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










Base de dados
Intervalo de ano de publicação
1.
Cytotherapy ; 25(6): 670-682, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36849306

RESUMO

BACKGROUND AIMS: Chimeric antigen receptor (CAR) T cells have demonstrated remarkable efficacy against hematological malignancies; however, they have not experienced the same success against solid tumors such as glioblastoma (GBM). There is a growing need for high-throughput functional screening platforms to measure CAR T-cell potency against solid tumor cells. METHODS: We used real-time, label-free cellular impedance sensing to evaluate the potency of anti-disialoganglioside (GD2) targeting CAR T-cell products against GD2+ patient-derived GBM stem cells over a period of 2 days and 7 days in vitro. We compared CAR T products using two different modes of gene transfer: retroviral transduction and virus-free CRISPR-editing. Endpoint flow cytometry, cytokine analysis and metabolomics data were acquired and integrated to create a predictive model of CAR T-cell potency. RESULTS: Results indicated faster cytolysis by virus-free CRISPR-edited CAR T cells compared with retrovirally transduced CAR T cells, accompanied by increased inflammatory cytokine release, CD8+ CAR T-cell presence in co-culture conditions and CAR T-cell infiltration into three-dimensional GBM spheroids. Computational modeling identified increased tumor necrosis factor α concentrations with decreased glutamine, lactate and formate as being most predictive of short-term (2 days) and long-term (7 days) CAR T cell potency against GBM stem cells. CONCLUSIONS: These studies establish impedance sensing as a high-throughput, label-free assay for preclinical potency testing of CAR T cells against solid tumors.


Assuntos
Glioblastoma , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Linfócitos T CD8-Positivos , Anticorpos , Citocinas , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T
3.
J Immunother Cancer ; 10(9)2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36382633

RESUMO

BACKGROUND: Chimeric antigen receptor (CAR) T cells have demonstrated high clinical response rates against hematological malignancies (e.g., CD19+ cancers) but have shown limited activity in patients with solid tumors. Recent work showed that precise insertion of a CAR at a defined locus improves treatment outcomes in the context of a CD19 CAR; however, it is unclear if such a strategy could also affect outcomes in solid tumors. Furthermore, CAR manufacturing generally relies on viral vectors for gene delivery, which comprise a complex and resource-intensive part of the manufacturing supply chain. METHODS: Anti-GD2 CAR T cells were generated using CRISPR/Cas9 within 9 days using recombinant Cas9 protein and nucleic acids, without any viral vectors. The CAR was specifically targeted to the T cell receptor alpha constant gene (TRAC). T cell products were characterized at the level of the genome, transcriptome, proteome, and secretome using CHANGE-seq, targeted next-generation sequencing, scRNA-seq, spectral cytometry, and ELISA assays, respectively. Functionality was evaluated in vivo in an NSG™ xenograft neuroblastoma model. RESULTS: In comparison to retroviral CAR T cells, virus-free CRISPR CAR (VFC-CAR) T cells exhibit TRAC-targeted genomic integration of the CAR transgene, elevation of transcriptional and protein characteristics associated with a memory-like phenotype, and low tonic signaling prior to infusion arising in part from the knockout of the T cell receptor. On exposure to the GD2 target antigen, anti-GD2 VFC-CAR T cells exhibit specific cytotoxicity against GD2+ cells in vitro and induce solid tumor regression in vivo. VFC-CAR T cells demonstrate robust homing and persistence and decreased exhaustion relative to retroviral CAR T cells against a human neuroblastoma xenograft model. CONCLUSIONS: This study leverages virus-free genome editing technology to generate CAR T cells featuring a TRAC-targeted CAR, which could inform manufacturing of CAR T cells to treat cancers, including solid tumors.


Assuntos
Imunoterapia Adotiva , Neuroblastoma , Humanos , Gangliosídeos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Receptores de Antígenos de Linfócitos T , Antígenos CD19 , Linfócitos T , Neuroblastoma/patologia
4.
Nat Biomed Eng ; 5(1): 77-88, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32719514

RESUMO

The function of a T cell depends on its subtype and activation state. Here, we show that imaging of the autofluorescence lifetime signals of quiescent and activated T cells can be used to classify the cells. T cells isolated from human peripheral blood and activated in culture using tetrameric antibodies against the surface ligands CD2, CD3 and CD28 showed specific activation-state-dependent patterns of autofluorescence lifetime. Logistic regression models and random forest models classified T cells according to activation state with 97-99% accuracy, and according to activation state (quiescent or activated) and subtype (CD3+CD8+ or CD3+CD4+) with 97% accuracy. Autofluorescence lifetime imaging can be used to non-destructively determine T-cell function.


