Anti-Human CD3 x BCMA (Teclistamab) – Fc Muted™

Anti-Human CD3 x BCMA (Teclistamab) – Fc Muted™

Product No.: C975

- -
- -
Product No.C975
Clone
JNJ-64007957
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 IgG4κ
Applications
Agonist
,
FA
,
FC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
BCMA-Fc recombinant protein
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 ?
Agonist,
FA,
FC
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 asthe therapeutic antibody Teclistamab. This product is for research use only. Teclistamab simultaneously binds CD3ε and B cellmaturation antigen (BCMA).
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.

Teclistamab is a humanized IgG4-proline, alanine, alanine (IgG4-PAA) DuoBody CD3xBCMA Bispecific T cell Engager (BiTE) antibody developed for treatment of MM2,3. Teclistamab treatment redirects CD3+ T cells to BCMA-expressing MM cells, leading to the secretion of perforin and certain granzymes from the cytotoxic T cells and ultimately inducing antibody-dependent cell cytotoxicity and tumor cell death of the BCMA-expressing B cells. The process is not specific and MHC I molecules on antigen presenting cells are not involved3. Teclistamab treatment also leads to the secretion of interferon-γ, TNF-α, IL-2, IL-6, IL-8, and IL-102 cytokines are induced2,3. Additionally, activity is increased by the γ-secretase inhibitor LY-4115752.

Teclistamab was generated by immunizing OmniRats (Open Monoclonal Technology) with BCMA-Fc recombinant protein and re-cloning hits on a relatively silent IgG4-PAA scaffold2. A controlled Fab-arm exchange of a BCMA antibody and a CD3 parental antibody derived from SP34 was then performed. The Fc region of Teclistamab contains S228P/L234A/L235A mutations to minimize its immunological effector functions.

Teclistamab has been approved for treatment of MM in patients who demonstrate disease progression despite treatment3.
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

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 Teclistamab biosimilars are commonly used as calibrators or reference controls in pharmacokinetic (PK) bridging ELISA assays to quantify Teclistamab levels in serum samples. These biosimilars are selected because they are structurally and functionally equivalent to the clinical product, enabling standardization and comparability when measuring drug concentrations across different study arms, including reference and biosimilar products.

Use as Calibration Standards:

  • Single calibrator approach: A single PK assay is validated to quantify both biosimilar and reference Teclistamab products, with the biosimilar serving as the analytical standard (calibrator) for the standard curve. This approach increases consistency, reduces variability, and eliminates the need to run multiple assays or perform cross-calibrations.
  • Standard preparation: The biosimilar standard is provided in a validated, lyophilized form. For example, in the KRIBIOLISA Teclistamab ELISA, the lyophilized biosimilar is reconstituted to a defined concentration (e.g., 2 μg/mL) and then serially diluted to generate a standard curve over a relevant concentration range for serum PK analysis.

Role in PK Bridging ELISA:

  • Analytical comparability: During assay validation, serum QC samples are prepared with both biosimilar and reference Teclistamab at multiple concentrations. Their quantification using the biosimilar calibration curve must show statistical bioanalytical equivalence, typically assessed by confidence intervals for response ratios within pre-set acceptance limits (often 0.8–1.25). This confirms the biosimilar is appropriate as a reference standard for both products.
  • Assay methodology: In a bridging (or sandwich) ELISA, microwells are coated with the Teclistamab target (e.g., recombinant BCMA). Serum samples, standards, and controls are added; Teclistamab binds the target. After washing, a secondary detection reagent (e.g., labeled CD3 protein or anti-human IgG) is used to quantify how much Teclistamab from samples or standards has bound, resulting in a signal proportional to concentration.

Quality controls:

  • Reference controls: Parallel to the calibration standards, QC samples derived from reference Teclistamab (or other validated biosimilar lots) are included at low, mid, and high concentrations to monitor assay performance throughout sample analysis.
  • Validation and consistency: The standard and control concentrations, together with run metrics, are rigorously documented and analyzed during assay validation and sample testing to ensure accurate, robust quantification in support of PK and bioequivalence studies.

Summary Table: Research-Grade Teclistamab Biosimilar Use in PK Bridging ELISA

FunctionDetail
Calibration StandardBiosimilar reconstituted, serially diluted to create the standard curve
Reference ControlsQC samples at defined concentrations, using biosimilar and reference molecules
Assay TypeBridging/sandwich ELISA with target-coated wells and labeled detection reagent
Analytical SharpeningStatistical demonstration of equivalence between biosimilar and reference
MatrixHuman serum or plasma

Key Point: Use of a research-grade Teclistamab biosimilar as the single calibrator ensures accurate, streamlined, and comparable PK quantification of both the biosimilar and reference antibody in serum, provided full analytical comparability is experimentally demonstrated and documented according to regulatory and scientific best practices.

