Anti-Human CD3 x BCMA (Elranatamab) [Clone RN613]

Anti-Human CD3 x BCMA (Elranatamab) [Clone RN613]

Product No.: C3270

- -
- -
Product No.C3270
Clone
RN613
Target
CD3 x BCMA
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
CD3ε: T-cell surface antigen T3/Leu-4 epsilon chain, T3E
BCMA: tumor necrosis factor receptor superfamily member 17, CD269
Isotype
Human IgG2κ

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO 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
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).
Ligand/Receptor
CD3ε: TCR BCMA: TNFSF13B/BLyS/BAFF, TNFSF13/APRIL, TRAF1, TRAF2, TRAF3, TRAF5 and TRAF6
NCBI Gene Bank ID
CD3ε: X03884
BCMA: Z14954
UniProt.org
CD3ε: P07766
BCMA: Q02223
Research Area
Adaptive Immunity
.
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Bispecific Antibody

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 Elranatamab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as the analytical standard to generate a calibration curve against which serum drug concentrations are determined. This approach supports accurate, comparable measurement of both the biosimilar and the reference product in samples from PK studies.

Key points on their use in PK bridging ELISA:

  • Single Analytical Standard: The consensus best practice in biosimilar PK analysis is to use a single, well-characterized biosimilar as the analytical standard for both biosimilar and reference products. This ensures measurement consistency, reduces assay variability, and avoids the need for separate calibration curves.
  • Calibration Curve Generation: The biosimilar is serially diluted in a matrix-matched medium (such as serum) to generate a standard calibration curve, typically across a range of concentrations suitable for the assay (for example, 50–12,800 ng/mL as described for PK assay validation). This curve is used to quantify unknown serum samples.
  • Validation of Analytical Equivalence: Prior to use as a PK standard, the biosimilar and the reference product are rigorously compared in the assay for acceptance criteria such as precision, accuracy, and parallelism. If results are bioanalytically equivalent—meaning the assay's readout is the same for both products—then the biosimilar standard is adopted for all measurements.
  • Quality Controls (QCs): Both the biosimilar and the reference product are also used to prepare QC samples at low, medium, and high concentrations. These QCs are measured alongside study samples to verify assay performance throughout the experimental runs.
  • Bridging Approach: In a bridging ELISA, the assay is designed so it can detect and measure both the biosimilar and the reference product with the same reagents and calibration, enabling direct comparisons of serum concentrations across studies or between products.
  • Assay Description: While the cited study focused on Teclistamab, the methodology is applicable to Elranatamab biosimilars: labeled forms of the bsAb (biotinylated or Alexa-labeled) can be used to check functional binding and assay performance.
  • Supporting Regulatory Bioequivalence: This unified approach underpins the regulatory demonstration of biosimilarity by ensuring PK data for biosimilar and reference drugs are directly comparable within the same validated assay system.

Summary Table: Role of Research-grade Elranatamab Biosimilars in PK Bridging ELISA

PurposeApproach
Calibration StandardUse biosimilar as analytical standard for all calibration curve points
Reference Control/QCPrepare QCs from both biosimilar and reference to validate assay performance
Analytical EquivalenceDemonstrate biosimilar and reference give equivalent signals and parallel responses in the ELISA
PK MeasurementQuantify serum drug levels in study samples using a single calibration curve
Regulatory RelevanceSupports comparative PK analysis of biosimilar vs. reference using the same assay methodology

This approach maximizes assay reliability and supports regulatory traceability for bioanalytical comparability in PK studies.

The primary preclinical in vivo models where anti-CD3 x BCMA bispecific antibodies are administered to study tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) are immunodeficient xenograft models (using human cells in NSG mice) and humanized/syngeneic immunocompetent mouse models engineered to express human CD3.

