Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Immunity ; 56(10): 2388-2407.e9, 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37776850

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy targeting CD19 has achieved tremendous success treating B cell malignancies; however, some patients fail to respond due to poor autologous T cell fitness. To improve response rates, we investigated whether disruption of the co-inhibitory receptors CTLA4 or PD-1 could restore CART function. CRISPR-Cas9-mediated deletion of CTLA4 in preclinical models of leukemia and myeloma improved CAR T cell proliferation and anti-tumor efficacy. Importantly, this effect was specific to CTLA4 and not seen upon deletion of CTLA4 and/or PDCD1 in CAR T cells. Mechanistically, CTLA4 deficiency permitted unopposed CD28 signaling and maintenance of CAR expression on the T cell surface under conditions of high antigen load. In clinical studies, deletion of CTLA4 rescued the function of T cells from patients with leukemia that previously failed CAR T cell treatment. Thus, selective deletion of CTLA4 reinvigorates dysfunctional chronic lymphocytic leukemia (CLL) patient T cells, providing a strategy for increasing patient responses to CAR T cell therapy.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Receptores Quiméricos de Antígenos , Humanos , Receptores de Antígenos de Linfocitos T/metabolismo , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Linfocitos T , Inmunoterapia Adoptiva , Antígenos CD19
2.
Nature ; 607(7918): 360-365, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35676488

RESUMEN

Synthetic receptor signalling has the potential to endow adoptively transferred T cells with new functions that overcome major barriers in the treatment of solid tumours, including the need for conditioning chemotherapy1,2. Here we designed chimeric receptors that have an orthogonal IL-2 receptor extracellular domain (ECD) fused with the intracellular domain (ICD) of receptors for common γ-chain (γc) cytokines IL-4, IL-7, IL-9 and IL-21 such that the orthogonal IL-2 cytokine elicits the corresponding γc cytokine signal. Of these, T cells that signal through the chimeric orthogonal IL-2Rß-ECD-IL-9R-ICD (o9R) are distinguished by the concomitant activation of STAT1, STAT3 and STAT5 and assume characteristics of stem cell memory and effector T cells. Compared to o2R T cells, o9R T cells have superior anti-tumour efficacy in two recalcitrant syngeneic mouse solid tumour models of melanoma and pancreatic cancer and are effective even in the absence of conditioning lymphodepletion. Therefore, by repurposing IL-9R signalling using a chimeric orthogonal cytokine receptor, T cells gain new functions, and this results in improved anti-tumour activity for hard-to-treat solid tumours.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Inmunoterapia Adoptiva , Subunidad gamma Común de Receptores de Interleucina , Neoplasias , Receptores de Interleucina-9 , Proteínas Recombinantes de Fusión , Linfocitos T , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Inmunoterapia Adoptiva/métodos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/inmunología , Interleucinas/genética , Interleucinas/inmunología , Melanoma/inmunología , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias Pancreáticas/inmunología , Receptores de Interleucina-9/genética , Receptores de Interleucina-9/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Factores de Transcripción STAT/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
4.
Nano Lett ; 20(3): 1578-1589, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-31951421

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy relies on the ex vivo manipulation of patient T cells to create potent, cancer-targeting therapies, shown to be capable of inducing remission in patients with acute lymphoblastic leukemia and large B cell lymphoma. However, current CAR T cell engineering methods use viral delivery vectors, which induce permanent CAR expression and could lead to severe adverse effects. Messenger RNA (mRNA) has been explored as a promising strategy for inducing transient CAR expression in T cells to mitigate the adverse effects associated with viral vectors, but it most commonly requires electroporation for T cell mRNA delivery, which can be cytotoxic. Here, ionizable lipid nanoparticles (LNPs) were designed for ex vivo mRNA delivery to human T cells. A library of 24 ionizable lipids was synthesized, formulated into LNPs, and screened for luciferase mRNA delivery to Jurkat cells, revealing seven formulations capable of enhanced mRNA delivery over lipofectamine. The top-performing LNP formulation, C14-4, was selected for CAR mRNA delivery to primary human T cells. This platform induced CAR expression at levels equivalent to electroporation, with substantially reduced cytotoxicity. CAR T cells engineered via C14-4 LNP treatment were then compared to electroporated CAR T cells in a coculture assay with Nalm-6 acute lymphoblastic leukemia cells, and both CAR T cell engineering methods elicited potent cancer-killing activity. These results demonstrate the ability of LNPs to deliver mRNA to primary human T cells to induce functional protein expression, and indicate the potential of LNPs to enhance mRNA-based CAR T cell engineering methods.


