You are viewing the site in preview mode

Skip to main content

TCR-T cell therapy: current development approaches, preclinical evaluation, and perspectives on regulatory challenges

A Correction to this article was published on 26 November 2024

This article has been updated

Abstract

TCR-T cell therapy represents a promising advancement in adoptive immunotherapy for cancer treatment. Despite its potential, the development and preclinical testing of TCR-T cells face significant challenges. This review provides a structured overview of the key stages in preclinical testing, including in silico, in vitro, and in vivo methods, within the context of the sequential development of novel therapies. This review aimed to systematically outline the processes for evaluating TCR-T cells at each stage: from in silico approaches used to predict target antigens, assess cross-reactivity, and minimize off-target effects, to in vitro assays designed to measure cell functionality, cytotoxicity, and activation. Additionally, the review discusses the limitations of in vivo testing in animal models, particularly in accurately reflecting the human tumor microenvironment and immune responses. Performed analysis emphasizes the importance of these preclinical stages in the safe and effective development of TCR-T cell therapies. While current models provide valuable insights, we identify critical gaps, particularly in in vivo biodistribution and toxicity assessments, and propose the need for enhanced standardization and the development of more representative models. This structured approach aims to improve the predictability and safety of TCR-T cell therapy as it advances towards clinical application.

Introduction

Currently, innovative personalized approaches to immunotherapy are undergoing significant development. One such approach, adoptive T-cell therapy, which involves the infusion of engineered T cells into patients, has led to breakthroughs in the treatment of certain cancers [1, 2]. This therapy entails the genetic modification of primary human T cells to express tumor-specific receptors, either T-cell receptors (TCR) or chimeric antigen receptors (CAR). These modifications enable T cells to recognize and destroy malignant cells [3]. While several CAR T-cell therapies have been approved by the Food and Drug Administration (FDA) for hematologic cancers, their effectiveness against solid tumors remains limited [4]. TCR-T cell therapy, although not yet approved for clinical use, is currently being evaluated in numerous preclinical and pilot trials targeting various antigens [4]. TCR-T cells are considered a more promising therapeutic approach for the treatment of solid tumors due to several key advantages. TCR-T cells are more sensitive to antigen levels in tumor cells compared to CAR-T cells and have the ability to recognize not only surface antigens but also intracellular antigens, making them applicable to a broader spectrum of tumors [5]. Moreover, TCR-T cells may demonstrate greater efficacy in the complex microenvironment of solid tumors, as they can interact with MHC-presented antigens, facilitating deeper tumor infiltration and enhanced activity within the tumor tissue. In addition, TCR-T cell therapy is associated with a potentially lower incidence of adverse effects, such as cytokine release syndrome, when compared to CAR-T therapy [6].

The main limitation of TCR-T cell therapy, when compared to CAR-T therapy, is the requirement for antigen recognition through major histocompatibility complex (MHC) molecules. Consequently, the effectiveness of TCR-T cell therapy is dependent on the patient’s specific human leukocyte antigen (HLA) profile, necessitating individualized cell modifications based on HLA compatibility. This poses a challenge for the widespread application of TCR-T cells, unlike CAR-T cells, for which standardized “off-the-shelf” options are being developed [6]. However, recent efforts have begun to develop pluripotent TCR-T cells capable of interacting with multiple HLA alleles [7].

Tumors can also develop resistance to TCR-T cell therapy by downregulating or losing HLA expression, which impairs antigen presentation and makes the tumor invisible to TCR-T cells. This phenomenon increases the risk of relapse and diminishes the therapeutic efficacy [8]. Additionally, successful antigen recognition by TCR-T cells requires proper antigen processing and presentation on the surface of tumor cells in complex with HLA. If this processing mechanism is disrupted, the effectiveness of the therapy may be significantly reduced [9, 10].

Before clinical trials can be approved and marketing authorization granted for T-cell-based medicinal products, preclinical trials must be conducted [11]. The goal of these preclinical trials is to evaluate the pharmacological, kinetic, and toxicological properties of the novel drug to predict its efficacy and safety in clinical practice. Unlike pharmaceuticals, there is currently no standardized approach for testing T-cell therapies. Therefore, preclinical study planning must consider potential risks identified in clinical trials of similar cell-based products [12].

Past safety issues associated with TCR-T cell therapy, particularly those related to unexpected "on-target, off-tumor toxicity," have resulted in severe adverse effects and fatalities in clinical trials. This phenomenon occurs because TCR-T cells may attack not only tumor cells expressing the target antigen but also healthy tissues that express the same antigen, even in minimal amounts. One of the most well-known examples involves TCR-T cells targeting the MAGE-A3 antigen (melanoma-associated antigen). In clinical trials, patients developed cardiotoxicity, which led to fatal outcomes [13, 14]. The cause of this toxicity was the cross-reactivity of TCR-T cells with titin, a protein expressed in cardiac muscle.

Another example of unexpected toxicity involves TCR-T cells targeting the MART-1 antigen, a melanoma antigen. In some clinical trials, patients developed autoimmune reactions against melanocytes, leading to vitiligo and other skin-related adverse effects [15, 16]. These reactions were caused by the cross-reactivity of TCR-T cells with normal tissues expressing MART-1.

In another clinical trial, patients receiving TCR-T cells engineered to target CEA (carcinoembryonic antigen) developed inflammatory colitis, which required systemic immunosuppression with high-dose steroids and anti-cytokine antibodies [17].

These examples underscore the importance of more stringent preclinical testing and thorough evaluation of potential cross-reactivity between tumor and normal tissues before initiating clinical trials.

To facilitate the planning of preclinical trials, we have consolidated and systematized data from existing protocols of T-cell-based drug studies available in the literature. Our analysis includes information from current international guidelines applicable to adoptive cell therapy, such as Fumagalli et al. [12]; the Guideline on the Non-Clinical Studies Required Before First Clinical Use of Gene Therapy Medicinal Products, 2019 [18]; the Guideline on Quality, Non-Clinical and Clinical Aspects of Medicinal Products Containing Genetically Modified Cells, 2019 [19]; and the Guideline on Quality, Non-Clinical, and Clinical Requirements for Investigational Advanced Therapy Medicinal Products in Clinical Trials, 2019 [20]; as well as the Preclinical Assessment of Investigational Cellular and Gene Therapy Products: Guidance for Industry, 2019 [21]. Additionally, we considered specific features of Russian legislation, including the Order of the Ministry of Health of the Russian Federation dated August 8, 2018, No. 512n “On the Approval of the Rules of Good Practice for Working with Biomedical Cellular Products” [22], and Federal Law dated June 23, 2016, No. 180-FZ “On Biomedical Cellular Products” [23].

This study aims to provide a summarized universal protocol for the key stages of TCR-T cell development and approval, while also addressing the existing challenges and issues in the field.

The key stages of the development and approval of TCR-T cell therapy

Figure 1 presents a generalized flowchart outlining the development of TCR-T cells and the conduct of their preclinical trials. This flowchart considers both the complex nature of these therapies and the limitations of currently available experimental models.

Fig. 1
figure 1

Flowchart of TCR-T cell manufacturing and preclinical trials. Stages that can be conducted using various in silico methods are marked with asterisks. These methods are typically employed during the “Development of Production Technology” stage. AG: antigen, TCR: T-cell receptor

This review focuses on the critical stages of TCR-T cell preclinical trials: “Development of Production Technology,” “Efficiency Assessment,” and “Safety Assessment,” each of which consists of multiple steps. While these stages align with the general phases of preclinical trials in drug development, TCR-T cell therapy presents unique challenges due to the technological and functional characteristics of autologous bioengineered cell-based products. A key challenge is the difficulty in selecting suitable animal models due to the infusion of human T cells, which necessitates a much broader use of in silico approaches compared to traditional pharmaceuticals. These approaches are employed not only at the initial stages of design and development but also in later phases of research to predict mechanisms of action and potential side effects. Previous studies have extensively described the efficacy tests used [2] and the safety tests [11] along with their advantages and limitations. However, our work is the first to organize these tests according to the stages of the technological production chain.

Development of production technology

The development of production technology encompasses all stages, from selecting a relevant antigen (AG) for TCR design and the corresponding genetic construct, to evaluating the efficiency of transducing selected cells with the synthesized construct (Fig. 2).

Fig. 2
figure 2

Flowchart of the “Development of Production Technology” stage. AG: antigen; TCR: T-cell receptor

Choosing an appropriate tumor antigen

Antigen selection is a critical factor in developing safe and effective adoptive cell therapy. TCRs recognize target antigens that are bound by major histocompatibility complex (MHC) molecules, which is a key advantage over CAR-T cells, as it allows for the detection of both surface and intracellular antigens [1]. Consequently, a wide range of antigens can be targeted in TCR-T cell therapy, though there are certain limitations related to how these antigens are presented on the cell surface. First, antigens with peptide epitopes identical to those recognized by TCR-T cells may also be expressed in healthy human tissues. Second, peptide epitopes in healthy tissues may structurally resemble target peptides sufficiently to be recognized by engineered TCR-T cells. Third, different MHC molecules may present off-target antigen peptides on healthy cells, leading to cross-reactivity with the engineered receptor [24]. Therefore, while an ideal target for TCR-T cells would be an MHC-peptide complex that is highly expressed and specific to all tumor cells, finding such an antigen is often challenging in practice.

Current clinical trials focus on two major classes of tumor antigens: tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs) [25].

Tumor-associated antigens (TAAs) are common antigens expressed across various healthy tissues but are overexpressed in tumors. Examples include HER2 [26] and mesothelin [27]. TAAs can be further classified into differentiation antigens and cancer germline antigens. Differentiation antigens are shared by tumors and their tissue of origin, such as MART-1 in melanoma [28]. Cancer germline antigens, whose expression is restricted to immune-privileged organs like the testes and placenta, include antigens such as MAGEs [29] and NY-ESO-1 [30]. Since these organs do not express MHC molecules, these antigens are not presented during spermatogenesis and are absent in healthy tissues [31].

In CAR-T cell therapy, the CD19 antigen, which is expressed on B cells, has become one of the primary targets for the treatment of B-cell lymphomas and leukemias [32, 33]. The depletion of B cells leads to B-cell aplasia, but this effect can be managed through immunoglobulin replacement therapy. Another example is BCMA (B-cell maturation antigen), which is expressed on mature B cells and plasma cells. BCMA is a target for CAR-T cells in the treatment of multiple myeloma [34]. Since BCMA is primarily expressed on plasma cells, targeting it does not result in severe side effects, similar to CD19.

TAAs are commonly found across different patients, making them promising targets for antitumor therapy [35]. However, because they are also expressed in normal tissues, albeit at low levels, the use of engineered receptors targeting these antigens may lead to on-target, off-tumor toxicity. Additionally, high-affinity T cells targeting these antigens might be eliminated during negative selection in the thymus, making it difficult to obtain highly active TCRs [1].

