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 Elranatamab. Clone RN613 (Elranatamab) is a
bispecific antibody that targets B-cell maturation antigen (BCMA) and CD3 on T cells.
Background
CD3 is an invariant antigen of the T cell TCR (T cell receptor)1. BCMA is a member of
the tumor necrosis factor family of receptors that regulates B cell maturation,
proliferation, and survival by activating p38/NF-κB and inducing upregulation of
antiapoptotic proteins2. BCMA is highly expressed on multiple myeloma (MM) cells
and is therefore a target of cancer immunotherapy.
Anti-Human CD3 x BCMA bispecific antibodies target both B-cell maturation antigen (BCMA)
on plasma cells and CD3 on T-cells, inducing T-cell activation and cytotoxic activity against
BCMA-expressing plasma cells.
Elranatamab (RN613), a bispecific antibody targeting BCMA and CD3 on T-cells, has shown
potent T-cell-mediated anti-myeloma activity by bringing T-cells close to myeloma cells.
Elranatamab is distributed in the body, with higher concentrations in blood and lymphatic
tissues. In laboratory settings, Elranatamab is used to study the efficacy of bispecific
antibodies in targeting multiple myeloma and to develop therapeutic strategies for relapsed
or refractory multiple myeloma3,4.
Antigen Distribution
CD3ε is a T cell surface glycoprotein. BCMA is predominantly
expressed on the surface of terminally differentiated B cells and is overexpressed
and activated on malignant multiple myeloma B cells (plasmablasts and plasma
cells).
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 Elranatamab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as the analytical standard to generate the standard curve for quantitative measurement of drug concentration in serum samples.
In a PK bridging ELISA, the procedure typically involves the following steps:
Preparation of Standards: Research-grade biosimilar Elranatamab is serially diluted in human serum or plasma to create calibration standards at specified concentrations, covering the expected range of drug concentration.
Assay Calibration: These standards are loaded into the ELISA wells coated with specific binding proteins (in Elranatamab's case, CD3 protein is used as the capture component). The biosimilar standard binds similarly to the capture and detection reagents as the innovator/reference drug because it is analytically and functionally equivalent.
Generation of Standard Curve: After the immunoassay stages (incubation, washing, adding detection reagents such as HRP-conjugated BCMA), the optical density (absorbance) of each standard is measured. The known concentrations of biosimilar standards are plotted against their corresponding absorbance values to generate a standard calibration curve.
Sample Quantification: Serum samples containing unknown concentrations of Elranatamab are tested in parallel. Their absorbance is interpolated against the standard curve to calculate their analyte concentration.
Why biosimilars are used:
If research-grade biosimilars meet analytical equivalence criteria with the reference product (innovator Elranatamab), they can be reliably used as calibration standards. This is validated through robust qualification studies comparing accuracy and precision parameters for both biosimilar and reference standards within the same assay, followed by thorough method validation.
Using a single analytical standard (usually the biosimilar) eliminates variability associated with running separate calibration curves for reference and biosimilar drugs, and ensures direct comparability in PK bridging studies for biosimilarity or bioequivalence.
Quality control (QC) samples and calibration standards may be prepared from either the biosimilar or reference product, but interpolation uses the biosimilar standard curve, as long as bioanalytical equivalence is demonstrated.
Reference controls may also be included alongside calibration standards to monitor assay performance for both biosimilar and innovator products.
Summary of benefits:
Ensures assay consistency and comparability between biosimilar and reference drugs during PK bridging studies.
Reduces analytical variability and regulatory complexity when assessing biosimilarity, especially in blinded clinical trials.
This approach aligns with best practices recommended by regulatory agencies and industry guidance for bioanalytical method development and compliance for biosimilar drug development.
For Elranatamab, such ELISA kits use capture reagents and detection systems (e.g., CD3-coated wells and HRP-conjugated BCMA) that measure functional drug binding, ensuring the calibration standard accurately mimics the reference product's behavior in the assay.
