Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
iScience ; 27(6): 109913, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38799557

RESUMEN

Here, we show that a NOT gated cell therapy (Tmod) can exploit antigens such as epidermal growth factor receptor (EGFR) and human leukocyte antigen-E (HLA-E) which are widely expressed on cancer cells. Noncancerous cells-despite high expression of these antigens-are protected from cytotoxicity by the action of an inhibitory receptor ("blocker") via a mechanism that involves blocker modulation of CAR surface expression. The blocker is triggered by the product of a polymorphic HLA allele (e.g., HLA-A∗02) deleted in a significant subset of solid tumors via loss of heterozygosity. Moreover, Tmod constructs that target mouse homologs of EGFR or HLA-E for activation, and a mouse-equivalent of HLA-A∗02 for inhibition, protect mice from toxicity caused by the CAR alone. The blocker also controls graft vs. host response in allogeneic T cells in vitro, consistent with the use of Tmod cells for off-the-shelf therapy without additional gene-editing.

2.
Front Immunol ; 15: 1304765, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38343543

RESUMEN

Clinical applications of CAR-T cells are limited by the scarcity of tumor-specific targets and are often afflicted with the same on-target/off-tumor toxicities that plague other cancer treatments. A new promising strategy to enforce tumor selectivity is the use of logic-gated, two-receptor systems. One well-described application is termed Tmod™, which originally utilized a blocking inhibitory receptor directed towards HLA-I target antigens to create a protective NOT gate. Here we show that the function of Tmod blockers targeting non-HLA-I antigens is dependent on the height of the blocker antigen and is generally compatible with small, membrane-proximal targets. We compensate for this apparent limitation by incorporating modular hinge units to artificially extend or retract the ligand-binding domains relative to the effector cell surface, thereby modulating Tmod activator and blocker function. By accounting for structural differences between activator and blocker targets, we developed a set of simple geometric parameters for Tmod receptor design that enables targeting of blocker antigens beyond HLA-I, thereby broadening the applications of logic-gated cell therapies.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Antígenos/metabolismo
3.
Mol Ther Oncolytics ; 27: 157-166, 2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36381658

RESUMEN

Innovative cell-based therapies are important new weapons in the fight against difficult-to-treat cancers. One promising strategy involves cell therapies equipped with multiple receptors to integrate signals from more than one antigen. We developed a specific embodiment of this approach called Tmod, a two-receptor system that combines activating and inhibitory inputs to distinguish between tumor and normal cells. The selectivity of Tmod is enforced by the inhibitory receptor (blocker) that recognizes an antigen, such as an HLA allele, whose expression is absent from tumors because of loss of heterozygosity. Although unwanted cross-reactivity of the blocker likely reduces efficacy rather than safety, it is important to verify the blocker's specificity. We have tested an A∗02-directed blocker derived from the PA2.1 mouse antibody as a safety mechanism paired with a mesothelin-specific activating CAR in our Tmod construct. We solved the crystal structure of humanized PA2.1 Fab in complex with HLA-A∗02 to determine its binding epitope, which was used to bioinformatically select specific class I HLA alleles to test the blocker's functional specificity in vitro. We found that this A∗02-directed blocker is highly specific for its cognate antigen, with only one cross-reactive allele (A∗69) capable of triggering comparable function.

4.
Clin Immunol ; 241: 109030, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35561999

RESUMEN

Immune cells that are engineered with receptors to integrate signals from multiple antigens offer a promising route to achieve the elusive property of therapeutic selectivity in cancer patients. Several types of multi-signal integrators have been described, among them mechanisms that pair activating and inhibitory receptors which are termed NOT gates by analogy to logical operations performed by machines. Here we review one such NOT-gated signal integrator called the Tmod system which is being developed for patients with solid tumors. Coupled with rigorous selection for patients with defined lesions in their tumor genomes (loss of heterozygosity), the Tmod approach presents an unusual opportunity to create truly selective therapies for certain cancer patients. Several of these agents are advancing toward the clinic, supported by a large body of quantitative preclinical data.


