Anti-Human FOLR1 (folate receptor 1) (Mirvetuximab) – Fc Muted™

Anti-Human FOLR1 (folate receptor 1) (Mirvetuximab) – Fc Muted™

Product No.: F535

- -
- -
Product No.F535
Clone
M9346A
Target
FOLR1 (folate receptor 1)
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Folate receptor alpha (FRα); Adult folate-binding protein (FBP); Folate receptor 1; Folate receptor, adult; KB cells FBP; MOv18
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 Mirvetuximab. Mirvetuximab (M9346A) is a monoclonal antibody that specifically targets Folate receptor 1 (FOLR1).
Background
Folate receptor 1 (FOLR1), also known as folate receptor alpha (FRα), is a glycosylphosphatidylinositol-anchored glycoprotein essential for transporting folate into cells. Elevated FOLR1 levels are linked to poor prognosis in cancers such as breast cancer and endometrial carcinoma, making it a potential target for cancer therapy. It has also been investigated as a biomarker for cancer diagnosis and a target for delivering cytotoxic agents, particularly in cancers affecting women1,2.

Mirvetuximab (M9346A) is an antibody-drug conjugate that targets folate receptor alpha (FRα), commonly overexpressed in cancers such as ovarian cancer. The initial phase III FORWARD I trial did not show significant benefits in FRα-positive tumors. However, subsequent studies focusing on patients with high FRα expression produced promising results. Consequently, the US FDA granted accelerated approval for Mirvetuximab in patients with FRα-positive, platinum-resistant ovarian, fallopian tube, or primary peritoneal cancer. Mirvetuximab has demonstrated efficacy in reducing the risk of tumor progression or death compared to chemotherapy, providing a valuable treatment option for advanced or recurrent ovarian cancer3.

This research-grade biosimilar does not contain the drug conjugate.

Antigen Distribution
While normally expressed on the apical surfaces of healthy epithelial tissues, FOLR1 is overexpressed in various solid tumors, including ovarian, breast, lung, and gastric cancers.
Ligand/Receptor
Folate and reduced folate derivates
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Tumor Suppressors

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 Mirvetuximab biosimilars serve as analytical calibration standards and reference controls in pharmacokinetic (PK) bridging ELISAs to reliably quantify Mirvetuximab concentration in serum samples from clinical studies.

Essential context and methodology

  • Single Analytical Standard Approach: For PK bridging assays when comparing the reference Mirvetuximab product with its biosimilar, the current best practice is to develop a single ELISA method using one analytical standard (usually the biosimilar) as the calibrator. This ensures both products are measured with the same assay conditions, minimizing operational variability and the need for complex cross-validation or crossover analysis.

  • Assay Calibration: The biosimilar is validated to ensure its bioanalytical equivalence with the reference drug. Calibration standards (typically a dilution series of the biosimilar in pooled human serum) are used to create the standard curve that determines drug concentration in test samples. The ELISA's performance characteristics—including accuracy, precision, and linearity—are validated using both biosimilar and reference Mirvetuximab samples at multiple concentrations.

  • Reference Controls: Quality control (QC) samples, prepared from both the biosimilar and the reference product at defined concentrations, are included in the assay runs. This verifies that the biosimilar standard provides equivalent response to the reference product, confirming that the PK data generated for both are directly comparable.

  • Bridging Design: The PK bridging ELISA essentially “bridges” the measurement between the reference and biosimilar by demonstrating that both can be accurately quantified using the same biosimilar-calibrated assay. Analytical equivalence is assessed statistically—often by comparing the 90% confidence interval of measured concentrations for both products against a predefined equivalence margin (e.g., 0.8–1.25).

  • Assay Performance Criteria

    • Precision and Accuracy: Inter-assay and intra-assay precision (<10% CV typical) and accuracy across relevant serum concentrations ranges must be within predefined validation targets to meet regulatory bioanalytical method standards.
    • Sensitivity and Linearity: The calibration range is established to reliably quantify the lowest to highest expected serum concentrations. Acceptable accuracy is typically 90-113% across the quantitation range.