Tumor-specific antigens, or neoantigens, are expressed exclusively by tumor cells and are absent in healthy tissues. For instance, mutation-derived antigens represent unique antigens that arise from specific point mutations and are expressed exclusively in tumor tissue [36]. Such “shared neoantigens,” like KRASG12D neoantigens [37] or PIK3CA [38], may be common among different patients with the same HLA allele [1]. TCR-T cells targeting these shared neoantigens are currently undergoing clinical trials [1]. Viral-derived neoantigens, like E6 and E7, which are expressed in human papillomavirus-associated cancers, are also noteworthy [39]. While the application of such antigens is restricted to certain types of cancer, viral antigens exhibit tumor specificity and are non-toxic to normal tissues [1]. Moreover, a single viral antigen can only be used to treat patients with a specific combination of virus-induced cancer and HLA type [5]. Alternative TSAs are highly specific tumor antigens derived from genomic sources other than nonsynonymous single nucleotide polymorphisms, such as frameshift mutation-derived antigens, splice variants, gene fusions, endogenous retroelements, etc. [40] (e.g., HERV-E in renal cell cancer [41]). These antigens are fairly common in multiple tumor types.

Neoantigen-based therapy, however, has several limitations. High-affinity T cells specific to TSAs are not eliminated during negative selection in the thymus and can be isolated from the patient’s tumor or from the peripheral blood of a healthy donor [43]. However, the number of neoantigens in a tumor is relatively limited and can vary among patients, making it difficult to identify appropriate targets for each individual and potentially reducing the efficacy of TCR-T cell therapy. Another critical limitation in the use of TSAs for TCR-T cell construction is the risk of cross-reactivity. To prevent this type of toxicity, in silico studies are required to exclude cross-reactivity between TCRs and extratumoral epitopes, including screening for potential amino acid and single nucleotide substitutions [1, 35].

One of the key approaches to overcoming the challenges of targeting neoantigens is the development of TCRs directed at unique antigens that are exclusively present in the tumor cells of a specific patient [44]. Currently, research teams are developing personalized TCRs that account for specific mutations in a patient’s tumor, increasing the likelihood of successfully eradicating the tumor without damaging normal tissues [45].

Accurately predicting target antigens is essential for developing TCR-based immunotherapy [35]. Today, in addition to selecting suitable antigens based on literature and clinical data, researchers can also utilize a variety of in silico methods. These tools help identify potential antigens or epitopes that could be effective targets for TCRs [42]. This process considers several factors, including the MHC's ability to present the antigen, its interaction with MHC molecules, and its specificity for tumor cells. Antigen prediction tools use algorithms to analyze the molecular characteristics of antigens and their association with MHC molecules [46]. This allows for the identification of antigens that can effectively present themselves on the cell surface and bind to TCRs. At the same time, identifying antigens with high specificity for tumor cells is crucial for minimizing adverse events and maximizing the efficacy of immunotherapy [1].

In summary, the key factors for successfully engineering TCRs for therapeutic applications include the choice of target antigen, the efficiency of antigen presentation by target tissue cells, the relative levels of antigen expression in tumor cells compared to healthy cells, and the appropriate HLA allele restriction.

Designing tcrs

The design of T-cell receptors (TCRs) in biotechnology relies heavily on various algorithms for sequence prediction, which play a crucial role in the development of biomedical engineering products. These algorithms help identify potential TCRs capable of recognizing specific antigens or epitopes, as well as analyze similarities among them [47].

At the early stages of TCR technology development, various methods were widely employed to identify specific receptors capable of recognizing tumor antigens. The main approaches used in this process included: the incubation of peripheral blood mononuclear cells (PBMC) or tumor-infiltrating lymphocytes (TIL) from cancer patients with tumor antigens [48], the use of MHC multimers incubated with T cells to select those that interact with target antigens [49], vaccinating mice with human tumor antigens to isolate high-avidity T cells capable of reacting to human tumors [50], and the use of hybridomas created by fusing T cells with tumor cells to generate stable T cell lines expressing specific receptors [51]. Additionally, PBMCs from healthy donors were incubated with tumor antigens to identify TCRs capable of recognizing tumors [52]. Each of these methods has played a crucial role in identifying specific TCRs that can be used in cancer immunotherapy. The advancement of these technologies has improved the process of identifying and selecting TCRs, enhancing their precision and effectiveness.

The widespread use of protein crystallization techniques has led to the accumulation of extensive structural data on atomic positions, bond lengths, dihedral angles, and more, including for T-cell receptors [53]. This structural information about TCRs, MHC–peptide complexes, and antigens facilitates the design of TCRs with improved binding and specificity, often utilizing methods such as molecular modeling and molecular dynamics [54]. Molecular modeling techniques, including molecular docking, can predict interactions between TCRs and MHC–peptide complexes, enabling predictions of TCR binding affinity and specificity to target antigens, as well as simulations of MHC–antigen complex structures [55, 56].

Various methods are employed to forecast the immunogenicity of engineered TCRs and antigens, helping to predict possible immune reactions and assess the safety of therapies [57]. Computer modeling is also used to predict the pharmacokinetic and pharmacodynamic characteristics of TCR-T cell therapy, optimizing dosage regimens and treatment strategies [58]. In silico tools can identify potential external targets of engineered TCRs, thereby minimizing adverse reactions. Machine learning and predictive modeling are further applied to anticipate and mitigate potential adverse events related to TCR-T therapy [11]. Additionally, various models assist in patient selection and the personalization of TCR-T therapy by predicting which patients are most likely to respond positively to treatment [59].

Several -omics technologies are used to obtain TCR sequences. One such method, repertoire sequencing (AIRR-seq) [60], is employed for systematic analysis of TCRs, allowing for the determination of their sequences and diversity. Single-cell sequencing technology is another method being actively developed, providing more precise data on receptor distribution in immune cells through both DNA and RNA analysis. This technology allows for the identification of specific alpha and beta chains that form TCRs, significantly enhancing analysis resolution [61].

Other modern tools and methods, such as RNA sequencing (e.g., Nanostring) and secretome analysis (e.g., Polyfunctionality with Isoplexis), are also utilized to study T-cell receptors and related processes in research and clinical practice. RNA sequencing provides insights into the T-cell transcriptome, helping to identify the diversity of TCRs and study their expression [62]. This method allows for a deeper understanding of T-cell dynamics and functionality in response to various stimuli and pathological conditions. The Nanostring technology, an innovative approach for analyzing gene transcriptional activity and protein translation, helps identify signatures and molecular markers specific to different T-cell subpopulations, and characterizes their functional activity [63]. The Polyfunctionality method, particularly using Isoplexis, enables the analysis of the secretome of individual cells, allowing simultaneous assessment of multiple T-cell functions, including their effector functions and cytokine production capabilities [64]. This is essential for better understanding the functional characteristics of T cells and their potential in immunotherapy. Additionally, 3D RNA sequencing is a relatively new technique that allows for the analysis of gene expression within the context of the cellular architecture of tissues [65], making it valuable for studying TCR interactions with surrounding cells and their microenvironment.

Advancements in these molecular genetic technologies provide researchers and medical professionals with a deeper and more detailed understanding of T-cell functionality and their roles in the immune system and various pathologies. These tools are contributing to improvements in diagnosis and the development of personalized TCR-based therapies, thereby opening new avenues in immunotherapy and biomedical research.

Tcrs production

The design and synthesis of genetic constructs are not always performed within a single institution. Design is primarily an R&D task and is often carried out by research departments, while the synthesis of constructs is frequently outsourced to commercial companies [66,67,68]. This process is followed by T-cell transfection, a technique used to insert genetic information into T cells to enhance their antitumor potential.

Numerous technological platforms have been developed for the genetic modification of primary T cells, including non-viral platforms such as transposon-based systems (PiggyBac, Sleeping Beauty) [69], TALEN [70], CRISPR-Cas9 [71], electroporation [72], and gene delivery technologies utilizing chemical compounds like polymers and lipids [73]. The production of viral vectors encoding TCRs remains the most commonly used method in both preclinical and clinical settings [74]. Despite the existing risk of random gene integration into critical regions of the genome, viral vectors provide significantly higher gene delivery efficiency compared to non-viral transduction methods [5]. They are capable of integrating genetic material into the host cell genome, ensuring long-term expression [75].

Viral vectors, such as retroviruses and lentiviruses, integrate genetic material into the genome of T cells to deliver the TCR gene [76,77,78]. Given that transduction using viral vectors is currently the most popular method for genetically modifying primary T cells, this review will focus on assessing the efficiency of this specific method.

Assessing the efficiency of TCR-T cell transduction

The efficiency of TCR-T cell transduction is a crucial parameter, as it directly impacts the number of genetically modified cells capable of recognizing and destroying tumor cells. High transduction efficiency ensures a sufficient number of functional TCR-T cells to achieve a therapeutic effect. Conversely, low efficiency may result in an insufficient number of active cells, reducing the overall efficacy of the therapy and the likelihood of achieving clinical remission. Moreover, evaluating transduction efficiency helps determine how well the technology is performing and what improvements can be made in the cell modification process [79].

The efficiency of genetic transduction can be assessed using various methods, including fluorescent labeling of marker genes, real-time PCR, monitoring of transgene expression levels, and analysis of transgene integration into the T-cell genome.

Fluorescent labeling of marker genes is an effective tool for evaluating the efficiency of TCR-T cell transduction [80, 81]. This method allows researchers to track and quantify successful transgene insertion and monitor the expression of markers in cells. Fluorescent labeling involves inserting a marker gene that codes for a fluorescent protein into engineered T cells. After successful transduction, these T cells can be examined under a microscope or by flow cytometry to determine the presence and intensity of fluorescence.

The design of fluorescently labeled tetrameric MHC–peptide complexes allows for direct visualization, quantification, and phenotypic characterization of antigen-specific T cells via flow cytometry [82]. MHC–peptide tetramers are complexes composed of four fluorophore-labeled MHC molecules, each of which binds to a specific peptide. Due to their tetrameric structure, these complexes can bind to up to three of the four monomeric MHC units, enhancing the avidity of their binding to targets [83]. Multimer binding is analyzed to verify the proper assembly and correct conformation of TCRs [1]. For example, Ishihara M. et al. used a retroviral vector encoding NY-ESO-1 157-165/HLA-A*02:01-specific TCR-α and TCR-β chains with increased affinity for interfering RNA constructs that specifically inhibit endogenous TCRs [52]. The efficiency of retroviral transduction was assessed using CD8 + and CD4 + T cells from healthy volunteers, demonstrating that 87.6% of CD8 + T cells and 89.0% of CD4 + T cells were tetramer positive.