Asunto(s)
Inmunoterapia , Neoplasias , Humanos , Inmunoterapia Adoptiva , Neoplasias/terapia
5.
Front Immunol ; 13: 826747, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35359952

RESUMEN

Progress toward improved solid-tumor treatment has long been hindered by the lack of truly tumor-specific targets. We have developed an approach to T cell therapy based on a dual-receptor system called Tmod™ that addresses this problem. The Tmod system exploits one of the few common genetic differences between tumor and normal cells: loss of heterozygosity (LOH). It utilizes the basic mechanistic logic that evolved in early vertebrates to mediate self vs. non-self discrimination, where an activation stimulus is blocked by self-ligands. Tmod constructs employ a chimeric antigen receptor (CAR) or T cell receptor (TCR) as activator component and a modified LIR-1 inhibitory receptor (blocker) to achieve high selectivity based on expression of the blocker antigen (Ag). Here we explore the in vitro pharmacology of a blocker directed at the HLA-A*02 Ag paired with either a mesothelin CAR or an HLA-A*11-restricted KRAS peptide TCR. While more sensitive to receptor expression changes on effector cells, we show that Tmod response is well-buffered against variations in Ag levels on target cells. In addition, the data reveal at least two distinguishable pharmacologic mechanisms of Tmod blocker function: (1) reducing activator sensitivity and (2) decreasing activation magnitude.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Animales , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos , Antígenos HLA-A , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T
6.
Sci Transl Med ; 14(634): eabm0306, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-35235342

RESUMEN

The CEACAM5 gene product [carcinoembryonic antigen (CEA)] is an attractive target for colorectal cancer because of its high expression in virtually all colorectal tumors and limited expression in most healthy adult tissues. However, highly active CEA-directed investigational therapeutics have been reported to be toxic, causing severe colitis because CEA is expressed on normal gut epithelial cells. Here, we developed a strategy to address this toxicity problem: the Tmod dual-signal integrator. CEA Tmod cells use two receptors: a chimeric antigen receptor (CAR) activated by CEA and a leukocyte Ig-like receptor 1 (LIR-1)-based inhibitory receptor triggered by human leukocyte antigen (HLA)-A*02. CEA Tmod cells exploit instances of HLA heterozygous gene loss in tumors to protect the patient from on-target, off-tumor toxicity. CEA Tmod cells potently killed CEA-expressing tumor cells in vitro and in vivo. But in contrast to a traditional CEA-specific T cell receptor transgenic T cell, Tmod cells were highly selective for tumor cells even when mixed with HLA-A*02-expressing cells. These data support further development of the CEA Tmod construct as a therapeutic candidate for colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Receptores Quiméricos de Antígenos , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Antígeno HLA-A2/genética , Humanos , Pérdida de Heterocigocidad
7.
Toxicol Appl Pharmacol ; 437: 115894, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35085592

RESUMEN

Cell therapy is an emerging therapeutic modality with the power to exploit new cancer targets and potentially achieve positive outcomes for patients with few other options. Like all synthetic treatments, cell therapy has the risk of toxicity via unpredicted off-target behavior. We describe an empirical method to model off-tumor, off-target reactivity of receptors used for investigational T cell therapies. This approach utilizes an optimal panel of diverse human cell-lines to capture the large majority of protein-coding gene expression in adult human tissues. We apply this cell-line set to test Jurkat and primary T cells engineered with a dual-signal integrator, called TmodTM, that contains an activating receptor (activator) and a separate inhibitory receptor (blocker). In proof-of-concept experiments, we use CD19 as the activating antigen and HLA-A*02 as the blocker antigen. This specific Tmod system, which employs a blocker targeting a ubiquitously expressed HLA class I antigen to inhibit CAR activation, has an inherent mechanism for selectivity/safety, designed to activate only when a specific HLA class I antigen is lost. Nonetheless, it is important to test off-target reactivity in functional assays, especially given the disconnect between ligand-binding and function among T cell receptors (TCRs) and chimeric antigen receptors (CARs). We show these cell-based assays yield consistent results with high sensitivity and specificity. The general strategy is likely applicable to more traditional single-receptor CAR- and TCR-T therapeutics.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/fisiología , Antígenos CD19/genética , Antígenos CD19/metabolismo , Línea Celular Tumoral , Biología Computacional , Eliminación de Gen , Regulación de la Expresión Génica , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
J Immunother Cancer ; 10(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35091455

