Anti-Human CD3 x DLL3 (Tarlatamab) [Clone AMG 757]

Anti-Human CD3 x DLL3 (Tarlatamab) [Clone AMG 757]

Product No.: C3320

- -
- -
Product No.C3320
Clone
AMG 757
Target
CD3 x DLL3
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
CD3ε: T-cell surface antigen T3/Leu-4 epsilon chain, T3E DLL3: Delta-like protein 3, Drosophila Delta homolog 3 (Delta3)
Isotype
Human scFv
Applications
ELISA
,
FC
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
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 ?
ELISA,
FC,
WB
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 Tarlatamab. Tarlatamab targets CD3 on T-cells and DLL3 on tumor cells.
Background
Anti-Human CD3 x DLL3 is a bispecific T-cell engager (BiTE) designed to target CD3 on T- cells and Delta-like ligand 3 (DLL3) on tumor cells. DLL3 is highly expressed in small cell lung cancer (SCLC) and other neuroendocrine tumors, making it an attractive target for immunotherapy. By redirecting T-cells to attack DLL3-expressing tumor cells, this bispecific antibody offers a novel treatment approach for SCLC, a disease known for its poor prognosis and limited therapeutic options. Preclinical studies have demonstrated that DLL3/CD3 bispecific antibodies can induce potent, DLL3-dependent T-cell-mediated lysis of tumor cells, recruit T-cells into non-inflamed tumor tissues, and lead to tumor regression in animal models. While specific efficacy data for this antibody are not provided, BiTE antibodies have generally shown high potency, with some studies indicating efficacy at sub-picomolar concentrations.1-3.

AMG 757, also known as Tarlatamab, is a promising example of a bispecific T-cell engager targeting DLL3 and CD3 for SCLC treatment. Studies have shown that AMG 757 induces potent tumor lysis and T-cell activation, resulting in significant tumor regression and even complete responses in preclinical models. Clinical trials have reported manageable safety profiles with encouraging response durability, including objective response rates of 23.4% to 40% and disease control rates of 51.4%. Notably, AMG 757 has demonstrated antitumor activity with durable responses and promising survival outcomes in patients with previously treated SCLC, making it a viable option for targeting DLL3-expressing tumors4-6.
Antigen Distribution
The distribution of Anti-Human CD23 x DLL3 in the body includes the blood and lymphatic tissues, where it can effectively engage with both T-cells and tumor cells.
Ligand/Receptor
CD3ε: TCR
DLL3: Notch-1
NCBI Gene Bank ID
CD3ε: X03884
DLL3: AH009222
UniProt.org
CD3ε: P07766
DLL3: Q9NYJ7
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 Tarlatamab biosimilars are typically used as calibration standards or reference controls in PK bridging ELISA by serving as the analytical standard against which both biosimilar and reference product drug concentrations in serum samples are measured.

In PK bridging ELISA assays for biosimilars:

  • A single analytical standard is selected—usually the biosimilar product with established analytical comparability to the reference drug, here Tarlatamab.
  • The method is robustly qualified to confirm that the biosimilar and reference product behave similarly in the assay (demonstrating bioanalytical equivalence within predefined parameters, such as precision, accuracy, and equivalence interval).
  • Quality Control (QC) samples are prepared with both the biosimilar and the reference drug, and their performance within the assay is assessed relative to the standard curve generated from the biosimilar calibration standards.
  • The standard curve is constructed by spiking increasing concentrations of the biosimilar Tarlatamab into human serum to cover the assay’s intended dynamic range (nominal concentrations may range from low ng/mL to tens of μg/mL, e.g., 50–12800 ng/mL).

Key steps:

  • Confirm that the biosimilar and reference Tarlatamab are analytically equivalent in the ELISA, minimizing confounding variability by relying on a single standard.
  • Use the biosimilar standard to calibrate the assay, and then quantify serum sample concentrations based on their signal compared to this standard.
  • This approach enables direct comparison of drug concentrations for both reference and biosimilar products in clinical PK studies, supporting bioequivalence and regulatory requirements.

Essential context:

  • The use of a single biosimilar standard avoids variability inherent in multiple calibration curves and enables blinded PK studies, as recommended by regulatory and industry best practices.
  • Analytical method validation involves extensive robustness testing, often over multiple days, with independent analyst sets to ensure reliable performance across different lots and runs.
  • This paradigm ensures scientifically rigorous, unbiased determination of pharmacokinetic profiles for biosimilar and reference Tarlatamab in study serum samples.

The primary in vivo models used to evaluate research-grade anti-CD3 x DLL3 bispecific antibodies for tumor growth inhibition and analysis of tumor-infiltrating lymphocytes (TILs) are humanized mouse models—specifically, mice implanted with DLL3-expressing human tumor cell lines and reconstituted with human immune cells.