The primary in vivo models used to study tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization when administering research-grade anti-CD3 x BCMA antibodies are xenograft models in immunodeficient mice with human T cell engraftment and syngeneic models in CD3-humanized mice with engineered tumors expressing human BCMA.

Key Model Types:

  • Xenograft (humanized) models:

    • NSG mice: Immunodeficient NSG (NOD SCID gamma) mice are injected with human multiple myeloma cell lines (e.g., MM1S-luciferase-positive), then reconstituted with human T cells. The anti-CD3 x BCMA antibody plus T cells are administered intravenously. This system allows for tumor growth inhibition studies and some TIL characterization but is limited by the lack of a fully functional endogenous immune system.
  • Syngeneic (humanized CD3) models:

    • CD3-humanized mice: These immunocompetent mice express human CD3 in place of murine CD3, permitting functional engagement by human-specific CD3 antibodies. Researchers implant murine tumor cell lines (e.g., B16 melanoma, MC38 colon carcinoma, EL4 thymoma) engineered to stably express human BCMA (e.g., B16/hBCMA, MC38/hBCMA, EL4/hBCMA) subcutaneously.
    • Anti-CD3 x BCMA bispecific antibodies are administered, alone or in combination with checkpoint blockade (e.g., anti-PD-1 antibody), to assess tumor volume inhibition and immune infiltration. This model enables high-resolution phenotyping of TILs under a more physiologically relevant, fully immunocompetent setting.

Essential Context:

  • Tumor growth inhibition is robustly observed in both xenograft and syngeneic humanized models after anti-CD3 x BCMA treatment.
  • TIL characterization is feasible in both models, but the CD3-humanized syngeneic system is superior for evaluating endogenous immune responses and TIL function within an intact, fully functional immune microenvironment. No syngeneic multiple myeloma mouse models fully recapitulate bone marrow conditions, so investigators use engineered surrogate tumors for BCMA expression.

Additional Relevant Information:

  • Combinatorial approaches: Combining anti-CD3 x BCMA antibodies with immune checkpoint inhibitors like anti-PD-1 further enhances antitumor efficacy and modulates TIL populations in the syngeneic model.
  • Limitations: Xenograft models with human T cell engraftment may not accurately reflect endogenous T cell metabolism and immune architecture. Syngeneic CD3-humanized mice with engineered human BCMA–expressing tumors provide better immunological fidelity for TIL studies but do not perfectly model multiple myeloma’s bone marrow environment.

Summary Table of Primary In Vivo Models

Model TypeHost Immune SystemTumor TypeT Cells SourceTIL CharacterizationKey References
Xenograft (NSG, humanized)ImmunodeficientHuman MM cell linesHuman (engrafted)Limited
Syngeneic (CD3-humanized)Immunocompetent (human CD3)Murine cell lines expressing human BCMAEndogenous (mouse)Extensive

Conclusion: The most widely used models for these studies are NSG xenografts with human T cell engraftment and syngeneic CD3-humanized immunocompetent mice bearing engineered tumors that express human BCMA. The latter offers the most comprehensive data on TIL phenotype and function.

Researchers studying synergistic effects in immune-oncology models use teclistamab biosimilars in combination with other checkpoint inhibitors—such as anti-CTLA-4 or anti-LAG-3 biosimilars—to engage distinct yet complementary immune pathways. This strategy is designed to enhance anti-tumor immunity beyond what each agent can achieve alone.

Experimental Approach:

  • Teclistamab is a bispecific antibody that brings CD3-positive T cells into close proximity with BCMA-expressing tumor cells, directly activating T cells to kill cancer cells regardless of their TCR specificity.
  • Checkpoint inhibitors (like anti-CTLA-4, anti-PD-1, or anti-LAG-3 agents) target pathways that suppress T cell function, essentially releasing "brakes" on immune responses. CTLA-4 acts mainly in lymph nodes and inhibits T cell priming, while LAG-3 and PD-1 are more involved at peripheral tumor sites in suppressing effector T cells.