RESUMEN

BACKGROUND: Mesothelin (MSLN) is a classic tumor-associated antigen that is expressed in lung cancer and many other solid tumors. However, MSLN is also expressed in normal mesothelium which creates a significant risk of serious inflammation for MSLN-directed therapeutics. We have developed a dual-receptor (Tmod™) system that exploits the difference between tumor and normal tissue in a subset of patients with defined heterozygous gene loss (LOH) in their tumors. METHODS: T cells engineered with the MSLN CAR Tmod construct described here contain (1) a novel MSLN-activated CAR and (2) an HLA-A*02-gated inhibitory receptor (blocker). A*02 binding is intended to override T-cell cytotoxicity, even in the presence of MSLN. The Tmod system is designed to treat heterozygous HLA class I patients, selected for HLA LOH. When A*02 is absent from tumors selected for LOH, the MSLN Tmod cells are predicted to mediate potent killing of the MSLN(+)A*02(-) malignant cells. RESULTS: The sensitivity of the MSLN Tmod cells is comparable with a benchmark MSLN CAR-T that was active but toxic in the clinic. Unlike MSLN CAR-T cells, the Tmod system robustly protects surrogate "normal" cells even in mixed-cell populations in vitro and in a xenograft model. The MSLN CAR can also be paired with other HLA class I blockers, supporting extension of the approach to patients beyond A*02 heterozygotes. CONCLUSIONS: The Tmod mechanism exemplified by the MSLN CAR Tmod construct provides an alternative route to leverage solid-tumor antigens such as MSLN in safer, more effective ways than previously possible.


Asunto(s)
Antígeno HLA-A2/genética , Inmunoterapia Adoptiva/métodos , Mesotelina/inmunología , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Animales , Línea Celular Tumoral , Femenino , Antígeno HLA-A2/inmunología , Humanos , Pérdida de Heterocigocidad , Ratones , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Res Commun ; 2(1): 58-65, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-36860694

RESUMEN

Neoantigens are among the most intriguing potential immuno-oncology targets because, unlike many cancer targets that are expressed on normal tissues, they are by definition restricted to cancer cells. Medicines directed at common neoantigens such as mutant KRAS are especially interesting because they may offer the convenience and cost of an off-the-shelf therapy. However, all common KRAS mutations produce proteins that differ from the wild type at a single amino acid, creating challenges for molecular discrimination. We have undertaken an effort to optimize single-chain variable fragments (scFv) against peptide/major histocompatibility antigen complexes composed of HLA-A*11 and either G12V- or G12D-mutant KRAS peptides. These scFvs could in principle be used in chimeric antigen receptor (CAR) T-cell therapies for selected patients whose tumors bear either of these mutations. Here we show that optimization of such CARs involves a trade-off between potency and selectivity. We further show that targeting this family without high selectivity engenders risks of cross-reactivity against other members of the G-protein family to which KRAS belongs. Significance: We report an effort to generate high potency, selective CARs directed at mutant KRAS peptides. Although the heavily optimized CARs maintain high selectivity against wild-type KRAS, they lose selectivity against other KRAS-related peptides derived from human proteins. To our knowledge, this work is the first to examine the trade-off between potency and selectivity with regard to KRAS pMHC-directed CARs, illustrating the challenge to achieve both sufficient potency and high selectivity.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Anticuerpos de Cadena Única , Humanos , Receptores Quiméricos de Antígenos/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Inmunoterapia Adoptiva , Anticuerpos de Cadena Única/genética
10.
Mol Immunol ; 138: 137-149, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34419823