Key models include:

  • Subcutaneous xenograft models in CD3+ T-cell humanized mice:

    • SHP-77 (small cell lung cancer, SCLC) human tumor cells are implanted subcutaneously in immune-deficient mice, typically NSG strains. These mice are "humanized" by engraftment of human PBMCs (peripheral blood mononuclear cells) or T cells.
    • The DLL3/CD3 bispecific antibody is administered intravenously. Tumor growth is monitored, and tumor regression is evaluated; analysis of TILs is performed post-treatment. The bispecific induces tumor infiltration by human T cells and converts "cold" (noninflamed) tumors into "hot" (inflamed) states, facilitating analysis of TILs and cytokine responses.
  • PBMC-reconstituted B-NDG mice implanted with human tumor cell lines:

    • Solid tumor models, such as SHP-77, are used; B-NDG mice are engrafted with human PBMCs to allow human T-cell mediated responses. Administration of an anti-CD3 x DLL3 bispecific antibody or analog results in robust tumor growth inhibition and allows characterization of the immune infiltrate within the tumor.

Other relevant approaches:

  • Orthotopic and metastatic xenograft models: Some studies use orthotopic delivery (directly into the lung) or systemic (tail vein) injection of DLL3-positive tumor cells (e.g., SCLC cell line H82) into NSG or NSG-SGM3 mice reconstituted with human T cells, followed by treatment with DLL3-targeting bispecific antibodies or CAR T cells. These models support the analysis of tumor growth inhibition and immune cell dynamics in appropriate tissue contexts.

Traditional syngeneic (fully immunocompetent murine tumor in mouse) models are generally not employed for anti-CD3 x DLL3 studies. This is because bispecific antibodies that engage human CD3 require a humanized immune system for activity; murine T cells do not respond to human CD3-targeting antibodies.

Summary Table: Models for anti-CD3 x DLL3 Bispecific Antibody Studies

Model typeTumor type (cell line)Mouse strain & humanizationImmune contextTIL analysis possibleReference
Subcutaneous xenograft, SHP-77SCLC (human)NSG/CD3+ T cell humanizedHuman PBMC/T cellsYes
PBMC-reconstituted B-NDG xenograft, SHP-77SCLC (human)B-NDG, PBMC humanizedHuman PBMC/T cellsYes
Orthotopic/metastatic SCLC (e.g., H82 cells in NSG)SCLC (human)NSG/NSG-SGM3, T-cell humanizedHuman T cellsYes*

Note: TIL analysis feasibility depends on the study design and tissue harvest timing.

No evidence supports the routine use of fully syngeneic mouse models (with mouse DLL3 and native mouse CD3) for anti-CD3 x DLL3 bispecifics, since these would not recapitulate human antibody/antigen/CD3 biology.

For all of these models, tumor growth inhibition and detailed characterization of TILs—including T-cell infiltration, activation, cytokine production, and exhaustion—can be systematically evaluated.

Researchers use the Tarlatamab biosimilar, a bispecific T-cell engager targeting DLL3 (on tumor cells) and CD3 (on T cells), to directly redirect T-cell cytotoxicity toward DLL3-expressing cancer cells—commonly in small cell lung cancer—within preclinical immune-oncology models. To study synergistic effects with other checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars, they often combine Tarlatamab with these agents in vitro (cell cultures) and in vivo (mouse or humanized mouse) models to assess additive or synergistic immune responses.

Context and Supporting Details:

  • Experimental Design:

    • Tarlatamab biosimilar is available for both in vitro and in vivo research use, specifically for modeling immune-mediated tumor killing.
    • In combination studies, researchers co-administer Tarlatamab biosimilar with checkpoint inhibitors (e.g., anti-CTLA-4 or anti-LAG-3 biosimilars) and evaluate endpoints such as T-cell activation, cytokine production, tumor lysis, and overall survival in animal models.
  • Immunological Mechanisms:

    • Tarlatamab acts by bridging T cells and DLL3-positive tumor cells, triggering targeted cytotoxic T-cell activation and tumor cell apoptosis, even in immunosuppressive tumor environments where traditional checkpoint blockade is less effective.
    • Checkpoint inhibitors, like anti-CTLA-4 and anti-LAG-3, enhance antitumor immunity by removing inhibitory signals from T cells (releasing immune "brakes").
      • For example, anti-CTLA-4 promotes direct activation of cytotoxic CD8+ T cells.
      • Anti-LAG-3 therapy increases CD4+ helper T-cell activity while reducing regulatory T-cell suppression, indirectly boosting CD8+ cytotoxic responses.
  • Rationale for Combining Modalities:

    • DLL3-targeting BiTEs like Tarlatamab force immune engagement with cancer cells, but efficacy may be hampered by exhaustion or other immune suppression within the tumor microenvironment.
    • Checkpoint inhibitors relieve distinct immunosuppressive pathways, potentially amplifying Tarlatamab-induced T-cell cytotoxicity.
    • By combining these, researchers can determine if checkpoint blockade potentiates the BiTE-induced tumor killing and identify which immune cell subsets are essential for synergy.
  • Assessment of Synergy:

    • Commonly measured outcomes include enhanced T-cell activation (CD4+ and CD8+), greater tumor cell killing, delayed tumor growth, and increased survival compared to single-agent therapy.
    • Use of biosimilars allows for preclinical combination studies without the regulatory or access limitations associated with clinical-grade drugs.

