GTOnco™ Redirected T Cell Cytotoxicity Assay Service

Introduction Overview Applications Strategy Mechanism Services Features Methods Platform FAQs

Currently, gene therapy-based I-O drugs that redirect cytotoxic T lymphocytes (CTLs) to kill tumor cells bearing a tumor-associated antigen have shown great promise in the clinic. For the purposes of monitoring redirected T-cell cytotoxicity induced by gene therapy-based I-O agents, Creative Biolabs has robustly developed in vitro platforms to provide reproducible cell-based cytotoxicity assays for our clients all over the world. We are able to design comprehensive research practices and handle high-quality in vivo studies to support your gene therapy-based I-O products development.

Introduction to Redirected T-Cell Cytotoxicity Assay

Redirected T-cell cytotoxicity assays are a crucial in vitro tool in the rapidly evolving field of immuno-oncology. This advanced functional assay is indispensable for evaluating the potency and efficacy of novel immunotherapies, particularly those designed to link T cells to target cells, such as bispecific antibodies (BsAb), trispecific antibodies (TsAb), and chimeric antigen receptor (CAR) T cells. By quantifying T-cell-mediated killing of specific target cells, this assay provides critical mechanistic and quantitative data, essential for advancing promising drug candidates from the laboratory to the clinic. It directly benefits drug developers and researchers, and through its support for basic research, makes a significant contribution to the scientific community and data repositories such as GEO (Gene Expression Comprehensive Database).

Figure 1: Illustrations of T-Cell Based Therapeutics in Clinical Development. (OA Literature)Figure 1. Examples of T-cell based therapeutics in clinical development.1

Overview of T Cell-Mediated Cytotoxicity

T cell-mediated cytotoxicity is a fundamental mechanism by which the adaptive immune system clears infected or malignant cells. Cytotoxic T lymphocytes (CTLs), primarily CD8+ T cells, recognize specific antigenic peptides presented on the surface of target cells via major histocompatibility complex (MHC) class I molecules. Upon recognition, CTLs are activated and rapidly induce apoptosis (programmed cell death) in target cells through two main pathways:

  • Perforin and Granase Pathway: CTLs release granules containing perforin and granase. Perforin forms a perforation in the target cell membrane, while granase enters the target cell and initiates a caspase cascade reaction.
  • Fas/FasL Pathway: The Fas ligand (FasL) on the surface of CTLs interacts with the Fas receptor on the target cell, triggering apoptosis in the target cell.

Why Are T-Cell Targeted Toxicity Assays Necessary?

Predictive and robust cytotoxicity assays are a crucial part of the immunotherapy development process. Here, they provide a fundamental understanding that allows for optimization of these therapeutics as well as translation to the clinic. With the rapid maturation of T cell redirecting technologies, there is a need to develop cytotoxicity assays that can accurately assess their mechanism of action and biology.

These assays are crucial for several key aspects:

Potential Efficacy Assessment

They quantitatively measure the ability of therapeutic drugs or engineered T cells to kill target cells (EC50 or IC50), which is essential for dose exploration and comparison between different constructs.

Mechanism of Action Validation

They validate whether bispecific antibody (BsAb) or chimeric antigen receptor (CAR) T cells are functional and whether they exert their effects through the intended T-cell targeting pathway.

Target Selection and Validation

They test candidate tumor-associated antigens (TAAs) and assess targeting specificity (targeted effect, off-target effect).

Translational Bridge

The results of these in vitro assays are prerequisites for submitting Investigational New Drug (IND) applications, providing fundamental evidence of the candidate drug's biological activity. (Gene Expression Comprehensive Database)

T Cell Redirection Strategy

Strategically directing the cytotoxicity of T cells towards malignant cells has become a cornerstone of modern cancer immunotherapy, with various technological platforms demonstrating clinical application value. The common goal of these methods is to bypass the traditional MHC-restricted antigen recognition system, endowing T cells with new specificity, thereby effectively reprogramming T cells to recognize tumor-associated antigens without considering the specificity of their own receptors.

Table 1: Comparison of Major T Cell Redirection Strategies

Strategy Mechanism of Action Key Advantages Clinical Stage
BiTEs Bispecific antibodies bridging T cells and tumor cells Transient engagement, no genetic modification required FDA-approved for some hematologic malignancies
CAR-T Cells Genetically engineered receptors with integrated signaling Persistent activity, memory formation Multiple FDA approvals for hematologic cancers
NOT Gates Paired activating and inhibitory receptors Enhanced specificity through logic gating Early clinical development

Scientific Mechanism of Redirected T Cell Cytotoxicity

Target cell clearance mediated by redirected T cell cytotoxicity is the result of a complex cascade of signaling and effector functions. At the most basic level, the central component of this process is the formation of a functional immunological synapse between the T cell and the target cell, which is initially triggered by cross-linking of connecting molecules between the two cells.

Molecular Basis of T Cell Activation

Upon binding to a bsAb or CAR, the CD3 complex transmits a signaling cascade from the immune receptor tyrosine activation motif (ITAM), which ultimately results in the release of intracellular calcium stores and activation of the influx of stored regulatory calcium ions into the cell. Calcium ion influx results in activation of calcineurin, which dephosphorylates NFAT transcription factors and induces expression of genes essential to T cell effector function.

Cytotoxicity Execution Pathways

Upon activation, cytotoxic T cells can clear target cells primarily via two mechanisms. The granule exocytosis pathway includes directed release of perforin and granzymes into target cells at the immunological synapse. Perforin then promotes entry of granzymes into the target cell cytoplasm; these serine proteases can then activate caspase cascades and other apoptotic pathways in the cytoplasm.

A Flexible Suite of Services at Creative Biolabs

As a CRO specializing in immunotherapy, many techniques have been used in our GTOnco™ platform to evaluate the redirected T cell cytotoxicity and therapeutic efficacy of the gene therapy-based I-O products. By utilizing appropriate animal models and approaches, we are able to monitor the interaction between T lymphocytes and tumor cells in vitro. Our in vitro imaging system including positron emission tomography (PET) and bioluminescence imaging (BLI) permits the visual analysis of redirected T cell cytotoxicity effect in animal models during the whole evaluation of I-O products. The T lymphocytes and tumor cells can be also qualitatively and quantitatively determined by flow cytometry to validate the therapeutic efficacy. In addition, we provide histologic analysis of animal tissue sections to test the redirected T cell cytotoxicity of I-O agents. With highly specific antibodies labeled with visible markers, the tumor cells and T lymphocytes are allowed for morphometric and visualization analysis.

Features of Our Services

  • Skillful technicians and extensive experience
  • Advanced techniques and custom assay service
  • Superior quality and timely feedback

Methods to Perform Redirected T Cell Cytotoxicity Assay

The Redirected T Cell Cytotoxicity Assay measures the capability of redirected T cells (engineered T cells or native T cells in the presence of a BsAb/TsAb) to kill target cells in vitro. There are multiple methods:

Method Principle Advantages Disadvantages
Lactate Dehydrogenase (LDH) Release Target cell lysis releases cytoplasmic LDH, which is measured enzymatically. Simple, high throughput, robust. Indirect measure, susceptible to background LDH.
Fluorescent/Luminescence Labeling Target cells are pre-labeled with a fluorescent (e.g., CFSE) or luminescent reporter (e.g., Cr51 historically, or non-radioactive alternatives like Eu3+ or ApoTox-Glo). Loss of signal or release of label indicates lysis. Highly sensitive, Eu3+ and ApoTox-Glo are non-radioactive. Cr51 is radioactive; labeling may affect cell viability.
Flow Cytometry (FACS) Target cells and effector cells are differentially labeled. Target cell death is measured by the loss of the target population or the uptake of viability dyes (e.g., PI, 7-AAD). Highly multiplexable (can assess T cell phenotype, activation, and target cell death simultaneously), most accurate for determining the E:T ratio. Lower throughput, requires skilled operator.
Real-Time Cell Analysis (RTCA) Measures cell adhesion and morphology changes (impedance) as an index of cell viability over time. Real-time kinetic killing curve, non-invasive. Sensitive to cell type and adhesion quality.

GTOnco™ Platform Advantages and Highlights

The GTOncoTM Platform is Creative Biolabs' integrated system for developing and validating oncology therapeutics. Its key advantages in the context of the Redirected T Cell Cytotoxicity Assay are:

  1. Biologically Relevant Assay: Uses freshly isolated, highly characterized human PBMCs and T cell subsets for an authentic immune environment in contrast to immortalized cell lines.
  2. Comprehensive Multiplexed Data Set: High content, multiplexed readout generated by advanced FACS technology allows for measurement of cytotoxicity, T cell activation, proliferation, and cytokine profile all in a single well to obtain the most data from limited samples.
  3. Standardized Protocol: Protocols are rigorously optimized and validated to ensure data is high quality, reproducible, and meets the standard needed to be accepted for regulatory submissions.
  4. Customizable Target Cell Engineering: Custom target cell lines can be engineered using CRISPR or lentiviral transduction to model on-target and off-target as needed.

Frequently Asked Questions

Q: What is an appropriate effector cell to target cell ratio for retargeted T cell cytotoxicity assays?

A: The optimal E: T ratio depends on the strength of the retargeted T cells and the susceptibility of the target cells to killing. As a general rule, we suggest first titrating a range of ratios (eg 25: 1 to 1:1) in the initial experiments to determine the best ratio to achieve detectable specific lysis while maintaining linearity. High E: T ratios may allow detection of suboptimal killing events, while low ratios may help define overly-potent effects that may saturate at high concentrations. Our team can advise on ratio selection for related projects and help determine the best ratio to achieve a broad dynamic range and optimal assay sensitivity.

Q: How to distinguish between specific retargeted cytotoxicity and non-specific activation?

A: Our assay design includes multiple negative controls to rule out the contribution of non-specific activation, including alone target cells (spontaneous release), alone effectors (background), and non-specific target cells lacking the cognate antigen. In addition, we included specificity-mismatched bispecific controls to measure any potential off-target effects. For CAR-T testing, we added untransduced T cells as an important control for antigen-specific lysis. The experimental design always factors in relevant controls to help differentiate specific redirected killing from background.

Q: Can your assays recapitulate the immunosuppressive tumor microenvironment?

A: Yes, we have developed our own TME mimicry models that include relevant soluble factors (TGF-β, PGE2, adenosine), low physiological oxygen tension (1-5% O2), and co-culture with suppressive cell populations (M2 macrophages, myeloid-derived suppressor cells). These more sophisticated models better mimic the hostile conditions encountered in solid tumors and are more predictive of in vitro efficacy. We can also tailor these systems to include patient-derived stromal elements or specific physiological barriers relevant to certain tumor types.

Q: How long does it typically take to complete a full cytotoxicity evaluation?

A: The standard project timeline is 4-8 weeks, depending on the assay complexity and number of conditions tested. A simple cytotoxicity screen based on flow cytometry can be completed within 2-3 weeks, while a more extensive evaluation including TME factors, longitudinal tracking, and multi-parameter analysis typically takes 6-8 weeks. Cell engineering services to construct stable effector cell lines or CRISPR-modified target cells will of course add additional time to the project. We provide a detailed project timeline at the outset and clearly define each milestone to ensure alignment with the R&D timeline.

Connect with Us Anytime!

At Creative Biolabs, we are committed to advancing the field of cancer immunotherapy through rigorous scientific evaluation of retargeted T-cell technologies. Our comprehensive cytotoxicity assessment platform provides crucial information to optimize candidate therapies and accelerate their translational application. We look forward to the opportunity to collaborate with you on innovative T-cell retargeting projects and contribute to their successful development. For any technical issue or products/services related question, please feel free to contact us.

Reference

  1. Strohl W R, Naso M. Bispecific T-cell redirection versus chimeric antigen receptor (CAR)-T cells as approaches to kill cancer cells. Antibodies, 2019, 8(3): 41. https://doi.org/10.3390/antib8030041 (Distributed under Open Access license CC BY 4.0, without modification.)

Online Inquiry

For research use only. Not intended for any clinical use.

This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.