RESUMEN

Though TCRs have been subject to limited engineering in the context of therapeutic design and optimization, they are used largely as found in nature. On the other hand, CARs are artificial, composed of different segments of proteins that function in the immune system. This characteristic raises the possibility of altered response to immune regulatory stimuli. Here we describe a large-scale, systematic comparison of CARs and TCRs across 5 different pMHC targets, with a total of 19 constructs examined in vitro. These functional measurements include CAR- and TCR-mediated activation, proliferation, and cytotoxicity in both acute and chronic settings. Surprisingly, we find no consistent difference between CARs and TCRs as receptor classes with respect to their relative sensitivity to major regulators of T cell activation: PD-L1, CD80/86 and IL-2. Though TCRs often emerge from human blood directly as potent, selective receptors, CARs must be heavily optimized to attain these properties for pMHC targets. Nonetheless, when iteratively improved and compared head to head in functional tests, CARs appear remarkably similar to TCRs with respect to immune modulation.


Asunto(s)
Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Humanos
11.
J Immunother ; 44(8): 292-306, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34432728

RESUMEN

Next-generation T-cell therapies will likely continue to utilize T-cell receptors (TCRs) and chimeric antigen receptors (CARs) because each receptor type has advantages. TCRs often possess exceptional properties even when tested unmodified from patients' T cells. CARs are generally less sensitive, possibly because their ligand-binding domains are grafted from antibodies selected for binding affinity or avidity and not broadly optimized for a functional response. Because of the disconnect between binding and function among these receptor types, the ultimate potential of CARs optimized for sensitivity and selectivity is not clear. Here, we focus on a thoroughly studied immuno-oncology target, the HLA-A*02/HPV-E629-38 complex, and show that CARs can be optimized by a combination of high-throughput binding screens and low-throughput functional assays to have comparable activity to clinical TCRs in acute assays in vitro. These results provide a case study for the challenges and opportunities of optimizing high-performing CARs, especially in the context of targets utilized naturally by TCRs.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias/terapia , Infecciones por Papillomavirus/terapia , Receptores Quiméricos de Antígenos/inmunología , Línea Celular , Proteínas Fluorescentes Verdes , Antígeno HLA-A2/inmunología , Humanos , Interferón gamma/inmunología , Luciferasas de Luciérnaga , Neoplasias/inmunología , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/inmunología , Péptidos/inmunología , Proteínas Represoras/inmunología , Anticuerpos de Cadena Única/inmunología
12.
Immunohorizons ; 5(5): 349-359, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039676

RESUMEN

We designed variant human TCRs composed of the full-length TCRα/ß or extracellular and transmembrane domains of the associated CD3 subunits fused to polypeptides derived from proteins thought to either enhance or inhibit normal T cell function. First, we showed that the C termini of both the TCR α- and ß-chains can accommodate specific additional sequences, without abrogating complex formation or acute sensitivity of the receptor. Replacement of ITAMs with ITIM-containing intracellular domains inverted the TCR signal (i.e., created a ligand-dependent inhibitory receptor). The normal signaling function of the CD3 complex was transferable to the TCR by eliminating all CD3 ITAMs and grafting three to six ITAMs onto the C termini of the α/ß-chains, with no effect on acute sensitivity. The observation that TCR variants of such diverse C-terminal composition can fold and function as signaling receptors demonstrates substantial structural and functional malleability of TCRs. These results add to knowledge about TCR structure-function with regard to acute signaling and may provide a route to use TCRs in different ways for T cell therapy.


