Anti-Human CD3 (Teplizumab) – Fc Muted™

Anti-Human CD3 (Teplizumab) – Fc Muted™

Product No.: LT2105

- -
- -
Product No.LT2105
Clone
PRV-031
Target
CD3
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Teplizumab, CD3ε
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Human CD3
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 (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Additional Applications Reported In Literature ?
ELISA,
WB,
IP,
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 as the therapeutic antibody Teplizumab. This product is for research use only. Teplizumab activity is directed against CD3 expressed on mature T cells.
Background
Type I diabetes is a chronic autoimmune disease that destroys insulin-producing beta-cells in the islets of Langerhans, leading to a dependence on exogenous insulin for survival1. Teplizumab (TZIELD) is a humanized, anti-CD3ε IgG1κ monoclonal therapeutic that delays the onset of Stage 3 Type 1 diabetes1, 2. CD3ε plays an essential role in T cell development and is part of the T cell-receptor CD3-complex, which acts as an external signal transducer3. Defects in CD3ε cause immunodeficiency and have been linked to susceptibility to type I diabetes in women.

Teplizumab is an Fc receptor-nonbinding anti-CD3 antibody4 whose Fc region is mutated (L234A; L235A) to reduce effector functions2. When Teplizumab is administered by intravenous infusion once daily for 14 consecutive days, it reduces the loss of beta-cell function1. Teplizumab treatment modifies CD8+ T lymphocytes, which are thought to kill beta-cells, to display a partially exhausted phenotype associated with delayed disease progression1, 5. Teplizumab delays the median onset of Stage 3 Type 1 diabetes by 2 years compared to placebo1, 2. Additionally, the effects of treatment persist over time. The median years to diabetes diagnosis after Teplizumab treatment is ~ 5 years compared to ~ 2 years in the placebo-treated group6.

In November 2022, the United States Food and Drug Administration approved Teplizumab injection to delay the onset of Stage 3 Type 1 diabetes in adults and pediatric patients aged 8 years and older who have Stage 2 Type 1 diabetes7.
Antigen Distribution
CD3 is found on the surface of mature T cells.
Ligand/Receptor
Peptide antigen bound to MHC
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Immunology

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 Teplizumab biosimilars are used as analytical standards or reference controls in pharmacokinetic (PK) bridging ELISAs to enable precise measurement of drug concentrations in serum samples. Their role is central for method standardization and quantitation in biosimilar and reference product comparability studies.

Essential Context and Supporting Details:

  • Single Analytical Standard Usage:
    The optimal practice is to use a single analytical standard, often a research-grade biosimilar prepared at known concentrations, for quantifying both the biosimilar (test product) and the innovator/reference product (e.g., FDA-licensed Teplizumab) in serum samples. This approach:

    • Minimizes variability between assays.
    • Avoids "crossover" calibration issues in blinded clinical studies.
    • Ensures both products are measured on the same curve.
  • Preparation of Calibration Standards:
    Calibration standards are prepared by reconstituting the research-grade biosimilar Teplizumab in serum or assay buffer at predefined concentrations—these standards establish the calibration curve for the ELISA, spanning the expected concentration range found in patient samples. In published validation protocols, standards cover a wide range (e.g., 50–12800 ng/mL).

  • Reference Controls (Quality Controls, QCs):
    Reference controls are typically both biosimilar and innovator Teplizumab samples, spiked in serum at known concentrations chosen to validate assay performance at low, medium, and high levels.

  • Bioanalytical Comparability and Bridging:
    Before bridging PK data between biosimilar and reference product, the assay must be scientifically proven to be bioanalytically equivalent—meaning that the biosimilar standard enables accurate quantitation of both products without bias. This involves rigorous validation:

    • Accuracy and precision evaluation across multiple runs and operators.
    • Statistical analysis to confirm equivalence, typically using confidence intervals (e.g., within 0.8–1.25 ratio for measured concentrations).
  • Practical ELISA Workflow:

    • Prepare serial dilutions of Teplizumab biosimilar to establish the standard curve.
    • Spike defined concentrations of Teplizumab biosimilar (and reference) into pooled human serum to create QC samples.
    • Test patient serum samples in parallel, interpolate their concentrations using the biosimilar-generated standard curve.
  • Justification for Biosimilar Standard Choice:
    If bioanalytical comparability is met, the biosimilar is commonly chosen as the universal calibrator because it streamlines all future PK measurements and regulatory submissions.

