Anti-Human Carbonate dehydratase IX (CAIX) (Girentuximab)

Anti-Human Carbonate dehydratase IX (CAIX) (Girentuximab)

Product No.: C3130

- -
- -
Product No.C3130
Clone
WX-G250
Target
Carbonate dehydratase IX (CAIX)
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Carbonate dehydratase IX, CA-IX, CAIX, Membrane antigen MN, P54/58N, RCC-associated antigen G250, pMW1
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC
,
IF
,
IP
,
N
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 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
Additional Applications Reported In Literature ?
ELISA,
FA,
FC,
IF,
IP,
N,
WB
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 sequenceas the therapeutic antibody Girentuximab. Girentuximab specifically targets and binds toCAIX. This product is research use only.
Background
Carbonic anhydrase IX (CAIX) is a type of transmembrane metalloenzyme that has a significant role in maintaining the pH balance within cells. It is predominantly found in the gastrointestinal tract and gallbladder, where it facilitates acid secretion. In various types of cancer, such as clear cell renal cell carcinoma (ccRCC), and breast, lung, and cervical cancers, CAIX is often overexpressed in response to low oxygen levels (hypoxia) and is linked to a poor prognosis. CAIX contributes to the acidification of the surrounding environment, thereby promoting the growth and spread of tumors1-3.

Girentuximab, also known as Rencarex, is a type of chimeric IgG1 monoclonal antibody that is designed to target CAIX3. Researchers have been studying its potential in treating renal cell carcinoma (RCC) because it effectively targets CAIX, which is typically found in high levels in most RCC cells. Girentuximab functions by initiating antibody-dependent cellular cytotoxicity (ADCC), destroying tumor cells that express CAIX. Despite the discontinuation of its development as a standalone antibody during phase III trials, scientists are still examining its use as a radioimmunoconjugate for both diagnostic and therapeutic applications4,5.
Antigen Distribution
CAIX is primarily expressed in the gastrointestinal tract and gallbladder. Its expression is significantly upregulated in various solid tumors, particularly in hypoxic environments.
Ligand/Receptor
Carbon dioxide
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Cancer Research

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 Girentuximab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA by preparing calibration curves, quality controls, and spiked samples, enabling quantification of Girentuximab drug concentrations in serum samples.

Essential context and supporting details:

  • Calibration Standards: In a PK bridging ELISA, calibration standards are prepared by spiking known concentrations of research-grade Girentuximab biosimilar into pooled human serum or another relevant matrix. These standards define the ELISA’s dynamic range, allowing construction of a standard curve correlating the ELISA signal (e.g., absorbance) with known drug concentrations.

  • Quality Controls (Reference Controls): Alongside the calibration standards, additional samples—called quality control (QC) samples—are prepared at low, medium, and high concentrations of the Girentuximab biosimilar in serum. These serve to monitor the accuracy and precision of the assay in each run and between runs.

  • Bridging ELISA Principle: The PK bridging ELISA typically uses anti-idiotypic antibodies or the antigen (CAIX for Girentuximab) in a “bridging” format, capturing Girentuximab from serum and detecting it with a secondary reagent.

  • Role of Biosimilars: Research-grade Girentuximab biosimilars are specifically used because they mimic the structure, binding properties, and immunoreactivity of the clinical drug, ensuring the signal measured in the ELISA reflects the presence of Girentuximab in patient samples. The biosimilar is often purified and characterized for high purity (>95%), which is essential for accurate standard and QC preparation.

  • Assay Calibration and Validation:

    • Assay standards and QCs are included on each ELISA plate to account for inter-plate and inter-assay variability.
    • The performance of the assay (sensitivity, specificity, linearity, precision) is validated using these standards, which may be cross-calibrated against internationally recognized reference materials or innovator (original) drug where available.
  • Measurement Process: Patient serum samples are analyzed in parallel with the standards and QCs. The drug concentrations in unknown samples are interpolated from the standard curve generated using the biosimilar-spiked calibration standards.

Summary Table: Use of Girentuximab Biosimilars in PK ELISA

ApplicationDetails
Calibration StandardSpiked into serum to make standard curve for quantification
Reference Control (QC)Spiked at defined concentrations to verify assay accuracy & consistency
Assay PrincipleTypically a bridging ELISA using CAIX antigen/anti-idiotypic Ab
Biosimilar PropertiesMatched to clinical Girentuximab for structure and binding; high purity

Additional relevant points:

  • It is essential that the biosimilar used as a standard is well-matched to the clinical drug in antibody sequence, affinity, and glycosylation profile.
  • Calibration with commercial or research-grade biosimilars is especially important in therapeutic drug monitoring or biosimilar/innovator comparability studies.
  • These procedures follow regulatory guidelines (FDA/EMA/ICH) for bioanalytical assay validation.

