glycosylation and proper folding .

  • E. coli: Used for non-glycosylated variants with His tags .

Activity Assays:

  • Kinase Activity: Measured via phosphorylation of PLCG1 or FRS2 .

  • Functional Validation: Inhibition of FGF1-dependent proliferation in Balb/c-3T3 cells .

Limitations and Future Directions

  • Glycosylation Variability: HEK293-derived proteins show batch-dependent glycosylation patterns affecting ligand affinity .

  • Therapeutic Potential: Small-molecule inhibitors (e.g., pheophorbide A) require further pharmacokinetic optimization .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 1xPBS, pH 7.4.
Form
Lyophilized powder
Lead Time
Typically, we can ship the products within 5-10 business days of receiving your order. The delivery time may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Notes
Repeated freezing and thawing is not recommended. For short-term storage, working aliquots can be stored at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial prior to opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting the solution at -20°C/-80°C. Our default final concentration of glycerol is 50%, which can be used as a reference.
Shelf Life
The shelf life is influenced by factors such as storage conditions, buffer composition, storage temperature, and the intrinsic stability of the protein. Generally, the shelf life of liquid forms is 6 months at -20°C/-80°C. For lyophilized forms, the shelf life is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
C-terminal hFc-tagged
Synonyms
ACH; CD 333; CD333; CD333 antigen; CEK 2; CEK2; FGFR 3; FGFR-3; FGFR3; FGFR3_HUMAN; Fibroblast growth factor receptor 3 (achondroplasia thanatophoric dwarfism); Fibroblast growth factor receptor 3; Heparin binding growth factor receptor; HSFGFR3EX; Hydroxyaryl protein kinase; JTK 4; JTK4; MFR 3; SAM 3; Tyrosine kinase JTK 4; Tyrosine kinase JTK4; Z FGFR 3
Datasheet & Coa
Please contact us to get it.
Expression Region
23-375aa
Mol. Weight
64.8 kDa
Protein Length
Partial
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Signal Transduction
Source
Mammalian cell
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Fibroblast growth factor receptor 3 (FGFR3) is a tyrosine-protein kinase that acts as a cell-surface receptor for fibroblast growth factors. It plays a critical role in regulating cell proliferation, differentiation, and apoptosis. FGFR3 is essential for normal skeletal development, particularly in regulating chondrocyte differentiation, proliferation, and apoptosis. It also plays a role in both osteogenesis and postnatal bone mineralization by osteoblasts. While FGFR3 can promote apoptosis in chondrocytes, it can also contribute to cancer cell proliferation. FGFR3 is crucial for the normal development of the inner ear. It phosphorylates PLCG1, CBL, and FRS2, activating various signaling cascades. Ligand binding triggers activation of these cascades, including the production of diacylglycerol and inositol 1,4,5-trisphosphate via PLCG1 activation. Phosphorylation of FRS2 leads to the recruitment of GRB2, GAB1, PIK3R1, and SOS1, mediating the activation of RAS, MAPK1/ERK2, MAPK3/ERK1, and the MAP kinase signaling pathway, as well as the AKT1 signaling pathway. FGFR3 is involved in the regulation of vitamin D metabolism. Mutations that lead to constitutive kinase activation or disrupt normal FGFR3 maturation, internalization, and degradation result in aberrant signaling. Overexpression or constitutive activation of FGFR3 promotes the activation of PTPN11/SHP2, STAT1, STAT5A, and STAT5B. The secreted isoform 3 retains its capacity to bind FGF1 and FGF2, potentially interfering with FGF signaling.
Gene References Into Functions
  1. Authors found that FGFR3-AS1 silencing decreased the activation of the PI3K/AKT signaling pathway. PMID: 29463348
  2. Our research identified FGFR3(high)/Ki67(high) papillary pTa tumors as a subgroup with poor prognosis, emphasizing the significance of histological grading for high-grade tumors. PMID: 30154342
  3. Patients with FGFR3 mutations or FGFR3-TACC3 fusion may be potential candidates for novel FGFR-targeted therapies in the perioperative setting. PMID: 30064409
  4. Data suggest that FGFR3 mutations found in SADDAN (but not those found in TDII) affect cytoskeleton organization in chondrocytes by inducing tyrosine hyperphosphorylation of paxillin; binding of FGFR3 to PLCG1 appears to be involved. (PLCG1 = phospholipase C gamma 1; SADDAN = Severe Achondroplasia with Developmental Delay and Acanthosis Nigricans; TDII = Thanatophoric Dysplasia type II) PMID: 29242050