Asunto(s)
Ingeniería Celular , Sistemas de Liberación de Medicamentos , Lípidos , Nanopartículas/química , ARN Mensajero , Receptores Quiméricos de Antígenos , Linfocitos T/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Humanos , Lípidos/química , Lípidos/farmacología , ARN Mensajero/química , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Mensajero/farmacología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo
5.
Nat Med ; 27(5): 842-850, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33888899

RESUMEN

While CD19-directed chimeric antigen receptor (CAR) T cells can induce remission in patients with B cell acute lymphoblastic leukemia (ALL), a large subset relapse with CD19- disease. Like CD19, CD22 is broadly expressed by B-lineage cells and thus serves as an alternative immunotherapy target in ALL. Here we present the composite outcomes of two pilot clinical trials ( NCT02588456 and NCT02650414 ) of T cells bearing a 4-1BB-based, CD22-targeting CAR in patients with relapsed or refractory ALL. The primary end point of these studies was to assess safety, and the secondary end point was antileukemic efficacy. We observed unexpectedly low response rates, prompting us to perform detailed interrogation of the responsible CAR biology. We found that shortening of the amino acid linker connecting the variable heavy and light chains of the CAR antigen-binding domain drove receptor homodimerization and antigen-independent signaling. In contrast to CD28-based CARs, autonomously signaling 4-1BB-based CARs demonstrated enhanced immune synapse formation, activation of pro-inflammatory genes and superior effector function. We validated this association between autonomous signaling and enhanced function in several CAR constructs and, on the basis of these observations, designed a new short-linker CD22 single-chain variable fragment for clinical evaluation. Our findings both suggest that tonic 4-1BB-based signaling is beneficial to CAR function and demonstrate the utility of bedside-to-bench-to-bedside translation in the design and implementation of CAR T cell therapies.


Asunto(s)
Ligando 4-1BB/metabolismo , Inmunoterapia Adoptiva/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores Quiméricos de Antígenos/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Linfocitos T/trasplante , Adulto , Animales , Antígenos CD19/metabolismo , Linfocitos B/inmunología , Antígenos CD28/genética , Células Cultivadas , Niño , Preescolar , Femenino , Humanos , Masculino , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Exp Med ; 217(12)2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-32860705

RESUMEN

Adoptive T cell therapy (ACT) with genetically modified T cells has shown impressive results against some hematologic cancers, but efficacy in solid tumors can be limited by restrictive tumor microenvironments (TMEs). For example, Fas ligand is commonly overexpressed in TMEs and induces apoptosis in tumor-infiltrating, Fas receptor-positive lymphocytes. We engineered immunomodulatory fusion proteins (IFPs) to enhance ACT efficacy, combining an inhibitory receptor ectodomain with a costimulatory endodomain to convert negative into positive signals. We developed a Fas-4-1BB IFP that replaces the Fas intracellular tail with costimulatory 4-1BB. Fas-4-1BB IFP-engineered murine T cells exhibited increased pro-survival signaling, proliferation, antitumor function, and altered metabolism in vitro. In vivo, Fas-4-1BB ACT eradicated leukemia and significantly improved survival in the aggressive KPC pancreatic cancer model. Fas-4-1BB IFP expression also enhanced primary human T cell function in vitro. Thus, Fas-4-1BB IFP expression is a novel strategy to improve multiple T cell functions and enhance ACT against solid tumors and hematologic malignancies.