The primary in vivo models used to study the effects of research-grade anti-CD3 × BCMA bispecific antibodies (bsAbs) for tumor growth inhibition and characterization of tumor-infiltrating lymphocytes (TILs) are:
Immunodeficient xenograft models (e.g., NSG mice) engrafted with human multiple myeloma cell lines and infused with human T cells: These models allow for assessment of tumor growth inhibition by the anti-CD3 × BCMA bsAb and facilitate analysis of human TILs within the tumor, as the transferred T cells are of human origin. For instance, MM1S-luciferase^+^ human multiple myeloma cells are injected into NSG mice, followed by administration of human T cells and the bispecific antibody, resulting in significant tumor growth inhibition and extended survival.
CD3-humanized syngeneic mouse models implanted with murine tumors engineered to express human BCMA: In these models, mice express human CD3 instead of murine CD3, and tumors such as B16/hBCMA melanoma or MC38/hBCMA colon carcinoma are implanted. Anti-CD3 × BCMA bsAbs administered in these models suppress tumor growth and permit TIL characterization in an immunocompetent context, since the mouse immune system remains broadly intact except for the humanized CD3 T-cell compartment.
Comparison of the models:
Model Type
Primary Use/Advantage
TIL Characterization
Tumor Source
Immune System Status
Xenograft (NSG + human T cells)
Human tumor and T-cell responses; robust for basic efficacy/TIL analyses
Yes (human T cells)
Human MM lines
Severely immunodeficient (NSG mouse); human T cells engrafted
CD3-humanized syngeneic (immunocompetent)
More physiological immune context; can combine with checkpoint blockade
Yes (mouse T cells with human CD3)
Engineered mouse tumor lines
Largely intact mouse immunity with human CD3 T cells
Key Points:
Xenograft models with NSG mice are most common for initial tumor growth inhibition and TIL studies using anti-CD3 × BCMA bsAbs, as they allow controlled introduction of human tumor and effector cells.
CD3-humanized syngeneic models are increasingly used to study combination therapies (e.g., with PD-1 blockade) and for more robust TIL phenotyping in an immunocompetent background, though the tumors must be engineered to express human BCMA.
There are no fully syngeneic multiple myeloma models expressing human BCMA that replicate the human bone marrow microenvironment.
These models are essential for both functional characterization and in-depth analysis of TILs induced by anti-CD3 × BCMA treatment, with each providing complementary insights depending on the immune and tumor cell origin.
Researchers investigating elranatamab biosimilars in combination with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) do so to analyze the synergistic effects on immune activation and anti-tumor efficacy, particularly in advanced and complex immune-oncology models. While no direct studies have been published specifically on elranatamab biosimilars combined with checkpoint inhibitor biosimilars, established research strategies and related studies with checkpoint combinations offer a clear framework.
Key Approaches and Rationale:
Elranatamab Mechanism: Elranatamab is a bispecific antibody that targets BCMA (expressed on plasma cells and myeloma cells) and CD3 (on T cells), effectively redirecting T-cell cytotoxicity toward tumor cells.
Checkpoint Inhibitor Mechanisms:
Anti-CTLA-4 and anti-LAG-3 inhibitors act by blocking negative regulators of T-cell activity, thereby further enhancing T-cell function and proliferation.
Studies with checkpoint inhibitor combinations (such as anti-PD-1 with anti-CTLA-4 or anti-LAG-3) demonstrate that synergy arises from targeting distinct immune-regulatory pathways and activating different T-cell subsets.
Synergy Hypothesis: In complex immune-oncology models (e.g., advanced melanoma mouse models), combined checkpoint inhibition can enhance anti-tumor activity by:
Increasing cytotoxic T-cell activation and infiltration into tumors.
Lowering regulatory cell (Treg) suppression.
Amplifying activation of CD4+ and/or CD8+ T cell responses, as shown by checkpoint inhibitor combinations.
When combined with T cell engagers like elranatamab, these effects are expected to further potentiate tumor killing, since both increased T cell activation and direct re-targeting of T cells at tumor cells occur.
Experimental Models:
Synergy Studies:
Mouse tumor models (often genetically engineered or with patient-derived xenografts) are treated with combinations of T-cell engagers (like elranatamab or surrogate biosimilars) and checkpoint inhibitors (or their biosimilars).