Additional relevant details

  • Kits may be validated against international standards where available, or against the innovator drug and commercial biosimilars, further boosting confidence in the calibration approach.
  • Matrix effects are assessed by spiking both calibration and control samples into pooled human serum to optimize detection and ensure there is no interference from serum proteins in patient samples.
  • Lyophilized biosimilar standards are preferred for long-term stability and reproducibility in commercial ELISA kits.
  • Confirmatory studies using other bioanalytical platforms (e.g., LC-MS/MS with biosimilar-based calibration) have shown biosimilars provide indistinguishable quantitative results compared to the innovator, affirming their appropriateness as calibration standards.

In summary, research-grade Mirvetuximab biosimilars are rigorously characterized and validated to be analytically equivalent to the reference product. They are then used as calibration standards and reference controls in PK bridging ELISA assays, ensuring consistent, reproducible, and regulatory-compliant measurement of Mirvetuximab levels in pharmacokinetic serum studies.

The primary in vivo models where research-grade anti-FOLR1 antibodies are administered to study tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) characterization are chiefly human tumor xenograft models in immunodeficient mice and, less commonly, various humanized mouse models.

Key Models:

  • Human Tumor Xenografts in Immunodeficient Mice (NSG, nude, SCID)

    • These models use human cancer cell lines (e.g., ovarian or lung cancer lines expressing FOLR1 such as NCI-H2170 or SKOV-3) implanted subcutaneously in immunodeficient mice.
    • Anti-FOLR1 antibodies, such as farletuzumab (MORAb-003), are administered to evaluate inhibition of tumor growth via mechanisms like antibody-dependent cellular cytotoxicity (ADCC).
    • Since these mice lack a fully functional immune system, native TIL analysis is limited mostly to innate populations or to humanized versions.
  • Humanized Mouse Models

    • Humanized NSG or NOG mice are engrafted with human hematopoietic stem cells or peripheral blood mononuclear cells (PBMCs), allowing for components of a human-like immune response.
    • Some recent approaches involve combining anti-FOLR1 bispecific antibodies (e.g., FOLR1-TCBs) that recruit human T cells, enabling analysis of tumor-infiltrating human lymphocytes (e.g., CD3+ T cells) following antibody treatment.
    • These models enable characterization of TILs such as CD8+ or CD4+ T cells and their functional status post-treatment.
  • Syngeneic Mouse Models

    • Standard murine syngeneic tumor models use mouse cancer cells in immunocompetent mice, but typically, mouse FOLR1 is not targeted by human anti-FOLR1 antibodies, limiting use for TIL research unless specifically engineered.
    • Some studies employ mouse surrogate FOLR1 antibodies if available, but this is rare for clinical-grade or translational research.

Additional Details:

  • Most TIL characterization following anti-FOLR1 therapy is performed using flow cytometry or immunohistochemistry to determine immune cell infiltration (e.g., CD3, CD8, granzyme B status, activation/exhaustion markers) in tumor tissue.
  • Antibody localization, tumor-binding kinetics, and ADCC have been evaluated in real-time using fluorescently labeled anti-FOLR1 antibodies in xenograft models.

Summary Table:

Model TypeFOLR1 TargetImmune ContextTIL Analysis Possible?
Human tumor xenograft (immunodeficient mice)HumanLargely innate (mouse)Limited/indirect
Humanized mouse modelsHumanHuman (engrafted)Yes (human TILs)
Syngeneic mouse models (immunocompetent mice)MouseMouseRarely (need mouse mAb)

Humanized and xenograft models are dominant for in vivo anti-FOLR1 antibody studies of tumor growth inhibition, with humanized mice required for direct TIL characterization. Syngeneic models are less relevant unless specifically engineered for cross-species FOLR1 targeting.

Researchers use Mirvetuximab biosimilar, which targets folate receptor alpha (FRα), in conjunction with checkpoint inhibitor biosimilars (such as anti-CTLA-4 or anti-LAG-3) to study synergistic immune responses in complex immune-oncology models by combining antigen-targeted antibody therapies with immunomodulatory agents in preclinical and functional assays.