If not specified by kit instructions, each lab should optimize and document their calibration strategy before use in regulated studies.

The primary in vivo models for administering a research-grade anti-carbonic anhydrase IX (CAIX) antibody to study tumor growth inhibition and characterization of tumor-infiltrating lymphocytes (TILs) are humanized mouse models bearing human tumor xenografts, particularly orthotopic renal cell carcinoma (RCC) models engrafted in immunodeficient mice with co-transfer of human immune cells.

Key models and approaches:

  • Humanized Orthotopic Xenograft Model:

    • Tumor: Human CAIX^+^ RCC cells, often luciferase-labeled for tracking, orthotopically implanted (in original anatomical site, e.g., the kidney) into immunodeficient mice (typically NOD/SCID/IL2Rγ^−/−, also known as NSG mice).
    • Immune Reconstitution: These mice are then injected with allogeneic human peripheral blood mononuclear cells (PBMCs), which provide human immune effectors (e.g., NK cells, T cells).
    • Antibody Administration: Human anti-CAIX monoclonal antibodies are administered in this setting to test for tumor inhibition and effects on human immune infiltration.
    • TIL Characterization: Tumors are analyzed by histology and FACS to quantify and phenotype tumor-infiltrating human immune cells, including T cells and NK cells, as a measure of immune activation and recruitment.
  • Standard Xenograft Models (without human immune cells):

    • Tumor: Human CAIX^+^ cell lines (commonly HT29, RCC cell lines) implanted into immunodeficient (nude or NSG) mice.
    • Limitation: These models allow assessment of direct anti-tumor effects of CAIX inhibition but are not well suited for comprehensive TIL characterization due to lack of a functional or a human-like immune compartment.
    • Syngeneic Models: Some studies use murine CAIX inhibitors/antibodies in syngeneic mouse tumor models (e.g., 4T1 breast tumor in BALB/c), but these do not allow for assessment of human TILs or direct translation of human anti-CAIX antibodies.

Summary Table: Primary Models for Anti-CAIX Antibody Studies

Model TypeTumor TypeHost MouseKey Immune ComponentTIL AssessmentKey References
Humanized orthotopicHuman RCC, CAIX^+^NSG (NOD/SCID/IL2Rγ^−/−)Human PBMCs injected post-engraftYes
Traditional xenograftHuman CAIX^+^Nude, SCID, or NSGNone or mouse-derivedLimited
Syngeneic murine CAIXMouse tumorImmunocompetent miceMouse immune systemYes (murine)

Conclusion:
The most widely used and scientifically appropriate model for administering anti-CAIX antibodies, evaluating tumor growth inhibition, and profiling human TILs is the humanized NSG mouse bearing an orthotopic human tumor and injected with human PBMCs, enabling both therapeutic and immunological assessments relevant to human biology. Traditional xenograft or syngeneic models do not fully recapitulate human immune responses or permit detailed study of human TILs following administration of research-grade anti-CAIX antibodies.

Researchers utilize the Girentuximab biosimilar in preclinical immune-oncology models to study its interactions and potential synergy with other checkpoint inhibitors, such as anti-CTLA-4 and anti-LAG-3 biosimilars, primarily by combining these agents to amplify anti-tumor immune responses and assess mechanisms of resistance.

Girentuximab biosimilar specifically targets carbonic anhydrase IX (CAIX), a protein highly expressed in hypoxic tumor regions, and mediates antibody-dependent cellular cytotoxicity (ADCC), which recruits immune effector cells like NK cells to destroy tumor cells. In research settings, this targeted mechanism is combined with checkpoint inhibitors such as anti-CTLA-4 (which blocks inhibition of T-cell activation) and anti-LAG-3 (which releases additional inhibitory brakes on T cells) to observe whether the combination leads to greater immune activation and tumor cell clearance than single agents alone.

Key strategies in these preclinical studies include:

  • Combination therapy protocols: Girentuximab biosimilar is administered with checkpoint inhibitor biosimilars to tumor-bearing animal models or tumor spheroids to probe for enhanced immune cell infiltration, cytokine production, and tumor regression compared to monotherapy groups.
  • Evaluation of synergistic effects: Researchers test whether the combination leads to additive or multiplicative effects on anti-tumor immunity. For example, combining Girentuximab's ADCC-mediated tumor targeting with immune checkpoint blockade may more effectively activate cytotoxic T cells and overcome immune suppression within the tumor microenvironment.
  • Mechanistic studies: Complex immune-oncology models allow mapping of cell populations, immune signaling, and the interplay between checkpoint pathways and antibody targeting. Synergy is assessed via flow cytometry, immunohistochemistry, and cytokine profiling to determine changes in T cell activation, exhaustion markers, and effector function.
  • Imaging diagnostics: Radiolabeled forms of Girentuximab (e.g., 124I-Girentuximab or 89Zr-DFO-Girentuximab) are used for precision PET imaging, enabling researchers to monitor tumor progression and therapeutic response dynamically in combination therapy settings.