The efficiency of TCR-T cell transduction can also be assessed through real-time polymerase chain reaction (PCR) [84]. This method allows for the quantification of the presence and expression levels of marker genes, which is crucial for monitoring and improving transduction and therapy effectiveness. Real-time PCR relies on DNA amplification using fluorophores, enabling real-time monitoring of the amount of PCR products [85]. When evaluating TCR-T cell transduction efficiency, the marker gene inserted alongside the TCR gene expresses a fluorescent protein, and its expression level is measured throughout the amplification process. Real-time PCR quantifies the level of transduced TCR-T cells and the expression level of the marker gene, which is essential for optimizing experimental conditions. Additionally, this method is used to monitor the stability of the marker in transduced T cells over extended periods.

Efficiency assessment

The “Efficiency Assessment” stage consists of two key steps: in vitro and in vivo proof of concept, aimed at comprehensively verifying the effectiveness of the T cell-based therapy under investigation (Fig. 3).

Fig. 3
figure 3

Flowchart of the “Effectiveness Assessment” stage. AG: antigen; TCR: T-cell receptor

In vitro proof of concept

Regardless of their origin, selected candidate TCR sequences must be tested to confirm their specificity and effectiveness [86]. Cell proliferation and cytotoxicity assays are used to determine whether the tested molecules affect cell proliferation and are capable of exhibiting direct cytotoxic effects.

The effectiveness of TCR-T cells largely depends on their ability to destroy target tumor cells and induce their lysis [87]. Cytotoxicity assays, where T cells are co-cultured with tumor cells or human cell lines, are the most common methods for assessing the antitumor activity of TCR-T cells in vitro [2].

There are numerous assays based on various functional characteristics of cells, including enzyme activity, cell membrane permeability, adhesion, ATP production, coenzyme production, nucleotide uptake activity, and cell proliferation [88]. To choose the optimal viability assay, one must carefully consider the cell type, culture conditions, and specific study objectives. Quantification of specific target cell lysis induced by modified T cells is typically performed using methods such as the chromium (51Cr) release assay, bioluminescence imaging (BLI) using luciferase [89], impedance-based cell analysis and flow cytometry [87]. These methods are often combined with approaches that measure the quantitative release of effector cytokines and/or degranulation markers, which are indicators of T-cell activation in response to specific signals from target cells, and they allow for the assessment of modified T cell functionality [2].

Cr-release assay

The 51Cr release assay, based on radioactive chromium labeling of cells and quantitative measurement of target cells with compromised plasma membranes, was first described over fifty years ago [90] and has become a standard for measuring cytotoxicity induced by T cells and NK cells [87]. In this method, target cells are labeled with radioactive chromium (51Cr) and incubated with effector cells at varying effector-to-target ratios. During effector-mediated killing, the target cells lose plasma membrane integrity, releasing 51Cr into the culture medium. The radioactivity measured in the supernatant using a gamma-ray scintillation counter is proportional to the number of target cells destroyed during a given period [87]. In the work by Ishihara M. and colleagues, cytotoxicity assays using the chromium release assay (Cr-release assay) demonstrated that NY-ESO-1 TCR-T cells effectively eliminated NY-ESO-1-positive melanoma cell lines in vitro [52].

Measurement of lactate dehydrogenase (LDH) activity

Another common method for assessing cytotoxicity is based on measuring the activity of cytoplasmic enzymes released by damaged cells. Lactate dehydrogenase (LDH), a stable cytoplasmic enzyme present in all cells, is rapidly released into the cell culture supernatant when the plasma membrane is damaged, and its activity can be easily quantified [91].

Bioluminescent imaging

Bioluminescence imaging (BLI) is another widely used technology for cytotoxicity assessment, which measures the activity of cytoplasmic enzymes released by damaged cells. BLI-based cytotoxicity assays utilize the luciferase reporter gene to measure the light signal (photons) emitted by target cells. This signal is produced exclusively by living cells expressing the luciferase transgene, so the cytotoxicity of effector cells can be assessed based on the decline in BLI signal intensity [92]. Mensali et al. demonstrated the cytotoxicity of electroporated TCR-T cells specific to the colorectal cancer cell line MSI + HLA-A2 + HCT 116 using a BLI-based assay [93].

Impedance analysis

Impedance-based cell analysis is an alternative to the 51Cr and BLI-based assays. This method uses microelectrodes embedded in the bottom of each well of a microtitration plate to measure the impedance of electric current between the electrodes during cell adhesion [87]. In the absence of cells, impedance (a measure of electrical resistance between electrodes) is near-zero; however, cell adhesion alters the impedance depending on the number, morphology, viability, and adhesion strength of each cell type [94]. Suspension cells or non-adherent cells typically do not significantly contribute to electrical impedance or cause much smaller changes compared to adherent cells. This principle is used to measure the cytolysis of adherent target cells by non-adherent effector cells such as T cells [95]. In the study by Jin and colleagues, the effector functions of E7 TCR T cells, including IFN-γ production and tumor cell lysis, were demonstrated in vitro. The ability of E7 TCR T cells to kill tumor cells was evaluated using a real-time impedance-based cytolysis assay [96].

Flow cytometry

Unlike the three methods mentioned above, flow cytometry analysis allows for the study and quantification of cytotoxicity within heterogeneous cell populations, offering a unique advantage when investigating the sensitivity of different cell types to target cells within a mixed population [97]. Flow cytometry distinguishes between target and effector cells based on properties such as cell size and granularity (as indicated by forward and side scatter, respectively), as well as specific staining with fluorescently labeled antibodies [87]. Cell death is typically assessed using fluorescent DNA intercalating agents such as propidium iodide or 7-aminoactinomycin D, which are preferentially absorbed by dead cells and exhibit spectral shifts when interacting with DNA [98].

Apoptosis stages can be identified by staining cells with annexin V, which specifically binds to phosphatidylserine expressed on the cell surface during apoptosis [99]. Standard methods involve assessing the cytotoxicity of engineered T cells against cell lines stained with cell-penetrating dyes, such as CFSE (5-(and-6)-carboxyfluorescein diacetate succinimidyl ester), which interact with intracellular free amines, followed by flow cytometry analysis [97, 100].

Therefore, flow cytometry enables simultaneous analysis of both target cell elimination and effector cell phenotyping, as well as the separation of heterogeneous cell populations [87].

In vivo proof of concept

The application of TCR-T cells in animal models is essential for verifying their viability, functionality, and persistence in vivo [1]. A significant challenge in these studies is the requirement to infuse human cells into animals.

Various mouse models are employed in experiments with engineered T cells, including humanized mouse models, syngeneic mouse models, and HLA class I transgenic mice [11]. However, most preclinical in vivo studies to date have utilized xenograft models derived from human cell lines, where tumor cells are grafted onto immunodeficient mice prior to the infusion of TCR-T cells [11].

For instance, in a study using a mouse sarcoma model resistant to immune checkpoint inhibition, Ishihara M. et al. investigated the preclinical efficacy of TCR-T cells in combination with a vaccine based on long peptide antigens (LPA) loaded into pullulan nanogel nanoparticles [52]. The LPA consisted of 38 amino acid residues containing three murine CD8 + T-cell epitopes, including 9 m. CMS5a murine fibrosarcoma cells were subcutaneously implanted into wild-type BALB/c mice. DUC18 CD8 + T cells were isolated from DUC18 transgenic mice expressing a murine TCR that recognized the 9 m epitope of the neoantigen, resulting in TCR-T cells capable of recognizing CMS5a tumors. The combination of the LPA-containing vaccine and DUC18 TCR-transgenic T cells successfully destroyed CMS5a sarcoma, whereas T cells alone were ineffective [52].

The in vivo ability of TCR-engineered T cells targeting E7 to kill cancer cells was also investigated using xenograft mouse models. The administration of TCR-engineered T cells targeting E7 led to complete regression of subcutaneous 4050 tumors and suppressed the growth of CaSki tumors in NSG mice [96].

Despite their utility, xenograft models have several limitations. These models do not accurately replicate components of the human tumor microenvironment, such as the presence of immunosuppressive cells. Additionally, engineered TCRs interact with peptide-HLA complexes, but major histocompatibility complex molecules are specific to humans. This species difference means that experiments using HLA-transgenic mice may not fully capture the complexity of human immune responses.

Given the similarities between the immune systems of humans and primates, primates are considered potential large animal models for better toxicity assessment of engineered T cells. However, challenges arise from the need to obtain autologous engineered T cells, and the natural occurrence of tumors in primates is relatively rare [11]. Nonetheless, primate models closely replicate the development of cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, which are among the most frequent and potentially fatal immune-related adverse events during engineered T cell therapy [101,102,103].

Spontaneous cancers in dogs share many similarities with human cancers in terms of clinical presentation, histological features, molecular profiles, and treatment response [104]. The biological similarities between cancer in dogs and humans also offer the possibility of testing engineered T cells in dogs with naturally occurring tumors [11]. However, there are limitations due to ethical considerations, such as the need to select a group of animals with relevant spontaneously occurring tumors, and the potential risk of anaphylactic reactions when injecting human biological products into dogs [105]. Nonetheless, studies are being conducted on CAR-T and TCR T-cell therapies for dogs, the results of which may serve as a model for the potential application of these T-cell therapies in humans [106, 107]. Conversely, the results of clinical trials in humans may contribute to the development of T-cell therapy in veterinary oncology [108].

The recent emergence of human ex vivo models, particularly organoids and organotypic systems, has the potential to elevate preclinical TCR-T cell research to a new level in the future, as these models can mimic human biology and provide personalized predictions of certain types of toxicity [11]. For example, van Amerongen et al. proposed using human-induced pluripotent stem cells, which can be differentiated into specific organoids of vital tissues, for toxicity studies [109]. Another study suggested using organoids derived from a patient's tumor tissue as a novel preclinical model [110]. This approach preserves key biological characteristics of the original tumor. Li et al. demonstrated that compared to lung cancer xenograft models, the success rate of culturing increases with patient-derived organoids, while the time and cost of creating the model are significantly reduced. These organoid models are also expected to be more personalized, offering better predictions of in vitro anticancer therapy efficacy. A recent review highlighted the potential of three-dimensional (3D) tumor models, such as spheroids from cervical cancer cell lines and patient-derived organoids, for evaluating new treatment modalities, particularly immunotherapy targeting tumor cells and modulating the tumor microenvironment [111]. Studies have already successfully used organoids to assess the cytotoxic effects of CAR-modified cells [112, 113].

In conclusion, the in vivo toxicity of adoptive T-cell therapy can currently be reliably assessed only in human clinical trials. Therefore, toxicity assessment primarily involves determining whether human tissue contains the target antigen and conducting in vitro tests to check TCRs for cross-reactivity against human peptides [96], as will be discussed in the following sections.

