Anti-Human Fibroblast Growth Factor 23 (Burosumab) – Fc Muted™

Anti-Human Fibroblast Growth Factor 23 (Burosumab) – Fc Muted™

Product No.: F545

- -
- -
Product No.F545
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

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Muted
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 analytical standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to ensure accurate and equivalent measurement of Burosumab concentrations in serum samples across different product batches or sources.

In a typical PK bridging ELISA for Burosumab:

  • A quantitative sandwich ELISA format is usually adopted, where capture and detection antibodies specific for Burosumab are used to bind and detect the drug in serum.
  • Calibration standards and quality controls (QCs) are prepared using the research-grade Burosumab biosimilar, often over a defined range of concentrations that bracket the expected levels in serum samples.
  • The biosimilar standard provides the reference for constructing a calibration (standard) curve, vital for determining unknown drug concentrations during sample analysis.

Key steps and considerations:

  • Method qualification: Both the biosimilar and any reference product (e.g., the originator Burosumab) are initially tested for bioanalytical comparability—ensuring that the assay measures both with similar accuracy and precision. Precision and accuracy data sets are analyzed statistically to confirm they are bioanalytically equivalent within the method. If comparability is confirmed, one (commonly the biosimilar) is selected as the sole analytical standard.
  • Single-standard approach: Using a single, well-characterized research-grade biosimilar as the standard reduces variability, simplifies assay validation, and helps eliminate analytical confounders in comparative PK studies.
  • Bridging calibration: The validated assay with the biosimilar standard is used to quantify serum concentrations from both biosimilar and reference product dosing arms in PK studies, supporting direct bioequivalence evaluations.

Practical Example:

  • Calibration standards are prepared at multiple concentrations (e.g., 50–12,800 ng/mL), and QC samples are also generated at critical points (e.g., low, medium, high concentrations) using both test and reference products.
  • Serum samples from study subjects are interpolated against this reference standard curve to report Burosumab concentrations.

Summary Table:

Use of Biosimilar StandardPurpose in PK ELISAKey Requirement
Standard curve constructionQuantify Burosumab in test samplesAnalytical comparability
Reference control for QCsValidate assay accuracy and monitor run performanceConsistency with reference
Single-standard PK bridgingSupport comparison between biosimilar and reference PK profilesValidation and equivalence

Critical controls:

  • Analytical comparability confirmation between biosimilar standard and reference product is required for regulatory acceptance in bioequivalence or bridging studies.

Limitation:

  • Research-grade biosimilar products are for research use only (RUO) and not suited for clinical diagnostic or therapeutic applications.

Thus, research-grade Burosumab biosimilars serve as key standards and controls in PK ELISA to enable accurate, robust, and comparable measurement of drug concentrations in bridging and bioequivalence studies.

The primary models for in vivo administration of research-grade anti-FGF23 antibodies to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are mouse syngeneic tumor models, though humanized models are theoretically possible but not widely documented for this purpose.

Supporting context and details:

  • Syngeneic tumor models utilize tumor cell lines derived from the same genetic background as the host mouse (e.g., MC38 in C57BL/6 mice, CT26 in BALB/c mice), preserving a fully functional immune system. These models are ideal for immunotherapy research, enabling detailed study of tumor growth, immune cell infiltration, and TIL phenotypes after antibody administration.
  • Studies profiling the immune microenvironment and examining TILs frequently utilize multi-color flow cytometry and other immunophenotyping approaches in syngeneic models, since this enables characterization of CD8+ T cells, Tregs, myeloid-derived suppressor cells, and others.
  • Anti-FGF23 antibodies have been reported in preclinical models to inhibit FGF23 actions, including studies in contexts such as bone cancers (e.g., multiple myeloma, prostate cancer), and these often involve murine settings where immune and tumor parameters can be measured.
Model TypeTypical ApplicationImmune SystemTumor SourceTIL Characterization Feasibility
SyngeneicMouse anti-tumor studiesIntact mouseMouse-derived linesHigh (standard approach)
HumanizedHuman immune cell studiesPartially reconstituted human immunity in miceHuman tumor or PDXTechnically possible, less common for anti-FGF23 antibody and more complex
  • Syngeneic models are specifically highlighted as a leading platform for evaluating immunotherapy efficacy, including checkpoint inhibitors and by extension, antibody therapies like anti-FGF23, because they enable robust immunophenotyping and are widely used in preclinical oncology.
  • While humanized mouse models (immunodeficient mice engrafted with human hematopoietic stem cells and/or human tumors) are used for certain human-specific immune studies, there is limited evidence in the literature for the use of anti-FGF23 antibodies in these models for TIL characterization and tumor growth inhibition. Most research so far has focused on syngeneic contexts due to convenience, cost, and immune system complexity.

In summary, syngeneic mouse tumor models are the primary and best-characterized in vivo system for evaluating anti-FGF23 antibody effects on tumor growth and immune infiltration. Humanized models are less frequently used for this specific application, primarily due to technical and biological constraints.