Researchers typically design studies that include:

  • Functional assays: Mirvetuximab biosimilar is used to engage FRα-expressing tumor cells, mimicking how the therapeutic antibody would target and potentially eradicate cancer cells in vivo. When combined with checkpoint inhibitors—biosimilars to anti-CTLA-4, anti-PD-1, or anti-LAG-3—they can measure enhanced T-cell activation, increased tumor-infiltrating lymphocytes (TILs), and reduction in regulatory T cells, indicating a more robust immune response.
  • Synergy evaluation: By using both agents together, researchers observe whether the direct tumor targeting of Mirvetuximab amplifies the effects of checkpoint blockade (e.g., increased cytokine production, improved antigen-specific immune activation, decreased immune suppression within the tumor microenvironment).
  • Model systems: These studies are conducted on complex immune-oncology model systems, including patient-derived xenografts, syngeneic mouse models expressing human targets, or co-culture systems combining human immune cells and FRα+ cancer cells, thereby replicating relevant aspects of tumor-immune interactions.

Checkpoint inhibitor biosimilars (e.g., anti-CTLA-4, anti-LAG-3) are often chosen to provide highly specific immune checkpoint blockade, allowing the research team to dissect mechanisms of immune activation. For instance, CTLA-4 blockade promotes T-cell priming and reduces regulatory T cells, while anti-PD-1/PD-L1 agents restore exhausted T cell function.

Mirvetuximab biosimilar used in combination studies is research-grade and lacks the cytotoxic drug conjugate, making it suitable for mechanistic immunology and synergy research without inducing direct cell death that would confound immune measurements.

In summary, by combining Mirvetuximab biosimilar with checkpoint inhibitor biosimilars, researchers systematically investigate how tumor-targeted antibody-drug conjugates and immune pathway modulators interact to enhance antitumor immunity in complex preclinical settings. This approach helps identify promising therapeutic combinations and clarify molecular mechanisms underlying potential synergy.

In a bridging ADA ELISA for immunogenicity testing, a Mirvetuximab biosimilar can serve as both the capture and detection reagent to monitor a patient's immune response against the therapeutic Mirvetuximab drug. The biosimilar is used in place of the originator molecule to reliably detect anti-drug antibodies (ADAs) that form in response to treatment.

Assay Principle and Reagent Functions:

  • The wells of an ELISA plate are coated with Mirvetuximab biosimilar (often in a biotinylated or otherwise immobilized format) to capture any ADAs present in the patient’s serum.
  • After the patient sample is added, ADAs that bind Mirvetuximab attach to the biosimilar-coated plate.
  • A second, labeled (e.g., HRP-conjugated or dye-labeled) Mirvetuximab biosimilar is then added, which binds to the other "arm" of any bivalent ADA bound to the capture reagent, forming a "bridge".
  • Following a wash, signal development (such as colorimetric change via TMB substrate) is measured, which is proportional to the amount of ADA present.

Rationale for Using a Biosimilar:

  • The biosimilar is structurally and functionally highly similar to the reference Mirvetuximab, ensuring equivalent ADA binding.
  • Using a biosimilar allows immunogenicity monitoring even if the original drug is scarce or reserved for other testing, supporting drug development and regulatory comparison studies.
  • Data indicate that well-developed and validated assays employing either the reference or the biosimilar molecule provide reliable ADA detection without altering immunogenicity profiles or assay sensitivity.

Clinical Application:

  • This format allows monitoring of ADA formation in patients receiving therapeutic Mirvetuximab, to evaluate safety, effectiveness, and potential for immune-mediated adverse events or decreased efficacy.
  • The bridging assay is sensitive to bivalent antibodies and compatible with most monoclonal antibody biologics, including Mirvetuximab and its biosimilars.

Key Steps Summarized:

  • Coat plate with Mirvetuximab biosimilar (capture reagent).
  • Incubate with patient serum (may contain ADAs).
  • Add labeled Mirvetuximab biosimilar (detection reagent).
  • Detect bridge formation via enzymatic or fluorescent signal.

This protocol is widely validated for monitoring immunogenicity to monoclonal antibodies in clinical studies.

References & Citations

1. Liu Y, Lian T, Yao Y. Biomarkers. 2020;25(5):367-374.
2. Ginter PS, McIntire PJ, Cui X, et al. Clin Breast Cancer. 2017;17(7):544-549.
3. G B, Rl C, I V, et al. International journal of gynecological cancer : official journal of the International Gynecological Cancer Society. 2024;34(4).
4. Mirvetuximab - Search Results - MyBioSource. Accessed October 5, 2024. https://www.mybiosource.com/search/mirvetuximab

Formats Available

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