Safety assessment

The safety assessment includes stages such as verifying the biodistribution of the tested T cells and evaluating their toxicity. This process involves determining virus-specific toxicity, off-tumor toxicity, and cross-reactivity (Fig. 4).

Fig. 4
figure 4

Flow diagram of the “Safety Assessment” stage. AG: antigen, TCR: T-cell receptor

In vivo biodistribution

In preclinical trials, the biodistribution stage using animal models is typically conducted alongside the in vivo proof-of-concept stage described earlier.

Studies in mice have shown that adoptively transferred T cells can localize and kill target cells expressing a cognate antigen, regardless of their anatomical location. For example, in the experiments by Hinrichs C.S. et al., mice received T cells targeting gp100, a melanocyte differentiation antigen expressed by both normal melanocytes and the B16 melanoma line. The T cells migrated indiscriminately to all tissues [24]. However, the engineered T cells exhibited effector functions only in gp100-expressing tissues, indicating an antigen-specific response during their ubiquitous trafficking [114]. In this mouse model, tumor regression was correlated with autoimmunity in the skin and eyes [115].

Nevertheless, animal models used for assessing T-cell biodistribution have several limitations. These models do not fully replicate the components of the human tumor microenvironment, such as the presence of human immunosuppressive cells. Moreover, they are not suitable for predicting the migration of T cells to metastatic tumor sites throughout the patient's body [96]. These challenges in biodistribution analysis remain unresolved, and new approaches are needed to address them.

Toxicity assessment

Virus-specific toxicity

Genotoxicity is a significant concern in adoptive cell therapy. Various tests are conducted to address the potential problems associated with this type of toxicity. Since engineered T-cell receptors are primarily delivered via viral vectors, safety concerns arise due to the risk of insertional mutagenesis and cellular transformation [116]. Clinical experience shows that these risks vary depending on the cell type. For example, the risk of transformation is higher for hematopoietic stem cells than for T cells [117].

Severe adverse events caused by insertional mutagenesis have been observed when using genetically modified hematopoietic stem cells, but no such adverse effects were observed during long-term follow-up of patients who received engineered T cells modified with retroviral constructs [11].

The use of lentiviral and retroviral vectors for generating TCR-T cells carries a risk of insertional mutagenesis. Random vector integration may result in uncontrolled genotoxic effects, cell proliferation, and clonal expansion, potentially leading to multiple primary malignancies [118].

Clonal expansion of engineered T cells due to vector insertion into the TET2 gene (a tumor suppressor gene and a master regulator of blood cell formation) has been documented in several studies. Insertion disrupted the gene’s structure and function, leading to the expansion of a therapeutically effective and significant cell clone [119]. Researchers expanded the study cohort to 39 patients, and Nobles et al. developed a model predicting the response to chronic lymphocytic leukemia treatment with CD19-specific CAR-T cells based on vector integration sites [120]. Another case of clonal expansion involved vector insertion into the gene encoding E3 ubiquitin–protein ligase CBL during the production of CAR-T cells targeting CD22 [121].

Adoptive cellular products have also been developed using transposon-based delivery methods. Early-phase clinical trials of CAR-T cells produced via the Sleeping Beauty system showed no severe toxicity [122,123,124,125]. The PiggyBac (PB) system was also successfully used to generate CAR-T cells [126], but later trials reported cases of CAR-T cell lymphoma [127, 128].

Recent studies suggest a potential link between genetic modification of T cells and their possible oncogenic transformation following CAR-T cell therapy [129, 130]. Several possible mechanisms underlying the development of secondary malignancies, such as lymphomas or leukemia, after CAR-T therapy are currently under discussion. One key factor is the genetic alterations that occur during T cell modification, such as the use of retroviral vectors for CAR delivery. These alterations may accidentally activate oncogenes or inactivate tumor suppressor genes, promoting cancer development [131].

A recent systematic review examines non-relapse mortality (NRM) following CAR-T cell therapy, including rare cases of secondary malignancies [132]. According to the meta-analysis, approximately 7.8% of NRM cases were caused by secondary malignancies. These findings highlight the need for further research and long-term monitoring of patients following CAR-T therapy.

Potential insertional mutagenesis can be evaluated by studying the vector integration patterns used in TCR-T cells [133]. Different variations of Integration Site Analysis are applied to identify integration sites [134]. While the original analysis was PCR-based, bioinformatic tools for processing integration site sequencing data are continuously being developed and improved [135, 136].

The field of genetic engineering has reached new heights with the use of site-specific genomic modifications, particularly with the CRISPR-Cas system in adoptive cell therapy. However, poorly predictable off-target genomic cleavage may occur. These off-target effects include insertions or deletions at unintended sites, which could result in chromosomal translocations between the target and off-target sites or even between different off-target sites [11]. Several genome-wide techniques for assessing off-target cleavage are now in use, including methods for detecting chromosomal translocations in engineered cells [137]. Various bioinformatic tools are available for predicting off-target effects when using CRISPR-Cas or other nucleases, including MIT CRISPR Design Tool [138], E-CRISP [139], GUIDE-seq [140], Digenome-seq [141], CIRCLE-seq [142], SITE-seq [143], and CAST-seq [137].

It’s important to note that the number and efficiency of off-target sites depend heavily on the specific gRNA. In silico methods may not predict experimentally identified off-target sites (leading to false negatives), while in vitro methods may result in false positives, reducing specificity [11, 144].

Off-tumor toxicity and cross-reactivity

Ideally, engineered T cells should selectively target malignant cells. However, target antigens are often expressed on both tumor and healthy tissues, raising concerns about on-target, off-tumor toxicity [145]. The severity of toxic effects depends on how accessible, widespread, and vital the target tissue is [11].

Cross-reactivity of TCRs represents a major safety hazard in TCR gene therapy. T-cell receptors recognize peptides presented by human leukocyte antigen (HLA) class I and II molecules. TCR binding involves interactions between loops of the complementarity-determining region and amino acid residues on HLA molecules and their presented peptides [146]. Thus, TCR binding includes both a peptide-independent HLA-binding component and a peptide-specific component [146]. Both components are crucial for achieving the binding affinity required for T-cell activation. Cross-reactivity occurs when the engineered TCR recognizes one of the autoantigens presented by the HLA allele used to recognize the cognate ligand, leading to off-tumor toxicity. This is especially critical if antigens targeted by the engineered T cells are expressed by essential normal tissues, even at very low levels [24].

Cross-reactivity can also arise when TCRs interact with different HLA alleles that present a variety of peptides, to which the TCR is intolerant. When engineering TCR-T cells, it is important to consider the HLA alleles present in patients. In the case of MHC allele mismatch, the designed TCR may not exhibit the required tolerance to HLA molecules [11].

The risk of autoimmune toxicity increases when using TCRs that have not undergone thymic negative selection, which is designed to remove autoreactive T cells. These include TCRs derived from immunized mice or artificially enhanced TCRs with mutations in the complementarity-determining region that increase affinity [24]. Although effective, this approach increases the risk of cross-reactivity and the recognition of non-specific peptides [11].

Both cross-reactivity scenarios must be evaluated in preclinical trials. Engineered T cells can strongly react with HLA alleles absent in the TCR donor, and allogeneic HLA molecules are among the most immunogenic antigens. Alloreactivity of therapeutic TCRs can be avoided by extensive in vitro screening using cell lines expressing different HLA alleles and by testing TCR-expressing engineered T cells on patient cells before treatment [11].

Additionally, off-target, off-tumor toxicity can result from cross-reactive binding to a mimotope—an epitope that is similar, but not identical, to the target, and is expressed on normal tissues. This off-target binding to cell surface proteins is difficult to predict in preclinical animal trials [11, 13].

To evaluate this risk, the fine specificity profile of the therapeutic TCR must be identified by varying individual residues in the cognate peptide and observing which changes result in the loss of T-cell activation [11].

Several methods for identifying critical amino acid residues for TCR-pMHC interactions are available. Alanine scanning is the most common method, involving the systematic replacement of each peptide residue with alanine. This identifies the combination of amino acids necessary for TCR binding to the pMHC complex [147, 148]. However, alanine scanning does not account for the potential substitution of an amino acid with one having similar physicochemical properties. Mutational position scanning (X-scanning), which uses a peptide library where each epitope residue is sequentially substituted with all possible amino acids, overcomes this limitation [149]. Higher-throughput methods include positional scanning with synthetic combinatorial libraries, where each amino acid is fixed at each position while the remaining peptide consists of random combinations of other amino acids [150,151,152]. The resulting sequences can be compared to the human proteome to predict potential cross-reactive epitopes.

Furthermore, 3D modeling and structure-based analysis strategies using X-ray crystallography data of TCR-pMHC complexes are also available [153, 154]. The extensive structural data collected on TCR-pMHC complexes allow machine learning algorithms to predict TCR-pMHC binding and potential cross-reactivity [155].

Due to the lack of adequate animal models, the on-target toxicity of engineered TCR-T cells cannot be effectively assessed in preclinical studies. This remains a technically challenging task [116].

Conclusions

In this review, we have summarized the requirements outlined in current guidelines for testing genetically modified TCR-T cells and proposed a flow diagram for conducting preclinical trials. Despite significant progress in this field, there remain knowledge gaps in certain stages of preclinical testing of modified T cells, as no existing in vivo models can fully demonstrate the biodistribution and potential toxic effects of these cells. Therefore, further research and clinical experience are necessary. The findings from ongoing studies should be carefully analyzed to improve and standardize the technologies associated with preclinical trials of TCR-T cells.

Data availability

Not applicable.

Change history

References

  1. Baulu E, Gardet C, Chuvin N, Depil S. TCR-engineered T cell therapy in solid tumors: state of the art and perspectives. Sci Adv. 2023;9(7):3700.

    Article  Google Scholar 

  2. Guedan S, Luu M, Ammar D, Barbao P, Bonini C, Bousso P, Buchholz CJ, Casucci M, De Angelis B, Donnadieu E, et al. Time 2EVOLVE: predicting efficacy of engineered T-cells—how far is the bench from the bedside? J Immunother Cancer. 2022;10(5): e003487.

    Article  PubMed  PubMed Central  Google Scholar 

  3. June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361–5.

    Article  CAS  PubMed  Google Scholar 

  4. Saez-Ibanez AR, Upadhaya S, Partridge T, Shah M, Correa D, Campbell J, et al. Landscape of cancer cell therapies: trends and real-world data. Nat Rev Drug Discov. 2022;21:631.

