Anti-Human CD3 (Teplizumab)

Anti-Human CD3 (Teplizumab)

Product No.: LT2100

- -
- -
Product No.LT2100
Clone
PRV-031
Target
CD3
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Teplizumab, CD3ε, 876387-05-2
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
Active
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 calibration standards or reference controls in PK bridging ELISA by preparing a quantitative standard curve with known concentrations of the biosimilar, enabling accurate measurement of Teplizumab concentration in serum samples.

In a PK bridging ELISA for measuring Teplizumab in serum, the process involves the following steps:

  • Preparation of Calibration Standards: The research-grade Teplizumab biosimilar is reconstituted and serially diluted to obtain a range of known concentrations, typically in a matrix-matched diluent such as human serum or plasma. These standards are essential for generating a standard curve during the ELISA.

  • Assay Principle: The ELISA is usually a quantitative sandwich format. For Teplizumab, ELISA plates are pre-coated with human CD3 proteins, which specifically bind Teplizumab (as the analyte). Calibrators (biosimilar standards), as well as serum samples, compete for binding on the same plate under identical conditions.

  • Detection and Quantification: After the binding and several washing steps to remove unbound material, an enzyme-labeled secondary antibody is added (such as Anti-Human IgG kappa:HRP), followed by a chromogenic substrate. The colorimetric signal generated is proportional to the Teplizumab present, whether in standards or unknowns.

  • Calculation of Drug Concentration: The absorbance values from the standards are used to construct a standard curve. Serum samples are then interpolated against this curve to determine the Teplizumab concentration.

  • Bridging/Reference Control Role: To ensure assay comparability—especially for biosimilar and reference (originator) products—a single PK assay and standard (often the biosimilar) is preferred. This minimizes inter-assay variability and supports regulatory expectations for demonstrating pharmacokinetic comparability in biosimilar development. During method development and validation, both the biosimilar and reference may be run in parallel to demonstrate analytical equivalence within predefined acceptance criteria (e.g., 90% confidence interval within 0.8–1.25 for the ratio of measured concentrations between biosimilar and reference). Once equivalence is established, the biosimilar is used as the default calibrator.

Key Technical Notes:

  • Standards and controls should ideally be matrix-matched (in human serum/plasma) to account for possible matrix effects.
  • All standards and samples are typically run in duplicate for increased accuracy and to meet assay validation requirements.
  • Proper validation includes evaluating precision, accuracy, and robustness—with both biosimilar and reference materials—during the development phase; however, routine use in PK studies employs the validated single standard (commonly the biosimilar).

This approach is consistent with industry best practices and regulatory guidance for ligand binding assays used in biosimilar PK bridging studies.

The primary preclinical models where a research-grade anti-CD3 antibody is administered in vivo to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models and humanized mouse models.

Key details:

  • Syngeneic Mouse Models: These involve implanting murine tumors into immunocompetent mice (e.g., C57BL/6 mice with B16 melanoma cells). Administration of a murine-reactive anti-CD3 antibody (such as the classic 2C11 clone) or bispecific antibodies engaging murine CD3 allows robust investigation of T cell-mediated tumor growth inhibition and detailed characterization of murine TILs within the intact murine immune system. This approach enables the study of native TIL expansion, activation, and response to therapy, closely mimicking physiological immune dynamics.

  • Humanized Mouse Models: These models use immunodeficient mice (like NSG or NOG strains) engrafted with human immune cells (typically PBMCs or hematopoietic stem cells) and human tumor xenografts. Administration of a human-reactive anti-CD3 antibody, often as part of a bispecific antibody (e.g., anti-GPRC5D × CD3 or BiTE constructs), can drive human T cell-mediated anti-tumor responses, tumor growth inhibition, and the profiling of human TILs in the tumor microenvironment. This system provides translational relevance for human-specific immune-tumor interactions, though immune reconstitution and functional diversity may be less complete than fully immunocompetent models.

Model Comparison Table

Model TypeHost Immune SystemTumor SourceAnti-CD3 SpecificityTIL Analysis (Species)Typical Uses
SyngeneicMurine (fully intact)Murine tumor linesMurine anti-CD3Mouse TILsMechanistic studies, immune profiling, drug efficacy assessment
HumanizedHuman (PBMC or HSC grafted)Human tumor xenograftHuman anti-CD3Human TILsTranslational studies, human immunotherapy, bispecific antibodies
  • Patient-Derived Xenograft (PDX) models use immunodeficient mice and human tumors but lack a full human immune system, so anti-CD3 immunotherapy studies require introducing human immune cells as in humanized models.
  • Bispecific anti-CD3 antibodies (BiTEs) are commonly used in these settings due to their ability to redirect T cells to tumor antigens and have been evaluated both in murine and humanized models.

Summary of applications:

  • Syngeneic models: For mechanistic immunology and conventional anti-CD3 antibody studies.
  • Humanized models: For translational immunotherapy research using human-reactive anti-CD3 agents and detailed analysis of human TILs.