Essential Context & Supporting Details:

  • Xenograft NSG Mouse Models:
    MM1S human multiple myeloma cells (often modified to express luciferase for imaging) are injected into NSG mice (NOD scid gamma, severely immunodeficient). Human T cells, together with anti-CD3 x BCMA antibodies, are administered intravenously. These models robustly demonstrate tumor growth inhibition and survival benefit. NSG mice lack a functional adaptive immune system, so human immune cell infiltration, including TIL characterization, is primarily limited to the transferred human T cells.

    • Example: “We tested the efficacy of BCMA-CD3 antibodies using the MM1S xenograft mouse NSG model… antibodies with T cells were injected intravenously. All antibodies except PBM0055 significantly decreased tumor growth”.
    • These studies have also examined TILs within the tumor, mainly assessing the fate of transferred human T cells.
  • CD3-Humanized Immunocompetent Mice (Syngeneic Tumor Models):
    Mice are genetically engineered to express human CD3, allowing testing of bispecific antibodies with human CD3 specificity in an otherwise immunocompetent environment. Tumors (such as MC38/hBCMA colon carcinoma or BCMA-expressing melanoma) are implanted, and the anti-CD3 x BCMA bsAb is administered. These models allow for the study of endogenous immune responses, including more physiologically relevant TILs and TME (tumor microenvironment) characteristics.

    • “We developed models in CD3-humanized immunocompetent mice to show BCMAxCD3 bsAb efficacy against BCMA-expressing syngeneic melanoma and colon carcinoma tumors… coadministration of BCMAxCD3 bsAb with PD-1 blockade exhibited combinatorial efficacy in these models”.
  • Limitations & Comparisons:
    Xenograft NSG models are standard for initial bsAb efficacy assessment but lack an intact immune system, so TIL analyses are limited to transferred human T cells and their activation/function.
    CD3-humanized immunocompetent mouse models enable more comprehensive TIL and TME studies, including effects of checkpoint inhibitors, but rely on murine tumors engineered to express human BCMA and CD3, which can differ from human disease biology.

Summary Table: In Vivo Models for Anti-CD3 x BCMA Antibody Testing

Model TypeHost Immune SystemTumor OriginTILs CharacterizedReferences
Xenograft (NSG)ImmunodeficientHuman MM cell linesMainly transferred human T cells
Humanized/SyngeneicHuman CD3 knock-inMurine (engineered BCMA)Endogenous murine/engineered human TILs

Additional Notes:

  • While most published in vivo work uses MM1S xenograft NSG models, recent efforts emphasize CD3-humanized mouse models for immunotherapy studies to better represent TIL dynamics and tumor microenvironment interactions.
  • No fully human/humanized spontaneous syngeneic MM models that comprehensively recapitulate the MM bone marrow microenvironment are currently available for bsAb testing.
  • Analyses typically include tumor burden assessment, T-cell activation and infiltration (by flow cytometry and immunohistochemistry), and sometimes combination studies with checkpoint blockade.

These models are foundational for demonstrating in vivo anti-tumor efficacy and TIL characterization with research-grade anti-CD3 x BCMA antibodies.

Researchers investigate synergistic effects of Elranatamab biosimilars in combination with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars by using complex immune-oncology models, including mouse models and advanced in vitro systems, to evaluate multi-agent immunotherapy strategies.

Elranatamab is a bispecific antibody targeting BCMA and CD3, approved for relapsed/refractory multiple myeloma; it engages T cells to attack malignant plasma cells. While the cited studies focus on single-agent clinical use, the scientific rationale for combining Elranatamab or its biosimilars with other checkpoint inhibitors builds on the mechanistic differences outlined in immune-oncology research:

  • Checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) modulate immune cell activation. Anti-CTLA-4 primarily enhances cytotoxic CD8 T-cell activity, while anti-LAG-3 indirectly boosts CD8 function through CD4 T-cell help and decreases Treg (regulatory T cell) suppression.