    Article  CAS  PubMed  Google Scholar 

  5. Chandran SS, Klebanoff CA. T cell receptor-based cancer immunotherapy: emerging efficacy and pathways of resistance. Immunol Rev. 2019;290(1):127–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Shafer P, Kelly LM, Hoyos V. Cancer therapy with TCR-engineered T cells: current strategies, challenges, and prospects. Front Immunol. 2022;13:835762.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Foy SP, Jacoby K, Bota DA, Hunter T, Pan Z, Stawiski E, Ma Y, Lu W, Peng S, Wang CL, et al. Non-viral precision T cell receptor replacement for personalized cell therapy. Nature. 2023;615(7953):687–96.

    Article  CAS  PubMed  Google Scholar 

  8. Perea F, Sánchez-Palencia A, Gómez-Morales M, Bernal M, Concha Á, García MM, Gonzalez-Ramirez AR, Kerick M, Martin J, Garrido F, et al. HLA class I loss and PD-L1 expression in lung cancer: impact on T-cell infiltration and immune escape. Oncotarget. 2018;9(3):4120–33.

    Article  PubMed  Google Scholar 

  9. Pang Z, Lu MM, Zhang Y, Gao Y, Bai JJ, Gu JY, Xie L, Wu WZ. Neoantigen-targeted TCR-engineered T cell immunotherapy: current advances and challenges. Biomark Res. 2023;11(1):104.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Han BS, Ji S, Woo S, Lee JH, Sin JI. Regulation of the translation activity of antigen-specific mRNA is responsible for antigen loss and tumor immune escape in a HER2-expressing tumor model. Sci Rep. 2019;9(1):2855.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Donnadieu E, Luu M, Alb M, Anliker B, Arcangeli S, Bonini C, De Angelis B, Choudhary R, Espie D, Galy A, et al. Time to evolve: predicting engineered T cell-associated toxicity with next-generation models. J Immunother Cancer. 2022;10(5): e003486.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Fumagalli O. Regulatory requirements for car-T cell therapy against cancer: a comparison between the EU and the US. 2019. https://www.dgra.de/media/pdf/studium/master-thesis/master_fumagalli_ornella_2019.pdf. Accessed 23 Aug 2021.

  13. Linette GP, Stadtmauer EA, Maus MV, Rapoport AP, Levine BL, Emery L, Litzky L, Bagg A, Carreno BM, Cimino PJ, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood J Am Soc Hematol. 2013;122(6):863–71.

    CAS  Google Scholar 

  14. Cameron BJ, Gerry AB, Dukes J, Harper JV, Kannan V, Bianchi FC, Grand F, Brewer JE, Gupta M, Plesa G, et al. Identification of a titin-derived HLA-A1–presented peptide as a cross-reactive target for engineered MAGE A3–directed T cells. Sci Transl Med. 2013;5(197):197ra103-197ra103.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS, Kammula US, Royal RE, Sherry RM, Wunderlich JR, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood J Am Soc Hematol. 2009;114(3):535–46.

    CAS  Google Scholar 

  16. Van Den Berg JH, Gomez-Eerland R, Van De Wiel B, Hulshoff L, Van Den Broek D, Bins A, Tan HL, Harper JV, Hassan NJ, Jakobsen BK, et al. Case report of a fatal serious adverse event upon administration of T cells transduced with a MART-1-specific T-cell receptor. Mol Ther. 2015;23(9):1541–50.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DAN, Feldman SA, Davis JL, Morgan RA, Merino MJ, Sherry RM, et al. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011;19(3):620–6.

    Article  CAS  PubMed  Google Scholar 

  18. Guideline on the non-clinical studies required before first clinical use of gene therapy medicinal products. https://www.ema.europa.eu/en/documents/scientific-guideline/guideline-non-clinical-studies-required-first-clinical-use-gene-therapy-medicinal-products_en.pdf. Accessed 3 Apr 2019.

  19. Guideline on quality, non-clinical and clinical aspects of medicinal products containing genetically modified cells. https://www.ema.europa.eu/documents/scientific-guideline/draft-guideline-quality-non-clinical-clinical-aspects-medicinal-products-containing-genetically_en.pdf. Accessed 30 Apr 2019.

  20. Guideline on quality, non-clinical and clinical requirements for investigational advanced therapy medicinal products in clinical trials. https://www.ema.europa.eu/en/documents/scientific-guideline/draft-guideline-quality-non-clinical-clinical-requirements-investigational-advanced-therapy-medicinal-products-clinical-trials-first-version_en.pdf. Accessed 3 Apr 2019.

  21. Preclinical assessment of investigational cellular and gene therapy products. https://www.fda.gov/regulatory-information/search-fda-guidance-documents/preclinical-assessment-investigational-cellular-and-gene-therapy-products. Accessed 30 Apr 2019.

  22. Order of the Ministry of Health of the Russian Federation dated August 8, 2018 No. 512n. On the approval of the rules of good practice for working with biomedical cellular products. (in Russian). https://base.garant.ru/72092774/. Accessed 3 Oct 2024.

  23. Federal Law dated June 23, 2016 No. 180-FZ. On biomedical cellular products. (in Russian). https://static-0.minzdrav.gov.ru/system/attachments/attaches/000/057/678/original/%D0%A4%D0%B5%D0%B4%D0%B5%D1%80%D0%B0%D0%BB%D1%8C%D0%BD%D1%8B%D0%B9_%D0%B7%D0%B0%D0%BA%D0%BE%D0%BD_%E2%84%96_180-%D0%A4%D0%97.DOCX?1628772692. Accessed 3 Oct 2024.

  24. Hinrichs CS, Restifo NP. Reassessing target antigens for adoptive T-cell therapy. Nat Biotechnol. 2013;31(11):999–1008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhang J, Wang L. The emerging world of TCR-T cell trials against cancer: a systematic review. Technol Cancer Res Treat. 2019;18:153303381983106.

    Article  Google Scholar 

  26. Alrhmoun S, Sennikov S. The role of tumor-associated antigen HER2/neu in tumor development and the different approaches for using it in treatment: many choices and future directions. Cancers. 2022;14(24):6173.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen Q, Sun Y, Li H. Application of CAR-T cell therapy targeting mesothelin in solid tumor treatment. Discov Oncol. 2024;15(1):1–23.

    Google Scholar 

  28. Rohaan MW, Gomez-Eerland R, van den Berg JH, Geukes Foppen MH, van Zon M, Raud B, Jedema I, Scheij S, de Boer R, Bakker NAM, et al. Mart-1 TCR gene-modified peripheral blood T cells for the treatment of metastatic melanoma: a phase I/IIa clinical trial. Immuno-Oncol Technol. 2022;15:100089.

    Article  CAS  Google Scholar 

  29. Alsalloum A, Shevchenko JA, Sennikov S. The melanoma-associated antigen family a (MAGE-A): a promising target for cancer immunotherapy? Cancers. 2023;15(6):1779.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Alsalloum A, Alrhmoun S, Shevchenko J, Fisher M, Philippova J, Perik-Zavodskii R, Perik-Zavodskaia O, Lopatnikova J, Kurilin V, Volynets M, et al. TCR-engineered lymphocytes targeting NY-ESO-1: in vitro assessment of cytotoxicity against tumors. Biomedicines. 2023;11(10):2805.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Gnjatic S, Nishikawa H, Jungbluth AA, Güre AO, Ritter G, Jäger E, Knuth A, Chen YT, Old LJ. NY-ESO-1: review of an immunogenic tumor antigen. Adv Cancer Res. 2006;95:1–30.

    Article  CAS  PubMed  Google Scholar 

  32. Abramson JS. Anti-CD19 CAR T-cell therapy for B-cell non-Hodgkin lymphoma. Transfus Med Rev. 2020;34(1):29–33.

    Article  PubMed  Google Scholar 

  33. Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood J Am Soc Hematol. 2015;125(26):4017–23.

    CAS  Google Scholar 

  34. Roex G, Timmers M, Wouters K, Campillo-Davo D, Flumens D, Schroyens W, Chu Y, Berneman ZN, Lion E, Luo F, et al. Safety and clinical efficacy of BCMA CAR-T-cell therapy in multiple myeloma. J Hematol Oncol. 2020;13:1–14.

    Article  Google Scholar 

  35. Li J, Xiao Z, Wang D, Jia L, Nie S, Zeng X, Hu W. The screening, identification, design and clinical application of tumor-specific neoantigens for TCR-T cells. Mol Cancer. 2023;22(1):141.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kast F, Klein C, Umaña P, Gros A, Gasser S. Advances in identification and selection of personalized neoantigen/T-cell pairs for autologous adoptive T cell therapies. Oncoimmunology. 2021;10(1):1869389.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Leidner R, Sanjuan Silva N, Huang H, Sprott D, Zheng C, Shih YP, Leung A, Payne R, Sutcliffe K, Cramer J, et al. Neoantigen T-cell receptor gene therapy in pancreatic cancer. N Engl J Med. 2022;386(22):2112–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chandran SS, Ma J, Klatt MG, Dündar F, Bandlamudi C, Razavi P, Wen HY, Weigelt B, Zumbo P, Fu SN, et al. Immunogenicity and therapeutic targeting of a public neoantigen derived from mutated PIK3CA. Nat Med. 2022;28(5):946–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Doran SL, Stevanović S, Adhikary S, Gartner JJ, Jia L, Kwong MLM, Faquin WC, Hewitt SM, Sherry RM, Yang JC, et al. T-cell receptor gene therapy for human papillomavirus–associated epithelial cancers: a first-in-human, phase I/II study. J Clin Oncol. 2019;37(30):2759.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Smith CC, Selitsky SR, Chai S, Armistead PM, Vincent BG, Serody JS. Alternative tumour-specific antigens. Nat Rev Cancer. 2019;19(8):465–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Nadal R, Cherkasova E, Barisic S, Granadier D, Aue G, Wells B, Hawks G, Hughes T, Shalabi R, Stronceket D, et al. A phase I study of HERV-E TCR transduced autologous T Cells (HERV-E TCR T Cells) in patients (pts) with metastatic clear cell renal cell carcinoma (mccRCC). Ann Oncol. 2018;29: viii329.

    Article  Google Scholar 

  42. Moore TV, Nishimura MI. Improved MHC II epitope prediction—a step towards personalized medicine. Nat Rev Clin Oncol. 2020;17(2):71–2.

    Article  PubMed  Google Scholar 

  43. Leko V, Rosenberg SA. Identifying and targeting human tumor antigens for T cell-based immunotherapy of solid tumors. Cancer Cell. 2020;38(4):454–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Davis L, Miller RE, Wong YNS. The landscape of adoptive cellular therapies in ovarian cancer. Cancers. 2023;15(19):4814.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chmielowski B, Ejadi S, Funke R, Stallings-Schmitt T, Denker M, Frohlich MW, Franzusoff AJ, Abedi M, Cristea MC. A phase Ia/Ib, open-label first-in-human study of the safety, tolerability, and feasibility of gene-edited autologous NeoTCR-T cells (NeoTCR-P1) administered to patients with locally advanced or metastatic solid tumors. J Clin Oncol. 2020;38(15): TPS3151.