Both platforms are foundational for evaluating T cell-focused therapies, profiling TIL phenotypes, and characterizing tumor growth inhibition in vivo.

Currently, there is no specific information available about researchers using Teplizumab biosimilars in conjunction with other checkpoint inhibitors, such as anti-CTLA-4 or anti-LAG-3 biosimilars, to study synergistic effects in complex immune-oncology models. Teplizumab is primarily used in the context of type 1 diabetes prevention and involves modulating immune responses, particularly CD8+ T cell exhaustion and modulation of other immune cell subsets.

However, the concept of combining different immunotherapies, including checkpoint inhibitors like anti-CTLA-4 and anti-PD-1, is well-established in cancer research. These combinations aim to enhance antitumor responses by targeting multiple checkpoints involved in immune regulation. For example, combining anti-CTLA-4 and anti-PD-1 therapies has shown synergistic effects in advanced melanoma by targeting different aspects of T cell activation and suppression.

To study synergistic effects in complex immune-oncology models, researchers typically employ a combination of in vitro and in vivo studies:

  1. In Vitro Studies: These involve culturing immune cells and analyzing how different checkpoint inhibitors influence cell activation, proliferation, and cytokine production.
  2. In Vivo Models: Mouse models of cancer are commonly used to study the effects of checkpoint inhibitor combinations. These models allow researchers to assess how different combinations impact tumor growth and survival.
  3. Mechanistic Analyses: Techniques like single-cell RNA sequencing and flow cytometry are used to detail changes in immune cell populations and signaling pathways following treatment with different checkpoints inhibitors.

If a Teplizumab biosimilar were to be used in immune-oncology research, the approach would likely involve similar methodologies to explore its potential synergistic effects with other checkpoint inhibitors. This would require adapting Teplizumab's mechanism of modulating immune responses in diabetes to oncology contexts, where immune suppression involves different pathways.

Potential Research Directions

1. Mechanistic Understanding

  • Objective: Understand how Teplizumab's modulation of CD8+ T cell exhaustion and other immune cell subsets could complement anti-CTLA-4 or anti-LAG-3 therapies in cancer models.
  • Approach: Use techniques like scRNA-Seq and flow cytometry to analyze changes in immune cell phenotypes and functions after combination treatments.

2. Synergistic Effects

  • Objective: Evaluate whether combining Teplizumab with anti-CTLA-4 or anti-LAG-3 biosimilars enhances antitumor responses in cancer models.
  • Approach: Conduct in vivo studies using mouse models of cancer to assess tumor growth and survival in response to combination therapies.

3. Clinical Relevance

  • Objective: Investigate the potential clinical benefits of combining these therapies in human cancer patients.
  • Approach: Design clinical trials to evaluate the safety and efficacy of these combinations in patients with specific cancer types.

By following these approaches, researchers can explore the potential of combining Teplizumab biosimilars with other checkpoint inhibitors in immune-oncology, although such specific studies are not currently documented.

A Teplizumab biosimilar can be used as both the capture and detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient's immune response to the therapeutic drug by detecting ADAs that bind to Teplizumab.

In this bridging ELISA format:

  • Capture Reagent: The Teplizumab biosimilar is immobilized (often biotinylated and bound to a streptavidin-coated plate). This setup allows any ADAs present in the patient's serum to bind the biosimilar Teplizumab on the plate.
  • Detection Reagent: After incubation, a labeled version of Teplizumab biosimilar (such as HRP-conjugated or dye-labeled) is added. If ADAs specific to Teplizumab are present, they will bind both the plate-immobilized and the labeled detection Teplizumab, forming a “bridge” between the two drug molecules via the ADA.

Key steps in the assay:

  • Patient serum is incubated on the capture Teplizumab biosimilar, enabling any specific ADAs to bind.
  • After washing, labeled Teplizumab biosimilar is added, binding to the other arm of the ADA.
  • Following further washes, a substrate is introduced for signal detection, correlating to ADA quantity.

Why use a biosimilar?

  • A biosimilar Teplizumab is structurally and functionally equivalent to the reference drug, making it suitable for ADA detection since patient antibodies will recognize both biosimilar and originator forms equally.
  • Biosimilars are often used to avoid consuming the clinical reference drug and to ensure consistent assay reagents, especially when large-scale or high-throughput ADA monitoring is needed.

Clinical significance:

  • The measured ADA response helps monitor immunogenicity, assess therapeutic efficacy, and identify potential adverse reactions such as loss of response or hypersensitivity.
  • Comparative immunogenicity assessments have confirmed biosimilars produce results matching the reference product, supporting their use in ADA monitoring assays.

In summary, using a Teplizumab biosimilar as both capture and detection reagent in a bridging ADA ELISA enables sensitive and specific quantification of anti-Teplizumab antibodies in patient sera, thereby helping assess the immune response against the therapeutic drug.

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.