菜单
Telephone sharing button Contact Us linkedin sharing button LinkedIn wechat sharing button YouTube wechat sharing button Twitter mailbox sharing button ps_marketing@wuxibiologics.com
arrow_left sharing button
arrow_right sharing button
CRO Services

Drug Development Expertise Empowering Research Services for Biologics

Bispecific T Cell Engager Assays


Bispecific T Cell Engager (TCE) Assay Services for Binding & Functional Characterization

Bispecific T Cell Engagers (TCEs) can simultaneously bind to T cells and tumor-associated antigens (TAAs), redirecting T cell-mediated cytotoxicity toward cancer cells. At WuXi Biologics, we offer comprehensive in vitro characterization to assess their binding efficiency, functional activity, and potency for downstream development.

 

Our full suite of assays support the discovery of TCE-based therapeutics, including:

  • Dual binding affinity assessments via ELISA, FACS, and SPR
  • High-throughput reporter gene assays for TCE screening
  • Custom T cell killing and cytokine release assays for TCE characterization

Assays for bispecific T cell engagers (TCEs), including dual binding affinity, T cell killing, cytokine release, and high-throughput reporter gene assays.

Bispecific T Cell Engager Assay Details:

Service Item Description Request A Quote
Reporter Gene Assays (RGA)​

Bispecific T cell engager RGA

Fc effector function: ADCC, ADCP

Request A Quote
TCE Cytotoxicity Assays CellTiter-Glo, FACS
T/PBMC Cell Activation and Expansion​ (Proliferation Assays)

p-ELISA/HTRF/AlphaLISA

Activation biomarkers (FACS)

Cytokine release assay​

CFSE/BrdU/CellTiter-Glo

T Cell Exhaustion​ FACS
Regulatory T Cell Assay​s

In vitro expansion​

T cell activation-suppression assay

Cytokine/Chemokine Profiling ELISA/AlphaLISA/Elispot/CBA/MSD
Ex vivo Tumor and TIL Profiling Schedule a Free Consultation
Cell Cycle Analysis​

Schedule a Free Consultation

Apoptosis Assay​s

PI/AnnexinV staining (FACS)​

Caspase 3/7 activation (Luminescence)​

Case Study #1: Dual Binding Evaluation of Bispecific T Cell Engagers (TCEs) to CD3 and TAA for Lead Screening and Characterization

In this case study, a FACS-based binding assay was developed to evaluate the simultaneous dual binding of bispecific T cell engagers to CD3 and tumor-associated antigens (TAA).

Case study on FACS-based binding assay to evaluate dual binding of bispecific T cell engagers to CD3 and tumor-associated antigens (TAA) for lead screening and characterization.

Figure 1: (A) Dual binding assay workflow. (B) Results demonstrated that the CD3 × CD38 bispecific T cell engager effectively bound both CD3 and CD38, confirming its dual-targeting capability.

Case Study #2: Reporter Gene Assay (RGA) for High-Throughput Screening of T Cell Activation by Bispecific TCEs

In this case study, we developed a Reporter Gene Assay (RGA) to assess the mechanism of action (MOA) of T cell activation induced by CD3 bispecific T cell engager.

Case study on Reporter Gene Assay (RGA) to assess T cell activation mechanism of action induced by CD3 bispecific T cell engagers for high-throughput screening.

Figure 1: (A) Reporter Gene Assay (RGA) workflow. (B) Results showed that the CD3 × TAA bispecific T cell engager effectively activated T cell signaling in the presence of tumor cells.

Case Study #3: Bispecific T Cell Engagers (TCEs) for Redirected T Cell Killing and Cytokine Release

In this case study, we developed T cell killing and cytokine release assays to screen and characterize CD3 × TAA bispecific TCEs.

Case study on T cell killing and cytokine release assays to screen and characterize CD3 × TAA bispecific T cell engagers (TCEs) for redirected T cell killing and cytokine release.

