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.
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 are often used as calibration standards (analytical standards) or reference controls in pharmacokinetic (PK) bridging ELISA assays to quantify drug concentrations in human serum. This practice ensures assay precision, accuracy, and comparability between the biosimilar and the innovator (reference) drug.
Context and Supporting Details:
Calibration standards made from biosimilars are typically validated against known concentrations and, where available, international standards (e.g., NIBSC/WHO). This provides traceability and reliability for quantification.
The current consensus and regulatory best practice is to use a single PK ELISA method with a single analytical standard (often the biosimilar) for quantifying both the biosimilar and reference samples. This reduces variability versus using multiple methods and eliminates the need for crossover analysis in multi-arm clinical comparability studies.
Method validation involves preparing the biosimilar calibration standards in serum over a range of concentrations covering the expected PK profile, such as serial dilutions from nanogram to microgram per milliliter. During validation, both the biosimilar and reference products are measured against the biosimilar standard curve to assess bioanalytical equivalence (accuracy, precision, linearity, specificity).
Analytical equivalence is established through statistical analysis of QC results for the biosimilar and reference drug, verifying that both perform comparably within set equivalence intervals (e.g., 90% CI within 0.8–1.25).
In practice, any cross-verification (measuring the reference with a biosimilar standard, and vice versa) should yield identical and interchangeable results if the biosimilar is analytically equivalent to the reference product. This allows the biosimilar standard to serve reliably throughout PK bridging assays.
Additional Information:
The sandwich ELISA format used in these PK assays commonly relies on anti-idiotypic antibodies for high specificity to the drug (Mirvetuximab), enabling detection at low concentrations without matrix interference.
Validation procedures and lot releases for ELISA kits incorporating biosimilar standards are typically performed following ICH/EMA guidelines, ISO 13485, and FDA/EMEA requirements for bioanalytical method precision (<10% CV inter/intra assay).
In summary, research-grade Mirvetuximab biosimilars are extensively validated and used as reliable calibration standards and reference controls in PK bridging ELISA assays, ensuring the quantitative comparability of biosimilar and innovator drug concentrations in serum samples.
Human tumor xenograft models (using immunodeficient mice implanted with human FOLR1-positive tumor cells) are the primary in vivo systems where research-grade anti-FOLR1 antibodies, such as farletuzumab, are administered to study tumor growth inhibition and the characterization of tumor-infiltrating lymphocytes (TILs). Syngeneic models are generally less common, as mouse FOLR1 differs antigenically from human FOLR1, limiting direct antibody targeting applications unless the antibody cross-reacts or genetically engineered mouse models are used.
Supporting details:
Farletuzumab (MORAb-003), a human FOLR1-targeting monoclonal antibody, has been most extensively studied in immunodeficient mouse models implanted with human ovarian cancer cells overexpressing FOLR1, assessing both tumor growth inhibition and mechanisms like antibody-dependent cellular cytotoxicity (ADCC).
These xenograft models enable direct investigation of antibody binding, tumor inhibition, and immune cell (including TILs) responses, sometimes using fluorescent dye labeling or engineered tumor cells expressing reporters (e.g., luciferase) for real-time tracking and quantification.
Analysis of TILs (including their activation and phenotype) can be performed post-treatment by isolating cells from the tumor and using flow cytometry or immunohistochemistry to profile immune infiltration and function.
Syngeneic models:
Mouse syngeneic models allow testing of immunotherapies in immunocompetent mice, but unless the anti-FOLR1 antibody cross-reacts with mouse FOLR1, their use is limited for this target.
Syngeneic tumor modeling paired with engineered tumor cells expressing human FOLR1, or transgenic mice expressing human FOLR1, is possible but much less frequently reported in published research.
Humanized mouse models:
If the goal is to study human immune cell responses to anti-FOLR1 antibody therapy and TILs in vivo, humanized mice (with a reconstituted human immune system and human FOLR1-positive tumors) provide a more advanced platform.
However, current experimental literature primarily features xenograft models rather than fully humanized systems for anti-FOLR1 antibody studies.
Key tumor types:
Most studies utilize models of ovarian cancer, as FOLR1 is highly overexpressed in these tumors, but lung and breast cancer models have also been explored.
Summary table: | Model Type | Host Immune System | Tumor Origin | Anti-FOLR1 Use | TIL Characterization | Example Reference ||------------------------ |-------------------- |---------------|------------------|---------------------|-------------------|| Human xenograft | Immunodeficient | Human | Yes (primary) | Yes | Farletuzumab study || Syngeneic (mouse) | Immunocompetent | Mouse | Rare (needs cross-reactivity or genetic engineering) | Yes, but rarely for FOLR1 | (general profiling) || Humanized mouse | Human (reconstituted) | Human | Possible, rare for FOLR1 studies | Yes | No reported major studies |
Conclusion: The gold standard for studying anti-FOLR1 antibody efficacy and TILs in vivo is the human tumor xenograft model using immunodeficient mice implanted with human FOLR1-positive tumors. Syngeneic and humanized models may be used but are much less common for direct anti-FOLR1 antibody administration and detailed immune characterization.