    Article  Google Scholar 

  46. Mecklenbräuker S, Skoczylas P, Biernat P, Zaghla B, Król-Józaga B, Jasiński M, Murcia Pienkowski V, Sanecka-Duin A, Popp O, Szatanek R, et al. Identification of tumor-specific MHC ligands through improved biochemical isolation and incorporation of machine learning. bioRxiv. 2023.06.08.544182. 2023.

  47. Springer I, Besser H, Tickotsky-Moskovitz N, Dvorkin S, Louzoun Y. Prediction of specific TCR-peptide binding from large dictionaries of TCR-peptide pairs. Front Immunol. 2020;11:1803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Robbins PF, Kassim SH, Tran TL, Crystal JS, Morgan RA, Feldman SA, Yang JC, Dudley ME, Wunderlich JR, Sherry RM, et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1–reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res. 2015;21(5):1019–27.

    Article  CAS  PubMed  Google Scholar 

  49. Hadrup SR, Maurer D, Laske K, Frøsig TM, Andersen SR, Britten CM, van der Burg SH, Walter S, Gouttefangeas C. Cryopreservation of MHC multimers: recommendations for quality assurance in detection of antigen specific T cells. Cytometry A. 2015;87(1):37–48.

    Article  PubMed  Google Scholar 

  50. Wong CP, Okada CY, Levy R. TCR vaccines against T cell lymphoma: QS-21 and IL-12 adjuvants induce a protective CD8+ T cell response. J Immunol. 1999;162(4):2251–8.

    Article  CAS  PubMed  Google Scholar 

  51. White J, Blackman M, Bill J, Kappler J, Marrack PPGD, Gold DP, Born W. Two better cell lines for making hybridomas expressing specific T cell receptors. J Immunol. 1989;143(6):1822–5.

    Article  CAS  PubMed  Google Scholar 

  52. Ishihara M, Kitano S, Kageyama S, Miyahara Y, Yamamoto N, Kato H, Mishima H, Hattori H, Funakoshi T, Kojima T, et al. NY-ESO-1-specific redirected T cells with endogenous TCR knockdown mediate tumor response and cytokine release syndrome. J Immunother Cancer. 2022;10(6): e003811.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Chai J. Atomic-resolution view of complete TCR-CD3 revealed. Protein Cell. 2020;11(3):158–60.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Zoete V, Irving M, Ferber M, Cuendet MA, Michielin O. Structure-based, rational design of T cell receptors. Front Immunol. 2013;4:268.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Mariuzza RA, Agnihotri P, Orban J. The structural basis of T-cell receptor (TCR) activation: an enduring enigma. J Biol Chem. 2020;295(4):914–25.

    Article  PubMed  Google Scholar 

  56. Fischer DS, Wu Y, Schubert B, Theis FJ. Predicting antigen specificity of single T cells based on TCR CDR3 regions. Mol Syst Biol. 2020;16(8): e9416.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. He J, Xiong X, Yang H, Li D, Liu X, Li S, Liao S, Chen S, Wen X, Yu K, et al. Defined tumor antigen-specific T cells potentiate personalized TCR-T cell therapy and prediction of immunotherapy response. Cell Res. 2022;32(6):530–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Dostalek M, Gardner I, Gurbaxani BM, Rose RH, Chetty M. Pharmacokinetics, pharmacodynamics and physiologically-based pharmacokinetic modelling of monoclonal antibodies. Clin Pharmacokinet. 2013;52:83–124.

    Article  CAS  PubMed  Google Scholar 

  59. Shublaq N, Sansom C, Coveney PV. Patient-specific modelling in drug design, development and selection including its role in clinical decision-making. Chem Biol Drug Des. 2013;81(1):5–12.

    Article  CAS  PubMed  Google Scholar 

  60. Benichou J, Ben-Hamo R, Louzoun Y, Efroni S. Rep-Seq: uncovering the immunological repertoire through next-generation sequencing. Immunology. 2012;135(3):183–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Pasetto A, Lu YC. Single-cell TCR and transcriptome analysis: an indispensable tool for studying T-cell biology and cancer immunotherapy. Front Immunol. 2021;12:689091.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Tu AA, Gierahn TM, Monian B, Morgan DM, Mehta NK, Ruiter B, Shreffler WG, Shalek AK, Love JC. TCR sequencing paired with massively parallel 3′ RNA-seq reveals clonotypic T cell signatures. Nat Immunol. 2019;20(12):1692–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Shin W, Mun S, Choi S, Han K. Application of NanoString technologies in angioimmunoblastic T cell lymphoma. Genes Genomics. 2020;42:485–94.

    Article  CAS  PubMed  Google Scholar 

  64. Dutton G. A functional cell biology company is formed: PhenomeX, created through Berkeley Lights’ acquisition of IsoPlexis, consolidates optofluidic and barcoding technologies to realize phenomic applications. Genet Eng Biotechnol News. 2023;43(5):14–5.

    Article  Google Scholar 

  65. Liu S, Iorgulescu JB, Li S, Borji M, Barrera-Lopez IA, Shanmugam V, Lyu H, Morriss JW, Garcia ZN, Murray E, et al. Spatial maps of T cell receptors and transcriptomes reveal distinct immune niches and interactions in the adaptive immune response. Immunity. 2022;55(10):1940-1952.e5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Ellis GI, Sheppard NC, Riley JL. Genetic engineering of T cells for immunotherapy. Nat Rev Genet. 2021;22(7):427–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Gee AP. Manufacturing genetically modified T cells for clinical trials. Cancer Gene Ther. 2015;22(2):67–71.

    Article  CAS  PubMed  Google Scholar 

  68. Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancer therapy. Nat Rev Cancer. 2013;13(8):525–41.

    Article  CAS  PubMed  Google Scholar 

  69. Clauss J. Efficient non-viral T cell engineering for TCR gene therapy by Sleeping Beauty minicircles. 2023.

  70. Grier AE. Genome engineering to expand applications of human T-cell immunotherapy. Ph.D. dissertation. 2017.

  71. Sicher I, Goff A, Loo J, Clary N, Zamarayeva A, Calero-Garcia M. Novel microfluidic platform for scalable transfection of CRISPR/Cas9 RNP in human T cells. 2020.

  72. Lai W, Chang CH, Farber DL. Gene transfection and expression in resting and activated murine CD4 T cell subsets. J Immunol Methods. 2003;282(1–2):93–102.

    Article  CAS  PubMed  Google Scholar 

  73. Pinto IS, Cordeiro RA, Faneca H. Polymer- and lipid-based gene delivery technology for CAR T cell therapy. J Control Release. 2023;353:196–215.

    Article  CAS  PubMed  Google Scholar 

  74. Edes I, Clauss J, Stahn R, Japp AS, Lorenz FKM. TCR and CAR engineering of primary human T cells. In: Walther W, editor. Gene therapy of cancer: methods and protocols. New York: Springer US; 2022. p. 85–93.

    Chapter  Google Scholar 

  75. Rossi M, Breman E. Engineering strategies to safely drive CAR T-cells into the future. Front Immunol. 2024;15:1411393.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Stärck L, Popp K, Pircher H, Uckert W. Immunotherapy with TCR-redirected T cells: comparison of TCR-transduced and TCR-engineered hematopoietic stem cell–derived T cells. J Immunol. 2014;192(1):206–13.

    Article  PubMed  Google Scholar 

  77. Hiasa A, Nishikawa H, Hirayama M, Kitano S, Okamoto S, Chono H, Yu SS, Mineno J, Tanaka Y, Minato N, et al. Rapid αβ TCR-mediated responses in γδ T cells transduced with cancer-specific TCR genes. Gene Ther. 2009;16(5):620–8.

    Article  CAS  PubMed  Google Scholar 

  78. Perro M, Tsang J, Xue SA, Escors D, Cesco-Gaspere M, Pospori C, Gao L, Hart D, Collins M, Stauss H, et al. Generation of multi-functional antigen-specific human T-cells by lentiviral TCR gene transfer. Gene Ther. 2010;17(6):721–32.

    Article  CAS  PubMed  Google Scholar 

  79. Cribbs AP, Kennedy A, Gregory B, Brennan FM. Simplified production and concentration of lentiviral vectors to achieve high transduction in primary human T cells. BMC Biotechnol. 2013;13:1–8.

    Article  Google Scholar 

  80. Reuß S, Sebestyén Z, Heinz N, Loew R, Baum C, Debets R, Uckert W. TCR-engineered T cells: a model of inducible TCR expression to dissect the interrelationship between two TCRs. Eur J Immunol. 2014;44(1):265–74.

    Article  PubMed  Google Scholar 

  81. Yusufi N, Mall S, Bianchi HO, Steiger K, Reder S, Klar R, Audehm S, Mustafa M, Nekolla S, Peschel C, et al. In-depth characterization of a TCR-specific tracer for sensitive detection of tumor-directed transgenic T cells by immuno-PET. Theranostics. 2017;7(9):2402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Sims S, Willberg C, Klenerman P. MHC–peptide tetramers for the analysis of antigen-specific T cells. Expert Rev Vaccines. 2010;9(7):765–74.

    Article  CAS  PubMed  Google Scholar 

  83. Campillo-Davo D, Flumens D, Lion E. The quest for the best: how TCR affinity, avidity, and functional avidity affect TCR-engineered T-cell antitumor responses. Cells. 2020;9(7):1720.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Lu A, Liu H, Shi R, Cai Y, Ma J, Shao L, Rong V, Gkitsas N, Lei H, Highfill SL, et al. Application of droplet digital PCR for the detection of vector copy number in clinical CAR/TCR T cell products. J Transl Med. 2020;18(1):1–7.

    Article  Google Scholar 

  85. Kubista M, Andrade JM, Bengtsson M, Forootan A, Jonák J, Lind K, Sindelka R, Sjöback R, Sjögreen B, Strömbom L, et al. The real-time polymerase chain reaction. Mol Aspects Med. 2006;27(2–3):95–125.

    Article  CAS  PubMed  Google Scholar 

  86. Hillerdal V, Boura VF, Björkelund H, Andersson K, Essand M. Avidity characterization of genetically engineered T-cells with novel and established approaches. BMC Immunol. 2016;17:1–10.

    Article  Google Scholar 

  87. Kiesgen S, Messinger JC, Chintala NK, Tano Z, Adusumilli PS. Comparative analysis of assays to measure CAR T-cell-mediated cytotoxicity. Nat Protoc. 2021;16(3):1331–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Adan A, Kiraz Y, Baran Y. Cell proliferation and cytotoxicity assays. Curr Pharm Biotechnol. 2016;17(14):1213–21.