Additional Relevant Information:

  • The methods described align with regulatory expectations for quantitative PK ligand binding assays (LBAs) in biosimilar development, including validation for accuracy, precision, specificity, and robustness.
  • Not all ELISA kits are validated for clinical or FDA-approved diagnostic use; some, including research-use-only kits, use biosimilar standards primarily for research and early development purposes.

Summary Table: Biosimilar Use in PK Bridging ELISA

AspectBiosimilar RoleReference Control Role
Standard CurvePrimary calibratorCan be used for comparison
Quality Control (QC)Validates assay rangeChecks assay accuracy
Bioanalytical BridgingEnsures comparabilityConfirms equivalence

In summary, research-grade Teplizumab biosimilars serve as calibration standards for generating reliable standard curves and as reference controls for QC in PK bridging ELISAs, provided analytical comparability to the reference drug is thoroughly demonstrated and validated.

Researchers often use two primary models to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) when administering a research-grade anti-CD3 antibody in vivo: syngeneic models and humanized mouse models.

Syngeneic Models

Syngeneic models involve using tumor cells derived from mice of a specific inbred strain, which are then implanted into genetically identical host mice. These models are critical for evaluating immunotherapies because they allow researchers to study the immune response in an immunocompetent host. For example, syngeneic models like the B16 melanoma model have been used to study bispecific antibodies targeting CD3 and a tumor antigen, demonstrating potent tumor growth inhibition and T cell activation.

Humanized Mouse Models

Humanized mouse models are typically used when studying human-specific interactions, such as with human CD3 antibodies. These models involve mice that have been engineered to express human genes or tissues, often for the study of human diseases like cancer. Humanized models supplemented with human T cells can be used to study the effects of bispecific anti-CD3 antibodies on tumor growth and the characterization of TILs. They are particularly useful for understanding how these antibodies work in a context closer to human biology.

Both models are essential for preclinical studies as they provide insights into how anti-CD3 antibodies interact with tumor cells and the immune system in vivo. However, syngeneic models are more commonly used for early-stage research on immunotherapies due to their ability to mimic an intact immune system, while humanized models offer a more direct approach to studying human-specific antibodies.

Researchers use Teplizumab biosimilars—anti-CD3 monoclonal antibodies—primarily to induce T cell modulation and tolerance, and they combine them with other checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars to study possible synergistic effects in complex immune-oncology models. This approach leverages the distinct but complementary mechanisms of each agent to overcome resistance and augment anti-tumor activity.

Key aspects of how these combinations are studied:

  • Distinct Mechanisms and Synergy Rationale:
    Checkpoint inhibitors like anti-CTLA-4 and anti-LAG-3 modulate T cell activation and immune suppression through different pathways. For example, anti-CTLA-4 mainly restores T cell activation and proliferation in lymph nodes, while anti-PD-1 and related agents prevent cytotoxic T cell exhaustion in the tumor microenvironment. Combining multiple checkpoint inhibitors is hypothesized to harness synergistic effects by concurrently targeting different immune regulatory barriers, which may increase overall anti-tumor efficacy.

  • Preclinical and Translational Models:
    Combination strategies are often first evaluated in mouse models of cancer. These models allow detailed monitoring of immune cell dynamics using techniques like flow cytometry and single-cell RNA sequencing. For example, in melanoma models, anti-PD-1/CTLA-4 and anti-PD-1/LAG-3 combinations show distinct requirements for CD4+ T cells and produce unique immune phenotypes:

    • The anti-PD-1/LAG-3 combo required CD4+ T cells, reduced Treg activity, and increased CD8+ activation via enhanced help from CD4+ cells.
    • The anti-PD-1/CTLA-4 combo mainly boosted CD8+ cytotoxic T cell numbers directly.
  • Teplizumab's T Cell Effects:
    Teplizumab induces persistent changes in antigen-specific CD4+ and CD8+ T cells, including activation followed by the development of an exhaustion or regulatory phenotype. Gene expression studies identify upregulation of pathways like PD-1 and CTLA-4 signaling in CD8+ T cells after teplizumab treatment, reflecting a shift towards an exhausted or tolerant state. This suggests combining teplizumab with checkpoint inhibitors that release these brakes (like anti-PD-1, anti-CTLA-4, or anti-LAG-3) might potentiate anti-tumor or anti-autoimmune responses by both inducing exhaustion/tolerance and liberating effector function when appropriate.