Figure 1: (A) Mechanism of redirected cytotoxicity through T cell engagement and tumor antigen targeting. (B) Both TCE1 and TCE2 exhibited potent cytotoxicity against TAA-containing tumor cells compared to the isotype control. (C) TCE-induced dose-dependent IL-2 release in co-cultured tumor cells and PBMCs. (D) Both TCEs promoted IFN-γ release, with TCE1 exhibiting higher potency.

Case Study #4: T Cell Activation Assays for the Characterization of Bispecific TCEs

This case study evaluates the activation of CD4+ and CD8+ T cells by bispecific TCEs using FACS. Antibody 1 demonstrated superior potency in upregulating CD25 and CD69 activation markers compared to antibody 2, indicating stronger T cell activation in the presence of tumor cells.

Case study on T cell activation by bispecific T cell engagers using FACS, showing superior potency of Antibody 1 in upregulating CD25 and CD69 activation markers compared to Antibody 2.

Figure A: CD25 and CD69 expression on CD4+ and CD8+ T cells by the treatment of antibody 1 and antibody 2. Dose-response curves suggested stronger T cell activation by antibody 1.

Figure B: In the table, the lower EC50 values indicated that CD25 and CD69 were upregulated by Ab1 and Ab2 on CD4⁺ and CD8⁺ lymphocytes in the presence of tumor cells. Ab1 effectively activated T cells in the presence of tumor cells, with potency greater than that of Ab2.

Frequently Asked Questions for Bispecific T Cell Engager Assays

Q: Do you recommend any assays for B cell-targeting TCEs?

A: For B cell-targeting TCEs, in addition to TDCC, cytokine release, and activation marker assays, you may also consider:

  • T cell proliferation assays to assess sustained activation.

  • Fratricide assays to evaluate unintended T cell killing.

  • B cell subset TDCC assays to determine preferential targeting of specific B cell subpopulations.

Q: How do you judge dual binding affinity in TCEs? Should it fall between CD3 and TAA binding affinities?

A: At WuXi Biologics, we assess CD3 and TAA binding separately using SPR or BLI, and confirm dual engagement using flow-based or ELISA assays. Dual binding isn’t simply the average of the two affinities; it’s influenced by factors like avidity, antigen density, and spatial orientation.

Our general guidance:

  • CD3 affinity: Weaker binding (KD ~10–100 nM) may reduce off-tumor toxicity; stronger binding (KD <1 nM) may boost potency.

  • TAA affinity: Typically stronger (sub-nanomolar KD) to ensure tumor targeting.

  • Dual binding: Often falls between CD3 and TAA affinities, but it’s more of a functional readout than a hard rule.

Ultimately, we recommend testing a range of CD3/TAA affinities and letting functional assays (cytotoxicity, cytokine release, etc.) guide lead selection.

Q: Do in vitro cytokine profiling results translate to in vivo efficacy and safety

A: In vitro assays are valuable for early screening but are not perfect predictors of in vivo outcomes. They help flag overly inflammatory or inactive candidates, compare formats, and de-risk candidates before animal studies. They are often predictive for trends in T cell activation and CRS risk but not always aligned with in vivo studies, for example, some molecules appear toxic in vitro but are tolerable in vivo, and vice versa.

 

We complement in vitro assays with:

  • Cross-reactivity and PK/PD studies

  • Receptor occupancy analysis

  • In vivo efficacy/toxicology in relevant models

Q: How do you use dual binding affinity in decision-making? Do you prefer strong or weak binding overall?

Dual binding assays are more about qualitative confirmation than quantitative thresholds. We use them to check simultaneous CD3 and TAA binding and functional relevance in a dose-dependent manner.

What we look for:

  • No binding → investigate potential issues (e.g., misfolding, steric hindrance, suboptimal format)

  • Clear binding → proceed to functional testing (e.g., TDCC or cytokine release)

  • Very strong binding at low doses → may flag stickiness or non-specific activity

We don’t rank leads based on dual binding affinity alone. And decisions are driven by cytotoxicity, cytokine profiling, and in vivo studies.

Your Project. Our Expertise.