Researchers have not documented the use of Burosumab biosimilars in combination with classic immune checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to study synergistic effects in immune-oncology models, because Burosumab targets a metabolic pathway (FGF23) rather than canonical tumor immune checkpoints. Current evidence and commercial biosimilar offerings show Burosumab's primary use is for investigating phosphate and vitamin D metabolism, bone biology, and endocrine FGF signaling.

Key contextual details:

  • Burosumab is a fully human IgG1 monoclonal antibody targeting FGF23 (fibroblast growth factor 23). Its research applications focus on metabolic bone diseases, phosphate regulation, and vitamin D metabolism.
  • Checkpoint inhibitors (such as anti-CTLA-4, anti-LAG-3, anti-PD-1) function by unleashing antitumor immunity through blockade of immune checkpoints that suppress T cell activity. Studies testing combinations of such checkpoint inhibitors (e.g., anti-CTLA-4 plus anti-PD-1, or anti-LAG-3 with anti-PD-1) are well-documented and have shown synergistic effects in tumor models.
  • Biosimilars are widely used in preclinical research to model the pharmacology of their originator molecules. Immuno-oncology research commonly employs biosimilars of checkpoint inhibitors to study combinatorial efficacy, immune cell phenotypes, and signaling pathways in tumor models.

No evidence supports Burosumab synergy studies with anti-CTLA-4 or anti-LAG-3 in immune-oncology:

  • There are no sources documenting studies where Burosumab biosimilars are combined with immune checkpoint inhibitors in immuno-oncology animal models or in vitro systems for the purpose of investigating antitumor synergy.
  • Burosumab’s mechanism—blocking FGF23 to restore phosphate homeostasis—does not overlap mechanistically with immune suppression pathways like CTLA-4, PD-1/PD-L1, or LAG-3.

If the intent is to study synergy in tumor immune responses:

  • Combinatorial studies in immuno-oncology models use checkpoint inhibitor biosimilars (anti-CTLA-4, anti-LAG-3, anti-PD-1, etc.) in parallel or sequential regimens. These studies examine immune cell activation, antitumor efficacy, and additive or synergistic effects, but do not include agents targeting metabolic hormones like FGF23.

Summary Table: Mechanistic Comparison

Antibody TargetPrimary ActionRole in Immuno-Oncology Research
FGF23 (Burosumab)Regulates phosphate and vitamin DUsed in metabolic/bone biology, not tumor models
CTLA-4 (Ipilimumab)Immune checkpoint (T-cell inhibition)Releases T-cell suppression in tumor immunity
LAG-3Immune checkpoint (T-cell regulation)Complements other checkpoint blockade in tumors

Conclusion:
Burosumab biosimilar is not used in combination with checkpoint inhibitor biosimilars like anti-CTLA-4 or anti-LAG-3 to investigate synergy in immune-oncology models, based on current research applications and available evidence. Standard practice for synergy studies in immuno-oncology focuses on combination checkpoint blockade involving established immune targets.

A Burosumab biosimilar can be used as a capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient's immune response (i.e., the development of ADAs) against the therapeutic drug Burosumab by exploiting its structure identical to the therapeutic molecule.

Principle of the Bridging ADA ELISA with Burosumab Biosimilar:

  • Bridging ADA ELISAs detect patient-derived antibodies (ADAs) against a therapeutic protein by "bridging" two labeled versions of the drug (or biosimilar): one as a capture reagent and one as a detection reagent.
  • The ADA, being bivalent, binds simultaneously to both the immobilized (capture) and the labeled (detection) form of the same drug, forming a "bridge" that can be detected.

How the Burosumab Biosimilar is Used:

  • The biosimilar, matching the variable regions and structure of the clinical Burosumab, is critical for both high specificity and sensitivity in ADA detection.
  • In a typical setup:
    • Capture step: The Burosumab biosimilar is immobilized—either directly on an ELISA plate or via a tag (e.g., biotin-streptavidin system).
    • Sample incubation: Patient serum is added; any ADAs specific to Burosumab will bind to the capture biosimilar.
    • Detection step: A differently labeled (e.g., enzyme-conjugated or biotinylated) Burosumab biosimilar is added; this binds to a different epitope on the captured ADA molecule.
    • Signal: After washing, the amount of detection reagent retained is proportional to the ADA levels and can be measured using a suitable substrate.

Why Use a Biosimilar?

  • The biosimilar is functionally identical to the marketed antibody (Burosumab) but is produced for research purposes.
  • It can be safely and reliably used in immunogenicity assays to mimic the drug in patient samples, enabling detection of patient anti-drug antibodies without depleting valuable clinical-grade product.

Relevant Properties of Burosumab Biosimilar:

  • Fully human IgG1, matching the therapeutic antibody's variable regions, and highly purified for specificity.
  • Suitable for labeling (e.g., biotinylation or HRP conjugation) needed for ELISA development.

Summary of Use in ADA ELISA:

  • The Burosumab biosimilar acts as both the capture and detection reagent in the bridging ELISA format.
  • This allows the detection of anti-Burosumab antibodies in patient samples as a marker of immunogenic response, crucial for monitoring safety and efficacy during therapy.

Note: The general protocol can be customized, and assay validation is required to ensure results are accurate for each laboratory and clinical situation.

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.

Formats Available

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