  • Study Designs:
    Researchers design studies to:

    • Administer teplizumab biosimilar with one or more checkpoint inhibitor biosimilars to tumor-bearing animals or in early phase clinical studies.
    • Assess tumor regression, survival, and changes in immune phenotype among T cell subsets.
    • Use omics technologies (single-cell RNA-seq, bulk gene expression, flow cytometry) to dissect how the different immune cell populations shift in response to these combinations.
  • Clinical Implications and Toxicity:
    The main rationale is to increase response rates and durability compared to monotherapy. However, these combinations also increase immune-related adverse events, which are closely monitored in both preclinical models and clinical translation.

Summary Table: Mechanisms in Combination Checkpoint Therapy

Agent/ClassPrincipal Target/EffectCombination Rationale
Teplizumab (anti-CD3)Induces T cell exhaustion/regulationModulate autoreactive/effector T cell responses, promote tolerance
Anti-CTLA-4Restores T cell priming in lymph nodesEnhance induction, proliferation of activated T cells
Anti-LAG-3Releases alternative inhibitory pathwayBoosts activation, mainly via CD4+ T cell help
Anti-PD-1/PD-L1Blocks peripheral T cell inhibitionPrevents exhaustion of tumor-infiltrating cytotoxic T cells

Currently, there are no published studies directly using teplizumab biosimilar in combination with anti-CTLA-4 or anti-LAG-3 in oncology models, but the mechanistic rationale and methodologies used in similar checkpoint-blockade combinations strongly inform future research directions. If you require more specific examples or technical protocols, these would often be found in preclinical immunotherapy research articles or ongoing clinical trial records.

Teplizumab biosimilar would be utilized in a bridging ADA ELISA as both the capture and detection reagent to create a sandwich-type assay that can specifically detect anti-drug antibodies (ADAs) formed against the therapeutic drug in patient samples.

Bridging ELISA Methodology for Teplizumab Biosimilar

In this assay format, biotinylated teplizumab biosimilar serves as the capture reagent by being immobilized onto streptavidin-coated microtiter plates. This creates a high-density surface of the drug molecule that can effectively capture any ADAs present in patient serum or plasma samples that are directed against teplizumab.

The detection component utilizes a labeled form of the same teplizumab biosimilar - typically conjugated with horseradish peroxidase (HRP) or a fluorescent dye. When bivalent anti-drug antibodies are present in the patient sample, they bind to the captured biotinylated teplizumab on the plate surface. The HRP-labeled teplizumab then binds to the other binding site of these bivalent ADAs, forming a "bridge" between the capture and detection reagents.

Detection and Quantification Process

The bound HRP-labeled teplizumab is detected using chromogenic substrates such as 3,3',5,5'-tetramethylbenzidine (TMB), which produces a measurable colorimetric signal proportional to the amount of ADAs present. This approach provides high sensitivity and allows for high-throughput screening of patient samples.

Advantages and Considerations

This bridging format offers several advantages for monitoring immune responses to teplizumab biosimilar therapy. The method can achieve very low detection limits and provides quantitative measurements of ADA levels over time. However, the specificity may be challenged by matrix components in human serum, soluble target molecules, or residual drug components that could interfere with the assay.

Clinical Monitoring Applications

The assay enables clinicians to track the development of neutralizing antibodies (NAbs) and assess their impact on therapeutic efficacy. Regular monitoring helps identify patients who may experience loss of response, hypersensitivity reactions, or other therapy-limiting side effects associated with ADA formation. This information is crucial for making informed decisions about continuing, modifying, or switching therapeutic regimens in patients receiving teplizumab biosimilar treatment.

References & Citations

1. Herold KC, Bundy BN, Long SA, et al. N Engl J Med. 381(7):603-613. 2019.
2. Kaplon H, Crescioli S, Chenoweth A, et al. MAbs. 15(1):2153410. 2023.
3. https://www.ncbi.nlm.nih.gov/gene/916
4. Herold KC, Hagopian W, Auger JA, et al. N Engl J Med. 346(22):1692-1698. 2002.
5. Long SA, Thorpe J, DeBerg HA, et al. Sci Immunol. 1(5):eaai7793. 2016.
6. Sims EK, Bundy BN, Stier K, et al. Sci Transl Med. 13(583):eabc8980. 2021.
7. https://www.fda.gov/news-events/press-announcements/fda-approves-first-drug-can-delay-onset-type-1-diabetes
8. Herold KC, Bluestone JA, Montag AG, et al. Diabetes. 41(3):385-391. 1992.
9. Herold KC, Gitelman SE, Ehlers MR, et al. Diabetes. 62(11):3766-3774. 2013.
Indirect Elisa Protocol
FA
Flow Cytometry
Immunoprecipitation Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

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