  5. This case presents prenatal ultrasonographic findings suggestive of TD and highlights the patient's postnatal dysmorphic features and typical radiographic findings. The definitive diagnosis of TD type I (TDI) was made postnatally, with molecular genetic analysis revealing the previously described p.R248C mutation in FGFR3. This case is reported due to its relatively long lifespan and the molecular diagnosis. PMID: 30226972
  6. FGFR3 expression indicated an adverse prognosis for individuals with lung adenocarcinoma and promoted metastatic potential of lung adenocarcinoma cells. PMID: 29850625
  7. FGFR1, along with its downstream regulatory PI3K/AKT kinases, may serve as potential biomarkers for the invasiveness and prognosis of laryngeal cancer. PMID: 29299828
  8. FGFR3-AS1 expression levels were higher in high-grade tumors than those in low-grade tumors and in invasive bladder tumors compared to non-invasive bladder tumors. PMID: 29226855
  9. Disease-free survival (DFS) was calculated based on FGFR3 IHC expression. PMID: 30061236
  10. The FGFR3 gene is responsible for the production of the FGFR 3 protein, which converts cartilage to bone. All individuals with a single copy of the mutated FGFR3 gene have Achondroplasia. PMID: 29185944
  11. Genetic association studies in a pediatric population in Japan suggest that mutations in ACAN (aggrecan), FGFR3 (fibroblast growth factor receptor-3), or GHRHR (growth-hormone-releasing-hormone receptor) are associated with idiopathic short stature in the population studied. PMID: 28768959
  12. HPV-positive vulvar squamous cell carcinoma is characterized by oncogenic FGFR3 mutations, classifying this subtype as a distinct disease. Inhibitors of FGFR3 are a potential therapeutic strategy for this neglected cancer in women. PMID: 28377483
  13. Results show that olfactory neuroblastoma tumors exhibit recurrent chromosomal copy-number changes, including FGFR3 amplification associated with overexpression. PMID: 28775129
  14. FGFR3-TACC3 is a recurrent resistance mechanism that can bypass EGFR blockade by all generations of EGFR TKIs in NSCLC. PMID: 28838400
  15. Mutation in the FGFR3 gene is associated with Klinefelter syndrome and achondroplasia. PMID: 28672740
  16. Genetic screening of the family revealed a previously reported heterozygous c.1138 G > A (p.G380R) mutation in the coding exon 8 of the FGFR3 gene. PMID: 28679403
  17. FGFR3 mutations have very limited urothelial tumorigenicity, and these mutations must collaborate with other genetic events to drive urothelial tumorigenesis. PMID: 27157475
  18. Long-term dovitinib administration was not feasible due to frequent toxicity. The absence of clinical activity suggests that patient selection based solely on pFGFR3 IHC does not enrich for response to FGFR3 kinase inhibitors in urothelial carcinoma. PMID: 27932416
  19. Results provide evidence that FGFR3 mutations in human papillomavirus-positive tonsillar and base of tongue cancer are indicative of a worse prognosis. PMID: 28525363
  20. Increased levels of FGFR3 and PIK3CA mutated DNA in urine and plasma are indicative of later progression and metastasis in bladder cancer. PMID: 28069289
  21. FGFR3 expression was increased in classical and neural subtypes of glioma and was associated with differentiated cellular functions. FGFR3 showed very limited correlation with other common receptor tyrosine kinases and predicted improved survival for glioma patients. PMID: 27829236
  22. REVIEW. FGFR3 is expressed in chondrocytes and mature osteoblasts where it functions to regulate bone growth. Analysis of the mutations in FGFR3 revealed increased signaling through a combination of mechanisms. PMID: 27987249
  23. There was a lower frequency of mutation in FGFR3, a gene associated with low-risk Bladder Cancer, than reported in The Cancer Genome Atlas database. PMID: 27520487
  24. Higher expression of FGFR3, phosphorylated AKT, and ZEB1 were observed. PMID: 27267856
  25. FGFR3 mutation status seems promising to guide decision-making on adjuvant anti-FGFR3 therapy, as it appeared homogeneous in RC and lymph nodes +. Based on the results of TUR, the deep part of the tumor needs to be assessed if neoadjuvant anti-FGFR3 treatment is considered. PMID: 27091807
  26. Our study identified the first actionable mutation spectrum in the Indian lung cancer genome. These findings implicate FGFR3 as a novel therapeutic target in lung adenocarcinoma. PMID: 27998968
  27. FGFR2, TWIST1, and FGFR3 mutations were identified in children with tracheal cartilaginous sleeve (TCS). All five children with a W290C mutation in FGFR2 had TCS, and most previously reported children with W290C had identification of TCS or early death. PMID: 27228464
  28. The Gly380Arg and Asn540Lys are hotspot mutations of the FGFR3 gene among patients with ACH/HCH. PMID: 28777845
  29. The FGFR3 gene is an infrequent target in the pathogenesis of Han Chinese urothelial cell carcinoma. PMID: 27029078
  30. Our research extends the genetic mutation spectrum of FGFR3. PMID: 29080836
  31. A study found FGFR3 gene mutation plus GRB10 gene duplication in a patient with achondroplasia plus growth delay with prenatal onset. PMID: 27370225
  32. Our family study confirms the consistent and unique phenotype of this condition and the specificity of the mutation in FGFR3. PMID: 27139183
  33. No insertions or deletions in FGFR3 have been reported to cause thanatophoric dysplasia types 1 or 2; therefore, this represents the first report describing such a mutation. PMID: 27028100
  34. Results suggest that FGFR3 kinase activity may regulate the papillomavirus reproductive program through phosphorylation of the E2 protein, although this is unlikely to occur through the Y102 residue of HPV E2. PMID: 28768864
  35. Our data reinforce the notion that molecular testing of FGFR3 must be included in the diagnostic approach of coronal craniosynostosis. This will allow accurate genetic counseling and optimal management of MS, which might otherwise go undiagnosed due to mild manifestations and wide variability of expression. PMID: 27568649
  36. We describe the first case of protein-losing enteropathy in a pediatric patient with severe skeletal dysplasia consistent with thanatophoric dysplasia type I and DNA analysis that revealed a c.1949A>T (p.Lys650Met) in exon 15 of the FGFR3 gene. PMID: 27214123
  37. High FGFR3 expression is associated with bladder cancer. PMID: 28320388
  38. The study provides evidence of the significant oncogenic potential of the FGFR3-TACC3 fusion protein. The presence of the TACC coiled-coil domain leads to increased and altered levels of FGFR3 activation, fusion protein phosphorylation, downstream signaling, cellular transformation, proliferation, and viability. PMID: 26869289
  39. Our research shows that low doses of NVP-BGJ398 improve in vivo condyle growth and correct dysmorphologies in Fgfr3(Y367C/+) mice, suggesting that postnatal treatment with NVP-BGJ398 mice might offer a new therapeutic strategy to improve mandible anomalies in achondroplasia (ACH) and other FGFR3-related disorders. PMID: 27260401
  40. FGFR3 mRNA missplicing in hepatocellular carcinoma (HCC) contributes significantly to its malignant character. PMID: 27267910
  41. Mutations in the FGFR3 gene are associated with tubular adenomas. PMID: 27438523
  42. High FGFR3 expression is associated with multiple myeloma. PMID: 27509849
  43. FGFR3 was predominantly mutated in infiltrative hepatocellular carcinoma (HCC) compared to nodular HCC. FGFR3 protein expression was higher in mutated infiltrative HCC compared to non-mutated infiltrative HCC and nodular HCC. FGFR3 may be a candidate oncogene in tumor progression. PMID: 28058595
  44. Our findings show that grade heterogeneity in urothelial carcinoma is characterized by increased MIB-1 labeling, particularly in the FGFR3 mutant pathway, with homozygous deletions of CDKN2A in low- and high-grade areas. PMID: 27530957
  45. We argue that routine use of molecular genomic tumor analysis in urothelial carcinoma may inform the selection of patients for appropriate trials, and we further investigate the potential of FGFR3 as a meaningful clinical target for this challenging disease. PMID: 27271022
  46. In conclusion, we identified four novel loci (TGFA, PIK3R1, FGFR3, and TREH) and confirmed two loci known to be associated with cartilage thickness. The identified associations were not caused by rare exonic variants. This is the first report linking TGFA to human Osteoarthritis, which may serve as a new target for future therapies. PMID: 27701424
  47. Case Report: FGFR3 epidermal naevus syndrome with urothelial mosaicism for activating p.Ser249Cys FGFR3 mutation. PMID: 27786351
  48. FGFR alterations are not frequent in low-grade gliomas, they are more common in hemispheric low-grade gliomas and are important since targeted therapies exist for FGFR receptors. PMID: 27659822
  49. FGFR3 gene mutations are associated with Urinary Bladder Cancer. PMID: 27356691
  50. We identified a novel FGFR3 mutation, p.Ser348Cys, in a patient with achondroplasia. A number of different FGFR3 mutations can cause achondroplasia; therefore, if the common p.Gly380Arg mutation is not found, complete analysis of FGFR3 is indicated in patients with achondroplasia. PMID: 26754866

Show More

Hide All

Database Links

HGNC: 3690

OMIM: 100800

KEGG: hsa:2261

STRING: 9606.ENSP00000339824

UniGene: Hs.1420

Involvement In Disease
Achondroplasia (ACH); Crouzon syndrome with acanthosis nigricans (CAN); Thanatophoric dysplasia 1 (TD1); Thanatophoric dysplasia 2 (TD2); Hypochondroplasia (HCH); Bladder cancer (BLC); Cervical cancer (CERCA); Camptodactyly, tall stature, and hearing loss syndrome (CATSHLS); Multiple myeloma (MM); Lacrimo-auriculo-dento-digital syndrome (LADDS); Keratinocytic non-epidermolytic nevus (KNEN); Muenke syndrome (MNKS); Keratosis, seborrheic (KERSEB); Testicular germ cell tumor (TGCT); Achondroplasia, severe, with developmental delay and acanthosis nigricans (SADDAN)
Protein Families
Protein kinase superfamily, Tyr protein kinase family, Fibroblast growth factor receptor subfamily
Subcellular Location
[Isoform 1]: Cell membrane; Single-pass type I membrane protein. Cytoplasmic vesicle. Endoplasmic reticulum. Note=The activated receptor is rapidly internalized and degraded. Detected in intracellular vesicles after internalization of the autophosphorylated receptor.; [Isoform 2]: Cell membrane; Single-pass type I membrane protein.; [Isoform 3]: Secreted.; [Isoform 4]: Cell membrane; Single-pass type I membrane protein.
Tissue Specificity
Expressed in brain, kidney and testis. Very low or no expression in spleen, heart, and muscle. In 20- to 22-week old fetuses it is expressed at high level in kidney, lung, small intestine and brain, and to a lower degree in spleen, liver, and muscle. Isof

Q&A

What is the structure of FGFR3 and how does it function in normal cells?

FGFR3 belongs to the fibroblast growth factor receptor family, which are type I transmembrane receptor tyrosine kinases with immunoglobulin-like domains. The receptor consists of an extracellular domain containing three Ig-like loops, a transmembrane domain, and an intracellular tyrosine kinase domain. In its normal state, FGFR3 binds to various FGF ligands, particularly FGF1, FGF2, FGF8, and FGF9 in the case of the FGFR3c splice variant . This binding triggers receptor dimerization, autophosphorylation, and activation of downstream signaling cascades.

The activation of FGFR3 leads to multiple signaling outputs primarily through the MAPK pathway and PLCγ signaling, with additional pathways including STAT activation and RSK2 . The cellular outcomes of FGFR3 signaling are highly context-dependent and can lead to proliferation, migration, or differentiation, depending on the cell type and specific conditions. In normal urothelial cells, both wildtype FGFR3 and its splice variants are expressed, with expression levels changing during differentiation and confluence .

How do FGFR3 isoforms differ and what are their functional implications?

FGFR3 exists in multiple isoforms resulting from alternative splicing. The two major splice variants are FGFR3b and FGFR3c, which differ in their third Ig-like domain, resulting in different ligand binding specificities. The FGFR3c variant is primarily activated by FGF1, FGF2, FGF8, and FGF9 .

Additionally, a truncated splice variant (Δ8-10) has been identified in normal urothelial cells. This variant lacks the region encoding the second part of the third Ig-like loop and the transmembrane domain. This isoform is glycosylated and secreted, capable of binding FGF1 and dimerizing. Interestingly, it can block the response to FGF1 in cells expressing full-length FGFR3, suggesting a negative regulatory role in normal urothelium . This variant is expressed at lower levels in tumor cell lines, potentially contributing to the dysregulation of FGFR3 signaling in cancer.

What experimental systems are available for studying recombinant FGFR3 function?

Several experimental approaches have been developed to study FGFR3 function:

  • Micropatterned surfaces for live cell analysis: This technique allows for quantification of GRB2 recruitment to mature receptors at the plasma membrane, providing insights into signaling events specifically at the cell surface .

  • Western blotting: This traditional method quantifies the activation of FGFRs by determining the phosphorylation state of tyrosines in adaptor protein docking sites, comparing the signal of the phosphorylated protein to the signal intensity of the pan-protein .

  • Cell-based functional assays: These assays measure outcomes such as proliferation, migration, and differentiation in response to FGFR3 activation or inhibition.

  • Molecular dynamics simulations: Computational approaches using software such as GROMACS to study FGFR3 structure, conformational changes, and interactions with ligands or inhibitors .

How can researchers accurately quantify FGFR3 signaling activity in different cellular compartments?

Quantifying FGFR3 signaling in different cellular compartments requires specialized approaches beyond traditional Western blotting, which typically measures bulk cellular responses. A micropatterning method has been developed to specifically report on signaling events at the plasma membrane, providing a more nuanced understanding of receptor activation .

This approach involves:

  • Creating micropatterned surfaces that allow for precise spatial organization of cells

  • Live cell imaging to monitor GRB2 recruitment to FGFR3 at the plasma membrane

  • Quantitative analysis of these recruitment events as a measure of receptor activation

This method has revealed that FGFR3 activation at the cell surface can differ significantly from measurements in bulk cell extracts. For instance, some FGFR3 mutants (K650Q and K650E) demonstrated either unexpectedly high or low activation states compared to previous reports, likely because the micropatterning method specifically probes the receptor population at the plasma membrane rather than the entire cellular pool .

What are the molecular mechanisms underlying ligand-independent activation of FGFR3 mutants?

FGFR3 mutants often display increased basal (ligand-independent) receptor activity, which contributes to their pathological effects. Studies have examined various FGFR3 mutants to understand this phenomenon:

The mechanisms of ligand-independent activation may include:

  • Altered receptor conformation that mimics the ligand-bound state

  • Enhanced dimerization propensity in the absence of ligand

  • Changes in interactions with regulatory proteins

  • Altered subcellular localization affecting access to downstream signaling components

Understanding these mechanisms is crucial for developing targeted therapeutic approaches for conditions associated with FGFR3 mutations.

How do computational methods contribute to FGFR3 inhibitor discovery and optimization?

Advanced computational methods have become essential tools in the discovery and optimization of FGFR3 inhibitors:

  • Structure relaxation through molecular dynamics (MD) simulations: Using the FGFR3 crystal structure (e.g., PDB code: 6LVM) as the starting point, MD simulations with packages like GROMACS can explore the receptor's conformational space and identify stable states for virtual screening. These simulations typically employ force fields such as AMBER 99SB-ILDN with explicit solvation .

  • Free energy landscape analysis: By analyzing Gibbs free energy during simulations, researchers can identify energy basins and transition states, revealing the lowest energy conformations of FGFR3 after releasing bound inhibitors. This approach was used to determine that a 500 ns simulation successfully sampled FGFR3 after releasing Pyrimidine Derivative 37b .

  • Active learning-based virtual screening: This approach combines physics-based methods with machine learning to efficiently screen large compound libraries. The process involves:

    • Generating a receptor grid from a prepared protein

    • Preparing compound libraries (e.g., natural compound libraries)

    • Initial docking using Glide SP

    • Training machine learning models on physics-based data

    • Iterative improvement through multiple training rounds

These computational approaches significantly enhance the efficiency of drug discovery efforts by prioritizing compounds with favorable binding profiles for experimental validation.

What are the optimal experimental conditions for analyzing FGFR3 activation in response to different ligands?

Analyzing FGFR3 activation requires careful consideration of experimental conditions:

  • Ligand selection: FGFR3c is primarily activated by FGF1, FGF2, FGF8, and FGF9, with FGF1 and FGF2 showing differences in inducing kinase phosphorylation. FGF2 has been reported to have a stronger activating effect when added at saturating conditions .

  • Heparin co-factor: Heparin or heparan sulfate proteoglycans are typically required as co-factors for optimal FGF binding and receptor activation. For example, the effective concentration (ED50) for FGF3 effect is 0.02-0.1 μg/mL in the presence of 1 μg/mL of heparin .

  • Receptor isoform consideration: Experiments should specify which FGFR3 isoform is being studied (e.g., FGFR3b or FGFR3c) as they have different ligand binding profiles.