  • Combination studies in mouse models of melanoma found that distinct immune cell subtypes are activated depending on the specific checkpoint inhibitor pairing: anti-PD-1/LAG-3 regimens required CD4 T-cell presence, reduced Treg activity, and promoted CD4-driven CD8 activation, while anti-PD-1/CTLA-4 regimen directly activated more cytotoxic CD8 cells.

Though no publicly cited studies directly use Elranatamab biosimilars with checkpoint inhibitor biosimilars, the anticipated experimental approach includes:

  • Co-administering Elranatamab biosimilar (activates T cells to kill tumor cells via BCMA/CD3 engagement) with a checkpoint inhibitor biosimilar (removes immunosuppressive brakes, unleashing further T cell responses)
  • Employing mouse tumor models or patient-derived xenografts to assess whether the dual engagement of adaptive immunity (by Elranatamab) and relief from immune checkpoints (by anti-CTLA-4/LAG-3) produces additive or synergistic anti-tumor effects.
  • Profiling immune cell responses (e.g., CD4/CD8/Treg subsets) using flow cytometry, single-cell RNA sequencing, and functional assays.
  • Measuring clinical endpoints such as objective response rate, progression-free survival, and duration of response.

Key context:

  • Researchers systematically test immunotherapy combinations because checkpoint inhibitors and T-cell engagers have complementary mechanisms: engagers provide tumor-specific T cell activation, while checkpoint inhibitors prevent tumor-induced immune exhaustion or suppression.
  • The specific contributions of CTLA-4 vs. LAG-3 blockade in synergy are experimentally determined by dissecting immune cell dynamics in response to treatment, as described in animal studies.

In summary, researchers use Elranatamab biosimilars and checkpoint inhibitor biosimilars in tandem in preclinical immune-oncology models to dissect the cellular basis of synergy, leveraging the unique immune activation signatures produced by each class and measuring anti-tumor efficacy through objective and mechanistic endpoints.

In a bridging ADA ELISA for immunogenicity testing, an Elranatamab biosimilar can be used as both the capture and detection reagent to monitor a patient's immune response—specifically the development of anti-drug antibodies (ADA) against Elranatamab.

The process works as follows:

  • Biotinylated Elranatamab biosimilar is coated onto streptavidin plates as the capture reagent.
  • Patient serum is added; if ADA (antibodies against Elranatamab) are present, their bivalent nature enables them to bind the biosimilar via one arm.
  • For detection, enzyme- or dye-labeled Elranatamab biosimilar is added, which can bind to the opposite arm of the ADA—forming a “bridge” between the two biosimilar molecules via the patient’s ADA.
  • The resulting complex is detected via the enzymatic or dye reaction, providing a quantitative measure of ADA in the sample.

Key Details:

  • Using a biosimilar ensures the reagent mimics the therapeutic Elranatamab's structure and epitope presentation, enabling sensitive and specific ADA detection.
  • The assay’s reliance on the bivalent binding of ADA (ability to simultaneously bind two antigen molecules) underpins its high specificity.
  • This method is widely used for immunogenicity assessments of therapeutic antibodies and bispecific antibodies, as is relevant for Elranatamab, a BCMA-CD3 bispecific used in multiple myeloma therapy.
  • Correct selection and characterization of biosimilar reagents is critical to minimize potential matrix interference and yield reliable results.

This format allows clinicians and researchers to monitor the immunogenic response to the therapeutic, which is crucial for assessing efficacy, safety, and the risk of side effects like loss of drug response or hypersensitivity reactions.

References & Citations

1. Mariuzza RA, Agnihotri P, Orban J. J Biol Chem. 295(4):914-925. 2020.
2. Pillarisetti K, Powers G, Luistro L, et al. Blood Adv. 4(18):4538-4549. 2020.
3. Bahlis NJ, Costello CL, Raje NS, et al. Nat Med. 2023;29(10):2570-2576.
4. Lesokhin AM, Tomasson MH, Arnulf B, et al. Nat Med. 2023;29(9):2259-2267.

Formats Available

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