    Article  CAS  PubMed  Google Scholar 

  89. Cho MH, Niles A, Huang R, Inglese J, Austin CP, Riss T, Xia M. A bioluminescent cytotoxicity assay for assessment of membrane integrity using a proteolytic biomarker. Toxicol In Vitro. 2008;22(4):1099–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Brunner KT, Mauel J, Cerottini JC, Chapuis B. Quantitative assay of the lytic action of immune lymphoid cells of 51Cr-labelled allogeneic target cells in vitro; inhibition by isoantibody and by drugs. Immunology. 1968;14(2):181.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Kumar P, Nagarajan A, Uchil PD. Analysis of cell viability by the lactate dehydrogenase assay. Cold Spring Harb Protoc. 2018;2018(6):465–8.

    Google Scholar 

  92. Karimi MA, Lee E, Bachmann MH, Salicioni AM, Behrens EM, Kambayashi T, Baldwin CL. Measuring cytotoxicity by bioluminescence imaging outperforms the standard chromium-51 release assay. PLoS ONE. 2014;9(2): e89357.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Mensali N, Myhre MR, Dillard P, Pollmann S, Gaudernack G, Kvalheim G, Wälchli S, Inderberg EM. Preclinical assessment of transiently TCR redirected T cells for solid tumour immunotherapy. Cancer Immunol Immunother. 2019;68:1235–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Zhu J, Wang X, Xu X, Abassi YA. Dynamic and label-free monitoring of natural killer cell cytotoxic activity using electronic cell sensor arrays. J Immunol Methods. 2006;309(1–2):25–33.

    Article  CAS  PubMed  Google Scholar 

  95. Xi B, Berahovich R, Zhou H, Xu S, Wei Y, Guan J, Wu L, Santa Ana D, Cerignoil F, Lamarche B, et al. A real-time potency assay for chimeric antigen receptor T cells targeting solid and hematological cancer cells. JoVE. 2019;153: e59033.

    Google Scholar 

  96. Jin BY, Campbell TE, Draper LM, Stevanović S, Weissbrich B, Yu Z, Restifo NP, Rosenberg SA, Trimble CL, Hinrichs CS. Engineered T cells targeting E7 mediate regression of human papillomavirus cancers in a murine model. JCI Insight. 2018;3(8): e99488.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Jedema I, van der Werff NM, Barge RM, Willemze R, Falkenburg JH. New CFSE-based assay to determine susceptibility to lysis by cytotoxic T cells of leukemic precursor cells within a heterogeneous target cell population. Blood. 2004;103(7):2677–82.

    Article  CAS  PubMed  Google Scholar 

  98. Riccardi C, Nicoletti I. Analysis of apoptosis by propidium iodide staining and flow cytometry. Nat Protoc. 2006;1(3):1458–61.

    Article  CAS  PubMed  Google Scholar 

  99. Logue SE, Elgendy M, Martin SJ. Expression, purification and use of recombinant annexin V for the detection of apoptotic cells. Nat Protoc. 2009;4(9):1383–95.

    Article  CAS  PubMed  Google Scholar 

  100. Jin Q, Jiang L, Chen Q, Li X, Xu Y, Sun X, Zhao Z, Wei L. Rapid flow cytometry-based assay for the evaluation of γδ T cell-mediated cytotoxicity. Mol Med Rep. 2018;17(3):3555–62.

    CAS  PubMed  Google Scholar 

  101. Taraseviciute A, Tkachev V, Ponce R, Turtle CJ, Snyder JM, Liggitt HD, Myerson D, Gonzalez-Cuyar L, Baldessari A, English C, et al. Chimeric antigen receptor T cell–mediated neurotoxicity in nonhuman primates. Cancer Discov. 2018;8(6):750–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Künkele A, Taraseviciute A, Finn LS, Johnson AJ, Berger C, Finney O, Chang CA, Rolczynski LS, Brown C, Mgebroff S, et al. Preclinical assessment of CD171-directed CAR T-cell adoptive therapy for childhood neuroblastoma: CE7 epitope target safety and product manufacturing feasibility. Clin Cancer Res. 2017;23(2):466–77.

    Article  PubMed  Google Scholar 

  103. Berger C, Sommermeyer D, Hudecek M, Berger M, Balakrishnan A, Paszkiewicz PJ, Kosasih PL, Rader C, Riddell SR. Safety of targeting ROR1 in primates with chimeric antigen receptor–modified T cells. Cancer Immunol Res. 2015;3(2):206–16.

    Article  CAS  PubMed  Google Scholar 

  104. Gardner HL, Fenger JM, London CA. Dogs as a model for cancer. Annu Rev Anim Biosci. 2016;4:199–222.

    Article  CAS  PubMed  Google Scholar 

  105. Yoon JY, Lee D, Yun T, Koo Y, Chae Y, Kang JH, Kang BT, Yang MP, Kim H. Anaphylactic reaction after subcutaneous vitamin K1 injection in dogs: an experimental study and case report. J Biomed Transl Res. 2022;23(2):35–42.

    Article  Google Scholar 

  106. O’Connor CM, Wilson-Robles H. Developing T cell cancer immunotherapy in the dog with lymphoma. ILAR J. 2014;55(1):169–81.

    Article  CAS  PubMed  Google Scholar 

  107. Ramos-Cardona XE, Luo W, Mohammed SI. Advances and challenges of CAR T therapy and suitability of animal models. Mol Clin Oncol. 2022;17(3):1–11.

    Article  Google Scholar 

  108. Mochel JP, Ekker SC, Johannes CM, Jergens AE, Allenspach K, Bourgois-Mochel A, Knouse M, Benzekry S, Wierson W, LeBlanc AK, et al. CAR T cell immunotherapy in human and veterinary oncology: changing the odds against hematological malignancies. AAPS J. 2019;21:1–12.

    Article  Google Scholar 

  109. van Amerongen RA, Morton LT, Chaudhari UG, Remst DFG, Hagedoorn RS, van den Berg CW, Freund C, Falkenburg JHF, Heemskerk MHM. Human iPSC-derived preclinical models to identify toxicity of tumor-specific T cells with clinical potential. Molr Ther Methods Clin Dev. 2023;28:249–61.

    Article  Google Scholar 

  110. Li Y, Gao X, Ni C, Zhao B, Cheng X. The application of patient-derived organoid in the research of lung cancer. Cell Oncol. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13402-023-00771-3.

    Article  Google Scholar 

  111. Kutle I, Polten R, Hachenberg J, Klapdor R, Morgan M, Schambach A. Tumor organoid and spheroid models for cervical cancer. Cancers. 2023;15(9):2518.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Yuti P, Wutti-In Y, Sawasdee N, Kongkhla K, Phanthaphol N, Choomee K, Chieochansin T, Panya A, Junking M, Yenchitsomanus PT, et al. Anti-CD19 chimeric antigen receptor T cells secreting anti-PD-L1 single-chain variable fragment attenuate PD-L1 mediated T cell inhibition. Int Immunopharmacol. 2022;113:109442.

    Article  CAS  PubMed  Google Scholar 

  113. Schnalzger TE, de Groot MH, Zhang C, Mosa MH, Michels BE, Röder J, Darvishi T, Wels WS, Farin HF. 3D model for CAR-mediated cytotoxicity using patient-derived colorectal cancer organoids. EMBO J. 2019;38(12): e100928.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Palmer DC, Balasubramaniam S, Hanada K, Wrzesinski C, Yu Z, Farid S, Theoret MR, Hwang LN, Klebanoff CA, Gattinoni L, et al. Vaccine-stimulated, adoptively transferred CD8+ T cells traffic indiscriminately and ubiquitously while mediating specific tumor destruction. J Immunol. 2004;173(12):7209–16.

    Article  CAS  PubMed  Google Scholar 

  115. Palmer DC, Chan CC, Gattinoni L, Wrzesinski C, Paulos CM, Hinrichs CS, Powell DJ Jr, Klebanoff CA, Finkelstein SE, Fariss RN, et al. Effective tumor treatment targeting a melanoma/melanocyte-associated antigen triggers severe ocular autoimmunity. Proc Natl Acad Sci. 2008;105(23):8061–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Finck AV, Blanchard T, Roselle CP, Golinelli G, June CH. Engineered cellular immunotherapies in cancer and beyond. Nat Med. 2022;28(4):678–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Ferrari G, Thrasher AJ, Aiuti A. Gene therapy using haematopoietic stem and progenitor cells. Nat Rev Genet. 2021;22(4):216–34.

    Article  CAS  PubMed  Google Scholar 

  118. Bushman FD. Retroviral insertional mutagenesis in humans: evidence for four genetic mechanisms promoting expansion of cell clones. Mol Ther. 2020;28(2):352–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Fraietta JA, Nobles CL, Sammons MA, Lundh S, Carty SA, Reich TJ, Cogdill AP, Morrissette JJD, DeNizio JE, Reddy S, et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature. 2018;558(7709):307–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Nobles CL, Sherrill-Mix S, Everett JK, Reddy S, Fraietta JA, Porter DL, Frey N, Gill SI, Grupp SA, Maude SL, et al. CD19-targeting CAR T cell immunotherapy outcomes correlate with genomic modification by vector integration. J Clin Investig. 2020;130(2):673–85.

    Article  CAS  PubMed  Google Scholar 

  121. Shah NN, Qin H, Yates B, Su L, Shalabi H, Raffeld M, Ahlman MA, Stetler-Stevenson M, Yuan C, Guo S, et al. Clonal expansion of CAR T cells harboring lentivector integration in the CBL gene following anti-CD22 CAR T-cell therapy. Blood Adv. 2019;3(15):2317–22.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Kebriaei P, Singh H, Huls MH, Figliola MJ, Bassett R, Olivares S, Jena B, Dawson MJ, Kumaresan PR, Su S, et al. Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells. J Clin Investig. 2016;126(9):3363–76.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Magnani CF, Gaipa G, Lussana F, Belotti D, Gritti G, Napolitano S, Matera G, Cabiati B, Buracchi C, Borleri G, et al. Sleeping Beauty–engineered CAR T cells achieve antileukemic activity without severe toxicities. J Clin Investig. 2020;130(11):6021–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Prommersberger S, Reiser M, Beckmann J, Danhof S, Amberger M, Quade-Lyssy P, Einsele H, Hudecek M, Bonig H, Ivics Z. CARAMBA: a first-in-human clinical trial with SLAMF7 CAR-T cells prepared by virus-free Sleeping Beauty gene transfer to treat multiple myeloma. Gene Ther. 2021;28(9):560–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Srour SA, Singh H, McCarty J, de Groot E, Huls H, Rondon G, Qazilbash M, Ciurea S, Bardelli G, Buck J, et al. Long-term outcomes of Sleeping Beauty–generated CD19-specific CAR T-cell therapy for relapsed-refractory B-cell lymphomas. Blood J Am Soc Hematol. 2020;135(11):862–5.