The core purpose of these studies is to optimize combination regimens for solid tumors (especially renal cell carcinoma), as well as to uncover resistance mechanisms that may limit the effectiveness of checkpoint inhibitors alone. The use of biosimilars makes these analyses cost-effective and reproducible for large-scale experimentation.

In summary, Girentuximab biosimilar is paired with checkpoint inhibitor biosimilars in advanced immune-oncology models to explore synergistic anti-tumor responses, characterize mechanisms of action and resistance, and guide future clinical translation for combination immunotherapies.

A Girentuximab biosimilar can be used as the capture and/or detection reagent in a bridging Anti-Drug Antibody (ADA) ELISA to measure a patient's immune response against therapeutic Girentuximab. This is accomplished by exploiting the drug’s ability to simultaneously bind to both arms of patient-derived anti-drug antibodies (ADAs), forming a "bridge" between two Girentuximab molecules, one immobilized and one labeled for detection.

Key steps in the bridging ADA ELISA using a Girentuximab biosimilar:

  • The biosimilar Girentuximab is immobilized on the surface of an ELISA plate, serving as the "capture reagent."
  • Patient serum samples (containing potential ADAs) are incubated with the plate. Any ADA specific to Girentuximab will bind the immobilized Girentuximab.
  • A second Girentuximab biosimilar, typically labeled (for example, with HRP for colorimetric detection or biotin for subsequent streptavidin labeling), is then added. This labeled Girentuximab molecule binds to the other free arm of the ADA, completing the bridge.
  • After washing, detection is achieved via the label, indicating the presence and quantity of Girentuximab-specific ADAs in the patient sample.

This format ensures specificity for antibodies that recognize Girentuximab (and its biosimilar). High-sensitivity detection is achieved by using sufficiently pure biosimilar Girentuximab as both the capture and detection reagent, mimicking the therapeutic drug's epitopes. This approach is standard for monoclonal antibody therapeutics and their biosimilars when assessing immunogenicity.

Additional context:

  • The bridging ELISA format detects bivalent ADAs, meaning those capable of simultaneously binding two Girentuximab molecules.
  • Biosimilars must be highly similar in structure to the originator drug to ensure recognition by all relevant patient ADAs, which is why a Girentuximab biosimilar is used instead of unrelated antibodies.
  • Similar assay designs have been documented for numerous mAb drugs, confirming the broad acceptance of this approach for immunogenicity monitoring.
  • Assay sensitivity, specificity, and interference from endogenous Girentuximab (therapeutic levels in serum) are also considerations in final assay validation.

In summary, Girentuximab biosimilar acts as both capture and detection reagent in bridging ADA ELISA, allowing detection of antibodies formed against the therapeutic drug in patient samples by forming a Girentuximab–ADA–Girentuximab immunocomplex, which is then detected via an appropriate label.

References & Citations

1. McDonald PC, Dedhar S. Subcell Biochem. 2014;75:255-269.
2. John A, Sivashanmugam M, Natarajan SK, Umashankar V. J Biomol Struct Dyn. 2020;38(7):1995-2006.
3. Choschzick M, Woelber L, Hess S, et al. Virchows Arch. 2010;456(5):483-490.
4. Muselaers CHJ, Boers-Sonderen MJ, van Oostenbrugge TJ, et al. Eur Urol. 2016;69(5):767-770.
5. Zatovicova M, Jelenska L, Hulikova A, et al. Int J Oncol. 2014;45(6):2455-2467.
6. Girentuximab Overview - Creative Biolabs. Accessed August 15, 2024. https://www.creativebiolabs.net/girentuximab-overview.htm?gclid=Cj0KCQjwzva1BhD3ARIsADQuPnXnAYWy3vGompPNowNBCoWbRrzTxgMjMZ-_SqLl1_wCX_5YaQfDRPwaAhNYEALw_wcB
7. Anti-CA9 Antibody (girentuximab biosimilar) (WX-G250), 12-9100. Accessed August 15, 2024. https://www.abeomics.com/anti-ca9-antibody-girentuximab-biosimilar-wx-g250
Indirect Elisa Protocol
FA
Flow Cytometry
IF
Immunoprecipitation Protocol
N
General Western Blot Protocol

Certificate of Analysis

Formats Available

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