Asunto(s)
Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Proteínas Portadoras/metabolismo , Humanos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/citología , Linfocitos T/inmunología
13.
J Immunother ; 44(3): 95-105, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33284140

RESUMEN

In 2013, an innovative MAGE-A3-directed cancer therapeutic of great potential value was terminated in the clinic because of neurotoxicity. The safety problems were hypothesized to originate from off-target T-cell receptor activity against a closely related MAGE-A12 peptide. A combination of published and new data led us to test this hypothesis with current technology. Our results call into question MAGE-A12 as the source of the neurotoxicity. Rather, the data imply that an alternative related peptide from EPS8L2 may be responsible. Given the qualities of MAGE-A3 as an onco-testis antigen widely expressed in tumors and largely absent from normal adult tissues, these findings suggest that MAGE-A3 may deserve further consideration as a cancer target. As a step in this direction, the authors isolated 2 MAGE-A3 peptide-major histocompatibility complex-directed chimeric antigen receptors, 1 targeting the same peptide as the clinical T-cell receptor. Both chimeric antigen receptors have improved selectivity over the EPS8L2 peptide that represents a significant risk for MAGE-A3-targeted therapeutics, showing that there may be other options for MAGE-A3 cell therapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Proteínas de Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Línea Celular , Línea Celular Tumoral , Células HCT116 , Células HEK293 , Humanos , Células Jurkat , Leucocitos Mononucleares/inmunología , Células MCF-7 , Complejo Mayor de Histocompatibilidad/inmunología , Neoplasias/inmunología , Células PC-3 , Receptores Quiméricos de Antígenos/inmunología
14.
PLoS One ; 15(11): e0241421, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33166305

RESUMEN

Cell therapy is poised to play a larger role in medicine, most notably for immuno-oncology. Despite the recent success of CAR-T therapeutics in the treatment of blood tumors and the rapid progress toward improved versions of both CAR- and TCR-Ts, important analytical aspects of preclinical development and manufacturing of engineered T cells remain immature. One limiting factor is the absence of robust multivariate assays to disentangle key parameters related to function of engineered effector cells, especially in the peptide-MHC (pMHC) target realm, the natural ligand for TCRs. Here we describe an imaging-based primary T cell assay that addresses several of these limitations. To our knowledge, this assay is the first quantitative, high-content assay that separates the key functional parameters of time- and antigen-dependent T cell proliferation from cytotoxicity. We show that the assay sheds light on relevant biology of CAR- and TCR-T cells, including response kinetics and the influence of effector:target ratio.


Asunto(s)
Inmunoensayo/métodos , Linfocitos T/inmunología , Línea Celular , Proliferación Celular , Citotoxicidad Inmunológica , Humanos , Cinética , Análisis Multivariante , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/citología
15.
Mol Immunol ; 128: 298-310, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33012527

RESUMEN

We describe an approach to cancer therapy based on exploitation of common losses of genetic material in tumor cells (loss of heterozygosity) (Basilion et al., 1999; Beroukhim et al., 2010). This therapeutic concept addresses the fundamental problem of discrimination between tumor and normal cells and can be applied in principle to the large majority of tumors. It utilizes modular activator/blocker elements that integrate signals related to the presence and absence of ligands displayed on the cell surface (Fedorov et al., 2013). We show that the targeting system works robustly in vitro and in a mouse cancer model where absence of the HLA-A*02 allele releases a brake on engineered T cells activated by the CD19 surface antigen. This therapeutic approach potentially opens a route toward a large, new source of cancer targets.


Asunto(s)
Pérdida de Heterocigocidad/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Alelos , Animales , Antígenos CD19/inmunología , Línea Celular Tumoral , Femenino , Antígenos HLA-A/inmunología , Humanos , Células Jurkat , Ligandos , Ratones , Ratones Endogámicos NOD
16.
Mol Immunol ; 126: 56-64, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32768859

RESUMEN

Chimeric antigen receptors (CARs) and their parent signaling molecule, the T cell receptor (TCR), are fascinating proteins of increasing relevance to disease therapy. Here we use a collection of 1221 pMHC-directed CAR constructs representing 10 pMHC targets to study aspects of CAR structure-activity relationships (SAR), with particular focus on the extracellular and transmembrane structural components. These experiments that involve pMHC targets whose number/cell can be manipulated by peptide dosing in vitro enable systematic analysis of the SAR of CARs in carefully controlled experimental situations (Harris and Kranz, 2016). We find that CARs tolerate a wide range of structural variation, with the ligand-binding domains (LBDs) dominating the SAR of CAR antigen sensitivity. Notwithstanding the critical role of the LBD, CAR antigen-binding on the cell surface, measured by pMHC tetramer staining, is not an effective predictor of functional sensitivity. These results have important implications for the design and testing of CARs aimed toward the clinic.