Asunto(s)
Inmunoterapia Adoptiva , Proteínas Recombinantes de Fusión/farmacología , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Receptor fas/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Ingeniería Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Factores Inmunológicos/farmacología , Leucemia/inmunología , Leucemia/patología , Leucemia/terapia , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Fenotipo , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos
7.
Cancer Discov ; 10(4): 552-567, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32001516

RESUMEN

Primary resistance to CD19-directed chimeric antigen receptor T-cell therapy (CART19) occurs in 10% to 20% of patients with acute lymphoblastic leukemia (ALL); however, the mechanisms of this resistance remain elusive. Using a genome-wide loss-of-function screen, we identified that impaired death receptor signaling in ALL led to rapidly progressive disease despite CART19 treatment. This was mediated by an inherent resistance to T-cell cytotoxicity that permitted antigen persistence and was subsequently magnified by the induction of CAR T-cell functional impairment. These findings were validated using samples from two CAR T-cell clinical trials in ALL, where we found that reduced expression of death receptor genes was associated with worse overall survival and reduced T-cell fitness. Our findings suggest that inherent dysregulation of death receptor signaling in ALL directly leads to CAR T-cell failure by impairing T-cell cytotoxicity and promoting progressive CAR T-cell dysfunction. SIGNIFICANCE: Resistance to CART19 is a significant barrier to efficacy in the treatment of B-cell malignancies. This work demonstrates that impaired death receptor signaling in tumor cells causes failed CART19 cytotoxicity and drives CART19 dysfunction, identifying a novel mechanism of antigen-independent resistance to CAR therapy.See related commentary by Green and Neelapu, p. 492.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores Quiméricos de Antígenos/metabolismo , Receptores de Muerte Celular/metabolismo , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Transducción de Señal
8.
Curr Drug Discov Technol ; 11(4): 239-42, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25373791

RESUMEN

P. falciparum is highly virulent in nature because of its ability to modify the infected host red blood cells, adherence to the vascular endothelium and changes in antigenicity at different stages. Also slow migration time in the dermal and endothelial cells leads to decreased immune response. To overcome the problems, there is a need to design a vaccine which increases the migration time of the parasite, enhances the immune response, enables recognition of surface antigens and causes minimal clinical infection as a side-effect. An ITI-based (Infection-Treatment Immunization) vaccine development strategy is to be adopted to develop this novel vaccine. This will include administration of a liquid solution of purified, non-attenuated sporozoites from an infected female Anopheles mosquito, AS02A adjuvant and chlorate (a metabolic inhibitor of sulfation that decreases the extent of GAG sulfation). To control infection, a drug-cover of artemisinin will be administered as a part of the vaccination strategy along with a specific protease inhibitor MRT12113 which prevents RBC rupture and reinvasion by the parasite. This vaccine will intend to increase the overall migration time of the parasite in blood which is otherwise approximately 30 minutes, resulting in an overall enhanced immune response. It also intends to reduce parasite invasion in cells and their consequent rupture thus preventing the clinical condition-malaria.


Asunto(s)
Benzopiranos/administración & dosificación , Catecoles/administración & dosificación , Diseño de Fármacos , Quimioterapia Combinada/métodos , Inmunoterapia Activa/métodos , Vacunas contra la Malaria , Malaria Falciparum/tratamiento farmacológico , Adyuvantes Inmunológicos/administración & dosificación , Animales , Artemisininas/administración & dosificación , Artemisininas/farmacología , Benzopiranos/farmacología , Catecoles/farmacología , Movimiento Celular/efectos de los fármacos , Cloratos/administración & dosificación , Cloratos/farmacología , Femenino , Humanos , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/parasitología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/farmacología , Esporozoítos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA