Anti-Human Fibroblast Growth Factor 23 (Burosumab)

Anti-Human Fibroblast Growth Factor 23 (Burosumab)

Product No.: F540

- -
- -
Product No.F540
Clone
KRN-23
Target
FGF23
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Fibroblast growth factor 23, Phosphatonin, Tumor-derived hypophosphatemia-inducing factor, FGF23
Isotype
Human IgG1κ
Applications
ELISA
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Active
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
≤ 1.0 EU/mg as determined by the LAL method
Purity
≥95% by SDS Page
≥95% monomer by analytical SEC
Formulation
This biosimilar antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
State of Matter
Liquid
Product Preparation
Recombinant biosimilar antibodies are manufactured in an animal free facility using only in vitro protein free cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Pathogen Testing
To protect mouse colonies from infection by pathogens and to assure that experimental preclinical data is not affected by such pathogens, all of Leinco’s recombinant biosimilar antibodies are tested and guaranteed to be negative for all pathogens in the IDEXX IMPACT I Mouse Profile.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Regulatory Status
Research Use Only
Country of Origin
USA
Shipping
2 – 8° C Wet Ice
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.

Description

Description

Specificity
This non-therapeutic biosimilar antibody uses the same variable region sequence as the therapeutic antibody Burosumab. Burosumab (KRN-23) is a human monoclonal antibody that specifically targets fibroblast growth factor 23 (FGF23).
Background
Fibroblast growth factor 23 (FGF23) is a hormone produced by bone cells that plays a key role in regulating phosphate levels in the body. Excessive FGF23 production leads to hypophosphatemic conditions like rickets and osteomalacia. Disorders such as X-linked hypophosphatemia (XLH) and tumor-induced osteomalacia (TIO) are characterized by elevated FGF23 levels, resulting in low phosphate levels and impaired bone health. Measuring FGF23 levels is critical for diagnosing these hypophosphatemic diseases and guiding treatment decisions1-3.

Burosumab (KRN23) is a fully human monoclonal antibody that binds to and neutralizes excess FGF23, helping restore phosphate balance. Clinical trials have shown burosumab to be effective in treating both XLH and TIO in children and adults, improving serum phosphate levels, bone turnover, fracture healing, and reducing pain. Approved for use in these conditions, burosumab offers a targeted therapy that addresses the underlying cause of FGF23-related disorders. Ongoing studies continue to explore its long-term safety and efficacy, highlighting its potential for sustained clinical benefits2,4,5.
Antigen Distribution
FGF23 is primarily produced by osteocytes and osteoblasts in the bone. It acts mainly on the kidneys to regulate phosphate reabsorption and vitamin D metabolism. FGF23 can also be found in other tissues, such as the liver, heart, and brain, but its highest expression is in the bone.
Ligand/Receptor
FGFR1, FGFR2, FGFR3, FGFR4
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Oncology
.
Bone Disease
.
Osteomalacia

Leinco Antibody Advisor

Powered by AI: AI is experimental and still learning how to provide the best assistance. It may occasionally generate incorrect or incomplete responses. Please do not rely solely on its recommendations when making purchasing decisions or designing experiments.

Research-grade Burosumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as the analytical standard to generate a reference concentration curve, enabling quantification of Burosumab levels in test serum samples. This approach ensures assay consistency and comparability for both biosimilar and reference products.

Key details on their use include:

  • Analytical Standard/Calibration Curve: The biosimilar, characterized for purity and concentration, is serially diluted to prepare calibration standards over a range of concentrations (e.g., 50–12,800 ng/mL) in a matrix matched to study samples, such as human serum. These standards are included in each assay run to generate a standard curve, against which unknown serum sample concentrations are interpolated.

  • Reference Controls: QC samples can be prepared from both the biosimilar and the reference (originator) Burosumab to test assay performance, establishing that the ELISA quantifies both products equivalently when employing the biosimilar as the calibration standard.

  • Assay Bridging Rationale: Regulatory guidance and bioanalytical best practice recommend the use of a single, well-characterized standard (often the biosimilar) to minimize variability and facilitate direct comparison between biosimilar and reference product PK profiles in the assay. Equivalence in quantitation is established statistically during assay validation and qualification.

  • In the ELISA process:

    • Capture antibodies specific to Burosumab are pre-coated onto ELISA plates.
    • Serum samples, calibrator (biosimilar) standards, and QC reference controls are added.
    • After incubation and washing, a detection antibody (often enzyme-linked) is added, followed by substrate for signal development.
    • The developed signal corresponds to Burosumab concentration, interpolated from the standard curve based on the biosimilar standard.

In summary, research-grade Burosumab biosimilars function as the quantitation anchor for PK ELISAs—serving as the standard against which both biosimilar and reference product concentrations are measured in serum, and as reference controls to ensure assay comparability and accuracy. This ensures robust bioanalytical comparability essential for PK bridging studies in biosimilar development.

The primary preclinical models used to administer research-grade anti-FGF23 antibodies in vivo for studying tumor growth inhibition and characterizing tumor-infiltrating lymphocytes (TILs) are mouse syngeneic tumor models. There is no clear evidence in the current literature that humanized models have been routinely used for anti-FGF23 antibody studies focused on TIL characterization.

Essential Context and Supporting Details:

  • Syngeneic models involve implantation of mouse-derived tumor cell lines into immunocompetent mice of the same genetic background, allowing the study of tumor-immune interactions, TIL profiles, and therapeutic antibody effects in the context of a fully functional murine immune system.

  • These models are specifically favored for immunotherapy research, including studies of immune checkpoint inhibitors and profiling of TILs by flow cytometry, due to the presence of mouse-origin immune cells that accurately reflect the in vivo immune response to treatments.

  • Anti-FGF23 antibody intervention: Several studies and reviews discuss the use of anti-FGF23 antibodies or inhibitors to modulate tumor growth, particularly in cancers with elevated FGF23 signaling (e.g., multiple myeloma, prostate cancer, or bone tumors), and suggest that in vivo efficacy and immune profile changes—including TIL characterization—are best examined using mouse syngeneic models.

  • While humanized mouse models (e.g., NSG mice engrafted with human immune cells) are used for certain immuno-oncology studies, they are not commonly referenced in the literature for anti-FGF23 antibody evaluation with a focus on TILs, likely due to technical complexity and limitations in recapitulating human FGF23-immune interactions.

Additional Relevant Information:

  • Multiple syngeneic tumor models (MC38, CT26, RENCA, B16F10, TC-1, etc.) are widely used to enable comparative profiling of immune infiltrates following antibody treatment, providing critical data for preclinical immunotherapy development.

  • TIL characterization in these studies typically uses multicolor flow cytometry and gene expression profiling to analyze how interventions (such as anti-FGF23 antibodies) impact the immune composition within the tumor microenvironment.

  • For cancers with significant bone involvement, the biological impact of FGF23 (and its therapeutic targeting) appears especially relevant; mouse models of these tumor types are standard platforms for discovery and validation studies.

In summary, syngeneic mouse tumor models are the predominant platform where research-grade anti-FGF23 antibodies are administered in vivo to study both tumor growth inhibition and the characterization of TILs. Humanized models are mainly reserved for translational studies requiring human-specific immune responses and are less commonly utilized for anti-FGF23 and TIL research.

Researchers commonly use Burosumab biosimilars—which target and neutralize FGF23 signaling—in complex immune-oncology models to investigate how modifying the tumor microenvironment or host metabolism might interact with or potentiate the effects of immune checkpoint inhibitors such as anti-CTLA-4 and anti-LAG-3 biosimilars. This approach typically involves combining Burosumab with checkpoint inhibitors to evaluate synergistic effects on tumor immunity or therapeutic outcomes in preclinical models.

Context and Supporting Details:

  • Burosumab biosimilar functions by blocking FGF23, a hormone linked to phosphate metabolism and bone health. Though its primary research applications are in metabolic and bone diseases such as X-linked hypophosphatemia (XLH), researchers are investigating its impact on the immune system and tumor environments, as FGF23 pathways may modulate immune cell function.

  • Checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3 biosimilars) block negative regulators of T-cell activation, unleashing immune responses against tumor cells. These inhibitors act through distinct immune pathways—CTLA-4 mainly affects early T-cell activation, while LAG-3 inhibits T-cell proliferation and function in the tumor microenvironment.

Synergy Studies:

  • Combining Burosumab with checkpoint inhibitors in experimental models allows researchers to study synergistic effects such as:

    • Enhanced T-cell activation or infiltration into tumors.
    • Changes to the immunosuppressive tumor microenvironment mediated by FGF23 pathway blockade.
    • Effects on bone and mineral metabolism that might indirectly influence immune cell energetics or trafficking.
  • These combinations are assessed in functional assays and advanced animal models. Endpoints often include tumor growth inhibition, immune cell profiling, and analyses of cytokine production or bone phenotype changes.

  • Researchers select biosimilars rather than clinical-grade antibodies for cost, consistency, and reproducibility in basic and translational studies.

Additional Notes:

  • There is no direct clinical evidence yet for Burosumab/checkpoint inhibitor combinations; current research is largely preclinical or exploratory.
  • The complexity of these studies allows for testing multi-dimensional immune and metabolic responses that are relevant for developing new combinatorial cancer therapies.

In summary, researchers use Burosumab biosimilars with other checkpoint inhibitors in immuno-oncology models to dissect interactions between tumor immunity, metabolism, and immune regulation, focusing on functional and synergistic effects in controlled experimental systems.

API Error 503: <html><body><h1>503 Service Unavailable</h1>No server is available to handle this request.<script>(function(){function c(){var b=a.contentDocument||a.contentWindow.document;if(b){var d=b.createElement('script');d.innerHTML="window.__CF$cv$params={r:'986e2e6c9f0f4de2',t:'MTc1OTE3NzIzMS4wMDAwMDA='};var a=document.createElement('script');a.nonce='';a.src='/cdn-cgi/challenge-platform/scripts/jsd/main.js';document.getElementsByTagName('head').appendChild(a);";b.getElementsByTagName('head').appendChild(d)}}if(document.body){var a=document.createElement('iframe');a.height=1;a.width=1;a.style.position='absolute';a.style.top=0;a.style.left=0;a.style.border='none';a.style.visibility='hidden';document.body.appendChild(a);if('loading'!==document.readyState)c();else if(window.addEventListener)document.addEventListener('DOMContentLoaded',c);else{var e=document.onreadystatechange||function(){};document.onreadystatechange=function(b){e(b);'loading'!==document.readyState&&(document.onreadystatechange=e,c())}}}})();</script></body></html>

References & Citations

1. Fukumoto S. J Mol Endocrinol. 2021;66(2):R57-R65.
2. Imanishi Y, Ito N, Rhee Y, et al. J Bone Miner Res. 2021;36(2):262-270.
3. Athonvarangkul D, Insogna KL. Calcif Tissue Int. 2021;108(1):143-157.
4. Schindeler A, Biggin A, Munns CF. Front Endocrinol (Lausanne). 2020;11:338.
5. Lamb YN. Burosumab: First Global Approval. Drugs. 2018;78(6):707-714.
Indirect Elisa Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.