Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Sci Rep ; 10(1): 6919, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32332814

RESUMEN

To accelerate the cardiac drug discovery pipeline, we set out to develop a platform that would be capable of quantifying tissue-level functions such as contractile force and be amenable to standard multiwell-plate manipulations. We report a 96-well-based array of 3D human pluripotent stem cell (hPSC)-derived cardiac microtissues - termed Cardiac MicroRings (CaMiRi) - in custom 3D-print-molded multiwell plates capable of contractile force measurement. Within each well, two elastomeric microcantilevers are situated above a circumferential ramp. The wells are seeded with cell-laden collagen, which, in response to the gradual slope of the circumferential ramp, self-organizes around tip-gated microcantilevers to form contracting CaMiRi. The contractile force exerted by the CaMiRi is measured and calculated using the deflection of the cantilevers. Platform responses were robust and comparable across wells, and we used it to determine an optimal tissue formulation. We validated the contractile force response of CaMiRi using selected cardiotropic compounds with known effects. Additionally, we developed automated protocols for CaMiRi seeding, image acquisition, and analysis to enable the measurement of contractile force with increased throughput. The unique tissue fabrication properties of the platform, and the consequent effects on tissue function, were demonstrated upon adding hPSC-derived epicardial cells to the system. This platform represents an open-source contractile force screening system useful for drug screening and tissue engineering applications.


Asunto(s)
Células Madre Pluripotentes/citología , Ingeniería de Tejidos/métodos , Animales , Automatización , Cardiotónicos/farmacología , Células Cultivadas , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , Ratones , Contracción Miocárdica/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Impresión Tridimensional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...