Researchers use Mirvetuximab biosimilars—engineered antibodies that target folate receptor alpha (FOLR1)—in conjunction with immune checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 in preclinical studies to evaluate potential synergistic anti-tumor effects in complex immuno-oncology models. This strategy is designed to combine tumor-directed cytotoxicity (from Mirvetuximab) with the immune-activating effects of checkpoint inhibition to maximize tumor immunogenicity and immune-mediated cell killing.
Key points in how these studies are conducted:
Mirvetuximab biosimilars are used as research-grade antibodies (lacking the cytotoxic payload but retaining specificity for FOLR1), allowing focus on antibody-mediated effector functions and immune cell recruitment without direct drug-conjugate toxicity.
In co-culture assays or in vivo tumor models featuring FOLR1-overexpressing human tumor cells and reconstituted human or humanized immune systems, researchers introduce both Mirvetuximab biosimilar (to bind tumor cells) and checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 (to relieve T cell inhibition).
Researchers measure immune activation indicators, such as:
Increased tumor-infiltrating lymphocytes (TILs)
Cytokine production (e.g., IFN-γ, TNF-α)
Tumor regression rates or apoptosis markers
Depletion of immunosuppressive cells (such as regulatory T cells, Tregs), often enhanced by dual checkpoint blockade.
The rationale for combination is to overcome the limited efficacy of single-agent checkpoint inhibitors or antibody–drug conjugates by engaging both innate (via antibody-dependent mechanisms) and adaptive (via checkpoint inhibitor-facilitated T cell activity) arms of the immune system.
Studies often include multiple combination arms, comparing single agents to various dual or even triple combinations (e.g., Mirvetuximab plus anti-CTLA-4, Mirvetuximab plus anti-LAG-3, or all three) to determine which interactions provide the greatest synergistic benefit.
Experimental approaches might involve:
Functional assays such as cytotoxicity or proliferation assays in vitro, using human cancer cell lines and effector cells (like PBMCs or T cells).
In vivo humanized mouse models bearing FOLR1-positive tumors, assessing tumor growth inhibition, immune infiltration, and survival metrics.
While most publicly reported studies combine checkpoint inhibitors with other forms of targeted therapies or with chemotherapy to overcome resistance, the use of research-grade biosimilars for Mirvetuximab specifically allows detailed mechanistic studies to inform and refine clinical combination strategies in immune-oncology research.
A Mirvetuximab biosimilar can be used as both a capture and detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient's immune response by detecting antidrug antibodies (ADAs) against Mirvetuximab in patient samples.
Context and Method: The bridging ADA ELISA format is widely used for immunogenicity testing of therapeutic antibodies and antibody-drug conjugates like Mirvetuximab. The core principle is as follows:
Capture reagent: The ELISA plate is coated with the Mirvetuximab biosimilar (the same protein sequence as the therapeutic Mirvetuximab). This biosimilar binds to any anti-Mirvetuximab antibodies (ADAs) present in the patient's serum.
Detection reagent: A labeled Mirvetuximab biosimilar (commonly biotinylated or conjugated with horseradish peroxidase, HRP) is then added. This will bind to free binding sites on the captured ADA, creating a "bridge" between the capture and detection reagents.
Detection Principle: ADAs in the patient's sample must be bivalent (able to bind two antigens) to form a bridge between the plate-bound Mirvetuximab and the labeled Mirvetuximab. The presence of this bridge allows for detection by a substrate reaction (e.g., colorimetric signal with TMB for HRP), which provides a quantitative or qualitative measure of ADA levels.
Why a biosimilar? A biosimilar can be used if it is structurally and functionally equivalent to the therapeutic Mirvetuximab. This ensures that the assay detects antibodies against the therapeutic product's clinically relevant epitopes without the need to use the (potentially scarce or costly) original drug.
Application to Mirvetuximab: Validated bridging ELISA assays using this format have been employed in clinical studies to assess the immunogenicity of Mirvetuximab, showing they are suitable to reliably detect ADAs specific to all components of the drug (antibody, linker, or payload).
Key advantages:
Highly specific for antibodies against the therapeutic.
Capable of detecting a wide range of immunoglobulin classes, as long as they're bivalent.
Sensitive and compatible with high-throughput screening.
Summary Table: Bridging ADA ELISA with Mirvetuximab Biosimilar
Step
Reagent
Function
Plate coating
Mirvetuximab biosimilar
Captures ADAs from patient serum
Add patient sample
Patient serum
Source of potential ADAs
Detection reagent
Labeled Mirvetuximab biosimilar
Binds captured ADA to form "bridge"
Signal development
Substrate (e.g., TMB for HRP)
Visual readout for ADA detection
This approach is standard and validated for immunogenicity assessment of therapeutic antibodies, including Mirvetuximab.
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