  • Cell context: The outcome of FGFR3 signaling depends heavily on cell context, so the choice of cell system is crucial. Responses in NIH-3T3 cells may differ significantly from those in urothelial cells or other relevant cell types .

  • Readout selection: Different readouts (phosphorylation of specific sites, GRB2 recruitment, downstream pathway activation) may give varying results and should be carefully chosen based on the specific research question.

How can researchers effectively study the interplay between FGFR3 signaling and immune checkpoint pathways?

Studying the interplay between FGFR3 signaling and immune checkpoint pathways requires integrated experimental and computational approaches:

  • Mathematical modeling: Developing validated mathematical models of FGFR3-mediated tumor growth can help investigate the impact of combined therapies. Such models can be calibrated with experimental data before exploring survival benefits and optimal dosing schedules .

  • Combined therapy experiments: Testing the effects of FGFR3 inhibitors in combination with immune checkpoint inhibitors (e.g., anti-PD-L1 therapy) under various conditions and sequences.

  • Parameter space exploration: Identifying regions where each monotherapy can outperform the other, and determining optimal combination strategies .

Recent mathematical models have suggested that:

  • FGFR3 mutation reduces the effectiveness of anti-PD-L1 therapy

  • There are specific parameter regions where either anti-FGFR3 or anti-PD-L1 monotherapy can outperform the other

  • Pretreatment with anti-PD-L1 therapy consistently results in greater tumor reduction, even when anti-FGFR3 therapy is the more effective monotherapy

These findings provide a rational framework for designing experimental studies and clinical trials of combination therapies targeting both FGFR3 and immune checkpoints.

What is the role of FGFR3 mutations in bladder cancer pathogenesis and progression?

FGFR3 plays a central role in bladder cancer pathogenesis:

  • Prevalence of alterations: Over 80% of non-muscle-invasive bladder cancers (NMIBC) and approximately 40% of muscle-invasive bladder cancers (MIBC) have upregulated FGFR3 signaling. These frequencies may be even higher when considering alternative splicing, ligand expression, and regulatory mechanism changes .

  • Types of alterations: FGFR3 alterations in bladder cancer include point mutations, overexpression of wildtype receptor, and FGFR3 gene fusions.

  • Prognostic significance: FGFR3 mutation generally identifies patients with favorable NMIBC disease, though only grade and stage remain single independent predictors of outcome in multivariate analyses .

  • Therapeutic implications: FGFR3 mutations rather than upregulated expression may represent better predictive biomarkers for FGFR inhibitor therapies. The FGFR inhibitor Erdafitinib has shown a 40% response rate in patients with FGFR3 point mutations or FGFR2/3 fusions, leading to FDA approval for locally advanced or metastatic bladder cancer .

  • Resistance mechanisms: Despite encouraging response rates, approximately 60% of patients with FGFR3 alterations do not respond to FGFR inhibitors, suggesting the presence of escape mechanisms, development of stable resistance, loss of dependence on FGFR3 during tumor progression, or intratumor heterogeneity .

How can researchers distinguish between oncogenic and non-oncogenic FGFR3 mutations in experimental systems?

Distinguishing between oncogenic and non-oncogenic FGFR3 mutations requires multiple complementary approaches:

  • Functional assays: Measuring basal and ligand-stimulated receptor activity using methods such as GRB2 recruitment in micropatterned cells. Oncogenic mutations typically show increased basal activity, though the response to ligand stimulation varies among mutants .

  • Signaling pathway analysis: Examining downstream signaling pathway activation (MAPK, PLCγ, STAT) in response to different mutations. Oncogenic mutations often show altered patterns of pathway activation compared to wildtype FGFR3 .

  • Cell transformation assays: Assessing the ability of mutant FGFR3 to transform NIH-3T3 or other relevant cell types. Oncogenic mutations generally promote anchorage-independent growth and other hallmarks of cellular transformation .

  • In vivo models: Evaluating the tumorigenic potential of FGFR3 mutations in animal models, particularly in tissue-specific contexts relevant to human cancers.

  • Computational prediction: Using in silico analysis methods such as CADD or SIFT scores, or merging information from multiple component methods with experimental data to predict the functional impact of mutations .

The integration of these approaches provides a more comprehensive understanding of the oncogenic potential of specific FGFR3 mutations, helping to prioritize therapeutic targets and stratify patients for clinical trials.

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.