Assuntos
Ativação Linfocitária/fisiologia , Imagem Óptica/métodos , Linfócitos T , Células Cultivadas , Humanos , Linfócitos T/classificação , Linfócitos T/citologia , Linfócitos T/fisiologia
5.
Biophys J ; 118(9): 2086-2102, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-31699335

RESUMO

Reprogramming of human somatic cells to induced pluripotent stem cells (iPSCs) generates valuable resources for disease modeling, toxicology, cell therapy, and regenerative medicine. However, the reprogramming process can be stochastic and inefficient, creating many partially reprogrammed intermediates and non-reprogrammed cells in addition to fully reprogrammed iPSCs. Much of the work to identify, evaluate, and enrich for iPSCs during reprogramming relies on methods that fix, destroy, or singularize cell cultures, thereby disrupting each cell's microenvironment. Here, we develop a micropatterned substrate that allows for dynamic live-cell microscopy of hundreds of cell subpopulations undergoing reprogramming while preserving many of the biophysical and biochemical cues within the cells' microenvironment. On this substrate, we were able to both watch and physically confine cells into discrete islands during the reprogramming of human somatic cells from skin biopsies and blood draws obtained from healthy donors. Using high-content analysis, we identified a combination of eight nuclear characteristics that can be used to generate a computational model to predict the progression of reprogramming and distinguish partially reprogrammed cells from those that are fully reprogrammed. This approach to track reprogramming in situ using micropatterned substrates could aid in biomanufacturing of therapeutically relevant iPSCs and be used to elucidate multiscale cellular changes (cell-cell interactions as well as subcellular changes) that accompany human cell fate transitions.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas , Diferenciação Celular , Humanos
6.
Biotechnol J ; 13(2)2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28840981

RESUMO

The next generation of therapeutic products to be approved for the clinic is anticipated to be cell therapies, termed "living drugs" for their capacity to dynamically and temporally respond to changes during their production ex vivo and after their administration in vivo. Genetically engineered chimeric antigen receptor (CAR) T cells have rapidly developed into powerful tools to harness the power of immune system manipulation against cancer. Regulatory agencies are beginning to approve CAR T cell therapies due to their striking efficacy in treating some hematological malignancies. However, the engineering and manufacturing of such cells remains a challenge for widespread adoption of this technology. Bioengineering approaches including biomaterials, synthetic biology, metabolic engineering, process control and automation, and in vitro disease modeling could offer promising methods to overcome some of these challenges. Here, we describe the manufacturing process of CAR T cells, highlighting potential roles for bioengineers to partner with biologists and clinicians to advance the manufacture of these complex cellular products under rigorous regulatory and quality control.


Assuntos
Bioengenharia , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/citologia , Terapia Baseada em Transplante de Células e Tecidos , Técnicas de Transferência de Genes , Humanos , Neoplasias/terapia , Controle de Qualidade , Receptores de Antígenos de Linfócitos T/metabolismo
7.
Tissue Eng Part A ; 24(3-4): 254-263, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28610471

RESUMO

Low-intensity pulsed ultrasound (LIPUS) has been shown to be effective for orthopedic fracture repair and nonunion defects, but the specific mechanism behind its efficacy is still unknown. Previously, we have shown a measurable acoustic radiation force at LIPUS intensities traditionally used for clinical treatment and have applied this force to osteoblastic cells encapsulated in type I collagen hydrogels. Our goal in this study is to provide insight and inform the appropriate design of a cell therapy approach to bone repair in which osteoblasts are embedded in collagen hydrogels, implanted into a bony defect, and then transdermally stimulated using LIPUS-derived acoustic radiation force to enhance bone formation at the earliest time points after bone defect repair. To this end, in this study, we demonstrate the ability to measure local hydrogel deformations in response to LIPUS-induced acoustic radiation force and reveal that hydrogel deformation varies with both LIPUS intensity and hydrogel stiffness. Specifically, hydrogel deformation is positively correlated with LIPUS intensity and this deformation is further increased by reducing the stiffness of the hydrogel. We have also shown that encapsulated osteoblastic cells respond to increases in LIPUS intensity by upregulating both cyclooxygenase 2 and prostaglandin E2 (PGE2), both implicated in new bone formation and well-established responses to the application of fluid forces on osteoblast cells. Finally, we demonstrate that combining an increase in LIPUS with a three-dimensional culture environment upregulates both markers beyond their expression noted from either experimental condition alone, suggesting that both LIPUS and hydrogel encapsulation, when combined and modulated appropriately, can enhance osteoblastic response considerably. These studies provide important information toward a clinically relevant cell therapy treatment for bone defects that allows the transdermal application of mechanical loading to bone defects without physically destabilizing the defect site.


Assuntos
Hidrogel de Polietilenoglicol-Dimetacrilato/química , Osteoblastos/citologia , Ultrassonografia/métodos , Animais , Linhagem Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Módulo de Elasticidade , Análise de Elementos Finitos , Consolidação da Fratura/fisiologia , Camundongos , Osteoblastos/efeitos da radiação , Osteogênese/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...