    CAS  Google Scholar 

  126. Li C, Sun Y, Wang J, Tang L, Jiang H, Guo T, Liu L, Wu Y, Ai L, Xia L, et al. PiggyBac-generated CAR19-T cells plus lenalidomide cause durable complete remission of triple-hit Refractory/Relapsed DLBCL: a case report. Front Immunol. 2021;12:599493.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Bishop DC, Clancy LE, Simms R, Burgess J, Mathew G, Moezzi L, Street JA, Sutrave G, Atkins E, McGuire HM, et al. Development of CAR T-cell lymphoma in 2 of 10 patients effectively treated with piggyBac-modified CD19 CAR T cells. Blood J Am Soc Hematol. 2021;138(16):1504–9.

    CAS  Google Scholar 

  128. Micklethwaite KP, Gowrishankar K, Gloss BS, Li Z, Street JA, Moezzi L, Mach MA, Sutrave G, Clancy LE, Bishop DC, et al. Investigation of product-derived lymphoma following infusion of piggyBac-modified CD19 chimeric antigen receptor T cells. Blood J Am Soc Hematol. 2021;138(16):1391–405.

    CAS  Google Scholar 

  129. Levine BL, Pasquini MC, Connolly JE, Porter DL, Gustafson MP, Boelens JJ, Horwitz EM, Grupp SA, Maus MV, Locke FL, et al. Unanswered questions following reports of secondary malignancies after CAR-T cell therapy. Nat Med. 2024;30(2):338–41.

    Article  CAS  PubMed  Google Scholar 

  130. Ghilardi G, Fraietta JA, Gerson JN, Van Deerlin VM, Morrissette JJ, Caponetti GC, Paruzzo L, Harris JC, Chong EA, Susanibar Adaniya SP, et al. T cell lymphoma and secondary primary malignancy risk after commercial CAR T cell therapy. Nat Med. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41591-024-02826-w.

    Article  PubMed  Google Scholar 

  131. Storgard R, Rejeski K, Perales MA, Goldman A, Shouval R. T-cell malignant neoplasms after chimeric antigen receptor T-cell therapy. JAMA Oncol. 2024;10(6):826–8.

    Article  PubMed  Google Scholar 

  132. dos Cordas Santos DM, Tix T, Shouval R, Gafter-Gvili A, Alberge JB, Cliff ERS, Theurich S, von Bergwelt-Baildon M, Ghobrial IM, Subklewe M, et al. A systematic review and meta-analysis of nonrelapse mortality after CAR T cell therapy. Nat Med. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41591-024-03084-6.

    Article  Google Scholar 

  133. Lebrec H, Maier CC, Maki K, Ponce R, Shenton J, Green S. Nonclinical safety assessment of engineered T cell therapies. Regul Toxicol Pharmacol. 2021;127:105064.

    Article  CAS  PubMed  Google Scholar 

  134. Schmidt M, Glimm H, Wissler M, Hoffmann G, Olsson K, Sellers S, Carbonaro D, Tisdale JF, Leurs C, Hanenberg H, et al. Efficient characterization of retro-, lenti-, and foamyvector-transduced cell populations by high-accuracy insertion site sequencing. Ann N Y Acad Sci. 2003;996(1):112–21.

    Article  CAS  PubMed  Google Scholar 

  135. Hocum JD, Battrell LR, Maynard R, Adair JE, Beard BC, Rawlings DJ, Kiem HP, Miller DG, Trobridge GD. VISA-Vector Integration Site Analysis server: a web-based server to rapidly identify retroviral integration sites from next-generation sequencing. BMC Bioinform. 2015;16:1–5.

    Article  Google Scholar 

  136. Yan A, Baricordi C, Nguyen Q, Barbarossa L, Loperfido M, Biasco L. IS-Seq: a bioinformatics pipeline for integration sites analysis with comprehensive abundance quantification methods. BMC Bioinform. 2023;24(1):286.

    Article  Google Scholar 

  137. Turchiano G, Andrieux G, Klermund J, Blattner G, Pennucci V, El Gaz M, Monaco G, Poddar S, Mussolino C, Cornu TI, et al. Quantitative evaluation of chromosomal rearrangements in gene-edited human stem cells by CAST-Seq. Cell Stem Cell. 2021;28(6):1136-1147.e5.

    Article  CAS  PubMed  Google Scholar 

  138. Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8(11):2281–308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Heigwer F, Kerr G, Boutros M. E-CRISP: fast CRISPR target site identification. Nat Methods. 2014;11(2):122–3.

    Article  CAS  PubMed  Google Scholar 

  140. Tsai SQ, Zheng Z, Nguyen NT, Liebers M, Topkar VV, Thapar V, Wyvekens N, Khayter C, Iafrate AJ, Le LP, et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat Biotechnol. 2015;33(2):187–97.

    Article  CAS  PubMed  Google Scholar 

  141. Kim D, Bae S, Park J, Kim E, Kim S, Yu HR, Hwang J, Kim JI, Kim JS. Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-target effects in human cells. Nat Methods. 2015;12(3):237–43.

    Article  CAS  PubMed  Google Scholar 

  142. Tsai SQ, Nguyen NT, Malagon-Lopez J, Topkar VV, Aryee MJ, Joung JK. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR–Cas9 nuclease off-targets. Nat Methods. 2017;14(6):607–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Cameron P, Fuller CK, Donohoue PD, Jones BN, Thompson MS, Carter MM, Gradia S, Vidal B, Garner E, Slorach EM, et al. Mapping the genomic landscape of CRISPR–Cas9 cleavage. Nat Methods. 2017;14(6):600–6.

    Article  CAS  PubMed  Google Scholar 

  144. Kim D, Luk K, Wolfe SA, Kim JS. Evaluating and enhancing target specificity of gene-editing nucleases and deaminases. Annu Rev Biochem. 2019;88:191–220.

    Article  CAS  PubMed  Google Scholar 

  145. Larson RC, Maus MV. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat Rev Cancer. 2021;21(3):145–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Stauss HJ. Immunotherapy with CTLs restricted by nonself MHC. Immunol Today. 1999;20(4):180–3.

    Article  CAS  PubMed  Google Scholar 

  147. Farquhar R, Van Rhijn I, Moody DB, Rossjohn J, Shahine A. αβ T-cell receptor recognition of self-phosphatidylinositol presented by CD1b. J Biol Chem. 2023;299(2):102849.

    Article  CAS  PubMed  Google Scholar 

  148. Svitek N, Saya R, Zhang H, Nene V, Steinaa L. Systematic determination of TCR–antigen and peptide–MHC binding kinetics among field variants of a Theileria parva polymorphic CTL epitope. J Immunol. 2022;208(3):549–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Wooldridge L, Laugel B, Ekeruche J, Clement M, van den Berg HA, Price DA, Sewell AK. CD8 controls T cell cross-reactivity. J Immunol. 2010;185(8):4625–32.

    Article  CAS  PubMed  Google Scholar 

  150. Schneider EL, Craik CS. Positional scanning synthetic combinatorial libraries for substrate profiling. In: Bugge T, Antalis T, editors. Proteases and cancer: methods and protocols. Totowa: Humana Press; 2009. p. 59–78.

    Chapter  Google Scholar 

  151. Naghavian R, Faigle W, Oldrati P, Wang J, Toussaint NC, Qiu Y, Medici G, Wacker M, Freudenmann LK, Bonté PE, et al. Microbial peptides activate tumour-infiltrating lymphocytes in glioblastoma. Nature. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-023-06081-w.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Wang J, Jelcic I, Mühlenbruch L, Haunerdinger V, Toussaint NC, Zhao Y, Cruciani C, Faigle W, Naghavian R, Foege M, et al. HLA-DR15 molecules jointly shape an autoreactive T cell repertoire in multiple sclerosis. Cell. 2020;183(5):1264-1281.e20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Hellman LM, Foley KC, Singh NK, Alonso JA, Riley TP, Devlin JR, Ayres CM, Keller GLJ, Zhang Y, Vander Kooi CW, et al. Improving T cell receptor on-target specificity via structure-guided design. Mol Ther. 2019;27(2):300–13.

    Article  CAS  PubMed  Google Scholar 

  154. Gupta S, Nerli S, Kandy SK, Mersky GL, Sgourakis NG. HLA3DB: comprehensive annotation of peptide/HLA complexes enables blind structure prediction of T cell epitopes. Nat Commun. 2023;14(1):6349.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Lu T, Zhang Z, Zhu J, Wang Y, Jiang P, Xiao X, Bernatchez C, Heymach JV, Gibbons DL, Wang J, et al. Deep learning-based prediction of the T cell receptor–antigen binding specificity. Nat Mach Intell. 2021;3(10):864–75.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This study was funded by Academic Leadership Program Priority 2030, Russian Science Foundation, project number 21-65-00004. It should be as follows: Preparing of sections 1-3 were supported by Academic Leadership Program Priority 2030 proposed by the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), preparing of sections 4-5 were supported by the Russian Science Foundation, project number 21-65-00004 (https://rscf.ru/project/21-65-00004/, accessed on 20 April 2021).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, E.A.G. and S.V.S.; methodology, E.A.G., A.A.A., N.A.S., and S.A.; investigation, E.A.G., A.A.A., and N.A.S.; resources, A.A.A. and S.V.S.; data curation, A.A.A., S.A. and N.A.S.; writing—original draft preparation, E.A.G. and A.A.A.; writing—review and editing, S.A., N.A.S., and S.V.S.; visualization, E.A.G. and A.A.A.; supervision, S.V.S.; project administration, E.A.G. and S.V.S.; funding acquisition, A.A.A. and S.V.S. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Alina A. Alshevskaya.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

The original version of this article was revised: Funding note was published incorrectly. It is now as follows: This study was funded by Academic Leadership Program Priority 2030, Russian Science Foundation, project number 21-65-00004. It should be as follows: Preparing of sections 1-3 were supported by Academic Leadership Program Priority 2030 proposed by the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), preparing of sections 4-5 were supported by the Russian Science Foundation, project number 21-65-00004 (https://rscf.ru/project/21-65-00004/, accessed on 20 April 2021)

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Golikova, E.A., Alshevskaya, A.A., Alrhmoun, S. et al. TCR-T cell therapy: current development approaches, preclinical evaluation, and perspectives on regulatory challenges. J Transl Med 22, 897 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05703-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05703-9

Keywords