Summary Table: Research Roles of Tarlatamab and Checkpoint Inhibitor Biosimilars

AgentMechanismKey Immune EffectsResearch Readout
Tarlatamab BiTEDLL3×CD3 targeting, T cell engagerRedirects T cells to kill DLL3+ tumor cellsTumor lysis, T-cell activation
Anti-CTLA-4 BiosimilarBlocks CTLA-4 to release CD8+ T cellsPromotes CD8+ T cytotoxicityExpanded cytotoxic T cells
Anti-LAG-3 BiosimilarBlocks LAG-3 to increase CD4+ T cell and reduce Treg inhibitionEnhances CD4+ T helper activity, decreases regulatory T cell suppressionIncreased CD4/CD8 activity

No direct preclinical study of "Tarlatamab plus anti-CTLA-4 or anti-LAG-3 biosimilar" is cited in these sources, but the underlying methodology is established by combining BiTEs with checkpoint inhibitors in complex immune-oncology models to measure potentially synergistic immune responses and antitumor outcomes. This approach enables translation to future clinical trial designs if synergy is established preclinically.

A Tarlatamab biosimilar is used as a capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA by leveraging its ability to bind to anti-Tarlatamab antibodies formed in patient serum, thus monitoring the patient’s immune response against the therapeutic drug.

Essential assay mechanism:

  • In a bridging ADA ELISA, the therapeutic biosimilar (Tarlatamab) is chemically labeled in two formats—commonly biotinylated for capture and enzyme/dye-labeled (e.g., HRP) for detection.
  • Capture reagent:
    • Biotinylated Tarlatamab biosimilar is immobilized on a streptavidin-coated plate.
    • Patient serum (potentially containing anti-Tarlatamab antibodies) is added; any ADA present forms a “bridge” by binding to both the immobilized (capture) and labeled (detection) Tarlatamab reagent, since the ADA recognizes two epitopes on the drug—typical of immunoglobulins.
  • Detection reagent:
    • The second (detection) Tarlatamab biosimilar molecule is conjugated to HRP or another reporter. It binds to the opposite paratope of the ADA, completing the bridge.
    • After incubation and washing, the presence of ADA is revealed by substrate conversion (e.g., TMB for peroxidase), measured as optical density.

Key details for immunogenicity monitoring:

  • Biosimilar Tarlatamab reagents ensure the assay measures immune response to the actual therapeutic (including relevant epitopes).
  • The ELISA is sensitive and allows screening of large sample volumes for immune responses against the drug itself.
  • The use of biosimilar reagents is crucial in clinical research where the originator drug may be limited or for manufacturing robust reagent supply. Both capture and detection reagents must retain native epitope conformation for ADA recognition.

Assay workflow summary:

  • Biotinylated Tarlatamab biosimilar coats the plate (capture reagent).
  • Patient serum is added; ADAs, if present, bind this coated drug.
  • HRP- or dye-labeled Tarlatamab biosimilar (detection reagent) is added, completing a bridge between two drug molecules via the ADA.
  • Wash steps remove unbound components; color development measures ADA binding quantitatively.

Additional notes:

  • Assay customization: Labeling protocols and blocking solutions are optimized to minimize matrix interference and maximize specificity in clinical samples.
  • A validated 3-tiered approach is often used (screening, confirmation, titer), with competitive inhibition by excess drug to confirm ADA specificity against Tarlatamab.

Summary Table: Bridging ADA ELISA using Tarlatamab biosimilar

ComponentRoleDescription
Tarlatamab biosimilar (biotin)Capture reagentImmobilizes ADA from patient serum on plate
Tarlatamab biosimilar (HRP/dye)Detection reagentBinds opposite paratope of ADA, enables detection
Patient serum (ADA)AnalyteContains anti-Tarlatamab antibodies (if formed)
Streptavidin-coated plateImmobilizationBinds biotinylated Tarlatamab
TMB/peroxidase substrateSignal developmentVisualizes ADA presence via enzyme activity

This approach enables sensitive and specific monitoring of immunogenicity to Tarlatamab by directly detecting ADA responses against the therapeutic drug in patient samples.

References & Citations

1. Lin S, Zhang Y, Yao J, et al. J Transl Med. 2024;22(1):766.
2. Hipp S, Voynov V, Drobits-Handl B, et al. Clin Cancer Res. 2020;26(19):5258-5268.
3. Dreier T, Lorenczewski G, Brandl C, et al. Int J Cancer. 2002;100(6):690-697.
4. Paz-Ares L, Champiat S, Lai WV, et al. J Clin Oncol. 2023;41(16):2893-2903.
5. Giffin MJ, Cooke K, Lobenhofer EK, et al. Clin Cancer Res. 2021;27(5):1526-1537.
6. Ahn MJ, Cho BC, Felip E, et al. N Engl J Med. 2023;389(22):2063-2075.
7. Tarlatamab (AMG-757) | anti-DLL3/CD3 BiTE antibody | MedChemExpress. MedchemExpress.com. Accessed September 30, 2024. https://www.medchemexpress.com/tarlatamab.html
Indirect Elisa Protocol
Flow Cytometry
General Western Blot Protocol

Certificate of Analysis

Formats Available

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