Combination Rationale and Model Use:

  • By combining teclistamab biosimilars (which force direct T cell-tumor engagement) with checkpoint inhibitors (which lift inhibition at various immune checkpoints), researchers can test whether freeing T cells from inhibition amplifies the cytotoxic response triggered by teclistamab-induced synapse formation.
  • In preclinical studies and complex models (e.g., syngeneic or humanized mouse tumor models), these combinations help dissect:
    • The specific T cell subpopulations involved (such as distinguishing CD4 vs. CD8 T cell requirements).
    • The effect on regulatory T cells (Tregs) versus helper or cytotoxic T cells.
    • Whether combining checkpoint inhibitors with teclistamab reshapes the tumor microenvironment for greater anti-tumor activity, or conversely, triggers increased toxicity.

Mechanistic Insights:

  • Recent research with other checkpoint inhibitor combinations (e.g., anti-PD-1/CTLA-4 versus anti-PD-1/LAG-3) demonstrates that these synergistic effects can be highly context-dependent—such as requiring CD4 T cells for efficacy in some combinations but not others, or modulating Treg suppression differently.
  • The mechanistic synergy is attributed to teclistamab increasing T cell-mediated tumor cell lysis, while checkpoint blockade supports sustained T cell activation and proliferation by blocking inhibitory signals.

Challenges and Considerations:

  • While combining these agents can yield improved anti-tumor responses, there is also a heightened risk of immune-related adverse events due to broad immune activation.
  • Models must be carefully selected to reflect the complexity of human immune-tumor interactions for findings to be translatable.

In summary, researchers utilize teclistamab biosimilars with checkpoint inhibitors in immune-oncology models to evaluate if dual targeting both engages T cells more robustly and overcomes tumor-induced immune suppression, thereby revealing potential new combination therapies for cancer.

A Teclistamab biosimilar can be used as the capture and/or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response against Teclistamab therapy by taking advantage of its identical variable regions, ensuring it recognizes the same epitopes as the clinical drug.

In a bridging ADA ELISA, the basic workflow involves:

  • Coating or capturing: A form of Teclistamab (or its biosimilar with the same variable regions) is immobilized—this can be via biotinylation on streptavidin-coated plates or direct adsorption to the microplate surface. This mimics the therapeutic drug as the target for endogenous ADAs in patient serum.
  • Sample incubation: Patient serum, potentially containing anti-Teclistamab antibodies (ADAs), is incubated with the coated biosimilar. Any present ADAs will bind to this immobilized drug.
  • Detection reagent: A labeled (e.g., HRP-conjugated or biotinylated) Teclistamab biosimilar is added. This reagent binds the opposite arm of bivalent ADAs, bridging the coated and labeled Teclistamab biosimilars via the ADA, creating a "sandwich" or "bridge".
  • Signal generation: The amount of label bound is detected via a chromogenic substrate, correlating with ADA concentration.

Key points for Teclistamab biosimilar use:

  • Same variable regions: Biosimilars match the therapeutic antibody’s specificity, ensuring ADAs will bind the biosimilar as they would the drug.
  • Fc mutations: Teclistamab biosimilars often have Fc region modifications to minimize effector functions, which is irrelevant for the ELISA’s antigen-binding role but crucial for safety and functional studies.
  • Non-therapeutic, research-grade: Using a biosimilar avoids depleting clinical drug supply and provides a consistent reagent for assay reproducibility.

Why use a Teclistamab biosimilar in ADA ELISA?

  • It allows detection of humoral immune responses (ADA formation) specific to the therapeutic, which is critical for monitoring immunogenicity and guiding clinical decisions.
  • Bridging ELISAs using biosimilars have high sensitivity and specificity when reagent quality and assay conditions are optimized.

Summary Table: Role of Teclistamab Biosimilar in Bridging ADA ELISA

StepReagent UsedPurpose
Plate coating/captureTeclistamab biosimilarCaptures anti-Teclistamab antibodies (ADA)
Immune complex formationPatient serum (with possible ADA)ADA bridges between capture and detection drug
DetectionLabeled Teclistamab biosimilarBinds ADA, enabling detection

This approach is widely used for various monoclonal antibody therapies and is recommended for drugs like Teclistamab to assess the risk and impact of ADA development during treatment.

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 Kang C. Drugs. 82(16):1613-1619. 2022
4 Usmani SZ, Garfall AL, van de Donk NWCJ, et al. Lancet. 398(10301):665-674. 2021.
5 Moreau P, Garfall AL, van de Donk NWCJ, et al. N Engl J Med. 387(6):495-505. 2022.
6 Glatte B, Wenk K, Grahnert A, et al. Blood Adv. 7(15):3842-3845. 2023.
7 Miao X, Wu LS, Lin SXW, et al. Target Oncol. 18(5):667-684. 2023.
Agonist
FA
Flow Cytometry

Certificate of Analysis

Formats Available

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