Tumor growth, immune cell infiltration, cytokine release, and survival outcomes are monitored.
Flow cytometry and single-cell RNA-seq are used to profile which immune cell subtypes are activated and how the tumor microenvironment is remodeled.
Endpoints:
Objective Response Rate (ORR), Duration of Response (DOR), and Progression-Free Survival (PFS), as in elranatamab monotherapy studies.
Immune profiling endpoints (e.g., ratios of CD8+ T cells to Tregs, CD4+ T cell activation status).
Translational Impact:
Such preclinical studies are crucial before clinical trials, mapping how combining T-cell engagers with checkpoint blockade can overcome resistance mechanisms and produce more durable responses in refractory cancers.
Evidence from related studies shows that different checkpoint combinations activate different immune-cell programs (e.g., anti-PD-1/LAG-3 requiring CD4+ T cells; anti-PD-1/CTLA-4 more directly activating CD8+ cytotoxic T cells), guiding the experimental design for biosimilar and novel combinations.
Summary Table: Potential Synergistic Effects in Combination Studies
Agent
Mechanism
Key Effects When Combined
Elranatamab
BCMA × CD3 TCE
Direct tumor targeting via T-cell redirection
Anti-CTLA-4
Checkpoint block
Enhanced T-cell priming; increased CD8+ T cell pool
Anti-LAG-3
Checkpoint block
Decreased Treg suppression; promoted CD4+ help
Limitations: There are no published results reporting elranatamab biosimilar use specifically in combination with checkpoint inhibitor biosimilars to date; the strategy outlined is based on how similar immune-oncology antibody combinations are mechanistically studied and on the functional synergy observed in other multi-agent checkpoint regimens in complex tumor models.
In a bridging ADA ELISA designed to monitor immunogenicity against a therapeutic such as Elranatamab, a biosimilar of Elranatamab can be used as either the capture or detection reagent by exploiting the ability of anti-drug antibodies (ADAs) in patient serum to bind two identical or similar drug molecules simultaneously.
Assay principle and reagent roles:
In the assay setup, the Elranatamab biosimilar is conjugated (e.g., with biotin for capture or with HRP/dye for detection).
The patient sample containing potential ADAs is incubated with the biosimilar in two forms:
Capture reagent: The biosimilar is coated on the plate (often via biotin-streptavidin binding, if biotinylated).
Detection reagent: A differentially labeled form (e.g., enzyme-conjugated) of the biosimilar detects the bound ADA.
Bivalent ADAs can bind the biosimilar on the plate with one binding site and the labeled biosimilar in solution with the other. This "bridges" the capture and detection reagents, forming a complex that produces a measurable signal in the presence of ADAs.
Why use a biosimilar as the reagent?
The biosimilar must have identical or highly similar epitopes to those present on the therapeutic Elranatamab to ensure efficient binding by all clinically relevant ADAs.
Using a biosimilar (rather than the originator) is appropriate if it shows equivalent immunoreactivity in assay validation.
Application in Immunogenicity Monitoring:
The assay detects and quantifies anti-Elranatamab antibodies formed in the patient after exposure to the therapeutic, indicating an immune response against the drug.
Positive samples may be further characterized to determine antibody domain specificity or neutralizing activity, per immunogenicity risk assessment best practices.
Considerations:
Matrix effects and potential interference from circulating drug or soluble target can impact specificity; therefore, reagent purity and assay optimization are critical for meaningful results.
For bispecific antibodies like Elranatamab, the assay must be validated to ensure no interference from the drug's dual-target nature.
Summary Table: Use of Elranatamab Biosimilar in Bridging ADA ELISA
Reagent Role
Formulation
Function in Assay
Capture reagent
Plate-bound (e.g., biotinylated)
Immobilizes ADAs from patient serum
Detection reagent
Labeled (e.g., HRP, dye)
Detects ADAs by forming a bridge complex
In summary, the Elranatamab biosimilar serves as both the capture and detection agent in a bridging ADA ELISA by enabling bivalent binding of anti-Elranatamab antibodies, thereby allowing monitoring of the patient’s immune response to the therapeutic.