Asunto(s)
Antígenos HLA-A/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Transducción de Señal/inmunología , Linfocitos T/inmunología , Sitios de Unión/inmunología , Antígenos HLA-A/metabolismo , Humanos , Células Jurkat , Ligandos , Células MCF-7 , Dominios Proteicos/inmunología , Multimerización de Proteína/inmunología , Receptores Quiméricos de Antígenos/inmunología , Relación Estructura-Actividad , Linfocitos T/metabolismo
17.
F1000Res ; 9: 1295, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33335718

RESUMEN

T-cell cancer therapy is a clinical field flush with opportunity.  It is part of the revolution in immuno-oncology, most apparent in the dramatic clinical success of PD-1/CTLA-4 antibodies and chimeric antigen receptor T-cells (CAR-Ts) to cure certain melanomas and lymphomas, respectively.  Therapeutics based on T cells ultimately hold more promise because of their capacity to carry out complex behaviors and their ease of modification via genetic engineering.  But to overcome the substantial obstacles of effective solid-tumor treatment, T-cell therapy must access novel molecular targets or exploit existing ones in new ways.  As always, tumor selectivity is the key. T-cell therapy has the potential to address target opportunities afforded by its own unique capacity for signal integration and high sensitivity.  With a history of breathtaking innovation, the scientific foundation for the cellular modality has often been bypassed in favor of rapid advance in the clinic.  This situation is changing, as the mechanistic basis for activity of CAR-Ts and TCR-Ts is backfilled by painstaking, systematic experiments-harking back to last century's evolution and maturation of the small-molecule drug discovery field.   We believe this trend must continue for T-cell therapy to reach its enormous potential.  We support an approach that integrates sound reductionist scientific principles with well-informed, thorough preclinical and translational clinical experiments.


Asunto(s)
Inmunoterapia Adoptiva , Melanoma , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Linfocitos T
18.
Sci Rep ; 9(1): 17291, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31754147

RESUMEN

Cell therapy using T cell receptors (TCRs) and chimeric antigen receptors (CARs) represents a new wave of immunotherapies garnering considerable attention and investment. Further progress in this area of medicine depends in part on improving the functional capabilities of the engineered components, while maintaining the overall size of recombinant constructs to ensure their compatibility with existing gene delivery vehicles. We describe a single-variable-domain TCR (svd TCR) that utilizes only the variable domain of the ß chain (Vß). This Vß module not only works in TCR and CAR formats, but also can be used to create single-chain bispecific CARs and TCRs. Comparison of individual ligand-binding Vß domains in different formats suggests that the lone Vß sequence controls the sensitivity and a major part of the specificity of the CAR or TCR construct, regardless of signaling format, in Jurkat and primary T cells.


Asunto(s)
Región Variable de Inmunoglobulina/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/trasplante , Ingeniería Celular , Células HEK293 , Humanos , Región Variable de Inmunoglobulina/genética , Células Jurkat , Ligandos , Neoplasias/inmunología , Cultivo Primario de Células , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores Quiméricos de Antígenos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Linfocitos T/inmunología , Transfección , Escape del Tumor
19.
F1000Res ; 5: 135, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26998234

RESUMEN

This editorial introduces the Preclinical Reproducibility and Robustness channel on F1000Research, which has been created to encourage and facilitate open and transparent publication and discussion of confirmatory and non-confirmatory studies in biomedical research.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA