Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 11(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36750252

RESUMEN

BACKGROUND: Glioma-induced immune dysregulation of the hematopoietic system has been described in a limited number of studies. In this study, our group further demonstrates that gliomas interrupt the cellular differentiation programming and outcomes of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow. HSPCs from glioma-bearing mice are reprogrammed and driven towards expansion of myeloid lineage precursors and myeloid-derived suppressor cells (MDSCs) in secondary lymphoid organs. However, we found this expansion is reversed by immunotherapy. Adoptive cellular therapy (ACT) has been demonstrably efficacious in multiple preclinical models of central nervous system (CNS) malignancies, and here we describe how glioma-induced dysfunction is reversed by this immunotherapeutic platform. METHODS: The impact of orthotopic KR158B-luc glioma on HSPCs was evaluated in an unbiased fashion using single cell RNAseq (scRNAseq) of lineage- cells and phenotypically using flow cytometry. Mature myeloid cell frequencies and function were also evaluated using flow cytometry. Finally, ACT containing total body irradiation, tumor RNA-pulsed dendritic cells, tumor-reactive T cells and HSPCs isolated from glioma-bearing or non-tumor-bearing mice were used to evaluate cell fate differentiation and survival. RESULTS: Using scRNAseq, we observed an altered HSPC landscape in glioma-bearing versus non-tumor-bearing mice . In addition, an expansion of myeloid lineage subsets, including granulocyte macrophage precursors (GMPs) and MDSCs, were observed in glioma-bearing mice relative to non-tumor-bearing controls. Furthermore, MDSCs from glioma-bearing mice demonstrated increased suppressive capacity toward tumor-specific T cells as compared with MDSCs from non-tumor-bearing hosts. Interestingly, treatment with ACT overcame these suppressive properties. When HSPCs from glioma-bearing mice were transferred in the context of ACT, we observed significant survival benefit and long-term cures in orthotopic glioma models compared with mice treated with ACT using non-glioma-bearing HSPCs.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Glioma , Ratones , Animales , Línea Celular Tumoral , Glioma/patología , Inmunoterapia , Células Madre Hematopoyéticas , Linfocitos T
2.
ACS Chem Neurosci ; 14(2): 235-245, 2023 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-36571847

RESUMEN

Aggregated α-synuclein, a major constituent of Lewy bodies plays a crucial role in the pathogenesis of α-synucleinopathies (SPs) such as Parkinson's disease (PD). PD is affected by the innate and adaptive arms of the immune system, and recently both active and passive immunotherapies targeted against α-synuclein are being trialed as potential novel treatment strategies. Specifically, dendritic cell-based vaccines have shown to be an effective treatment for SPs in animal models. Here, we report on the development of adoptive cellular therapy (ACT) for SP and demonstrate that adoptive transfer of pre-activated T-cells generated from immunized mice can improve survival and behavior, reduce brain microstructural impairment via magnetic resonance imaging (MRI), and decrease α-synuclein pathology burden in a peripherally induced preclinical SP model (M83) when administered prior to disease onset. This study provides preclinical evidence for ACT as a potential immunotherapy for LBD, PD and other related SPs, and future work will provide necessary understanding of the mechanisms of its action.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Vacunas , Ratones , Animales , alfa-Sinucleína/genética , Sinucleinopatías/patología , Ratones Transgénicos , Enfermedad de Parkinson/terapia , Enfermedad de Parkinson/patología , Modelos Animales de Enfermedad
3.
Nat Cancer ; 3(1): 11-24, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35121998

RESUMEN

Pediatric central nervous system tumors are the most common solid malignancies in childhood, and aggressive therapy often leads to long-term sequelae in survivors, making these tumors challenging to treat. Immunotherapy has revolutionized prospects for many cancer types in adults, but the intrinsic complexity of treating pediatric patients and the scarcity of clinical studies of children to inform effective approaches have hampered the development of effective immunotherapies in pediatric settings. Here, we review recent advances and ongoing challenges in pediatric brain cancer immunotherapy, as well as considerations for efficient clinical translation of efficacious immunotherapies into pediatric settings.


Asunto(s)
Neoplasias Encefálicas , Neoplasias del Sistema Nervioso Central , Neoplasias Encefálicas/terapia , Neoplasias del Sistema Nervioso Central/terapia , Niño , Humanos , Factores Inmunológicos , Inmunoterapia/efectos adversos , Sobrevivientes
4.
Cells ; 10(5)2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33919157

RESUMEN

Our understanding of the relationship between the immune system and cancers has undergone significant discovery recently. Immunotherapy with T cell therapies and checkpoint blockade has meaningfully changed the oncology landscape. While remarkable clinical advances in adaptive immunity are occurring, modulation of innate immunity has proven more difficult. The myeloid compartment, including macrophages, neutrophils, and dendritic cells, has a significant impact on the persistence or elimination of tumors. Myeloid cells, specifically in the tumor microenvironment, have direct contact with tumor tissue and coordinate with tumor-reactive T cells to either stimulate or antagonize cancer immunity. However, the myeloid compartment comprises a broad array of cells in various stages of development. In addition, hematopoietic stem and progenitor cells at various stages of myelopoiesis in distant sites undergo significant modulation by tumors. Understanding how tumors exert their influence on myeloid progenitors is critical to making clinically meaningful improvements in these pathways. Therefore, this review will cover recent developments in our understanding of how solid tumors modulate myelopoiesis to promote the formation of pro-tumor immature myeloid cells. Then, it will cover some of the potential avenues for capitalizing on these mechanisms to generate antitumor immunity.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Inmunoterapia/métodos , Células Mieloides/inmunología , Mielopoyesis/inmunología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Animales , Células Madre Hematopoyéticas/citología , Humanos , Factores Inmunológicos , Células Mieloides/citología
5.
Clin Cancer Res ; 26(21): 5689-5700, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32788225

RESUMEN

PURPOSE: Immunotherapy has been demonstrably effective against multiple cancers, yet tumor escape is common. It remains unclear how brain tumors escape immunotherapy and how to overcome this immune escape. EXPERIMENTAL DESIGN: We studied KR158B-luc glioma-bearing mice during treatment with adoptive cellular therapy (ACT) with polyclonal tumor-specific T cells. We tested the immunogenicity of primary and escaped tumors using T-cell restimulation assays. We used flow cytometry and RNA profiling of whole tumors to further define escape mechanisms. To treat immune-escaped tumors, we generated escape variant-specific T cells through the use of escape variant total tumor RNA and administered these cells as ACT. In addition, programmed cell death protein-1 (PD-1) checkpoint blockade was studied in combination with ACT. RESULTS: Escape mechanisms included a shift in immunogenic tumor antigens, downregulation of MHC class I, and upregulation of checkpoint molecules. Polyclonal T cells specific for escape variants displayed greater recognition of escaped tumors than primary tumors. When administered as ACT, these T cells prolonged median survival of escape variant-bearing mice by 60%. The rational combination of ACT with PD-1 blockade prolonged median survival of escape variant glioma-bearing mice by 110% and was dependent upon natural killer cells and T cells. CONCLUSIONS: These findings suggest that the immune landscape of brain tumors are markedly different postimmunotherapy yet can still be targeted with immunotherapy.


Asunto(s)
Glioma/terapia , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T/inmunología , Escape del Tumor/efectos de los fármacos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Glioma/genética , Glioma/inmunología , Glioma/patología , Xenoinjertos , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia Adoptiva/efectos adversos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Escape del Tumor/inmunología , Microambiente Tumoral/efectos de los fármacos
6.
Nat Commun ; 10(1): 3029, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31292444

RESUMEN

With improving biofabrication technology, 3D bioprinted constructs increasingly resemble real tissues. However, the fundamental principles describing how cell-generated forces within these constructs drive deformations, mechanical instabilities, and structural failures have not been established, even for basic biofabricated building blocks. Here we investigate mechanical behaviours of 3D printed microbeams made from living cells and extracellular matrix, bioprinting these simple structural elements into a 3D culture medium made from packed microgels, creating a mechanically controlled environment that allows the beams to evolve under cell-generated forces. By varying the properties of the beams and the surrounding microgel medium, we explore the mechanical behaviours exhibited by these structures. We observe buckling, axial contraction, failure, and total static stability, and we develop mechanical models of cell-ECM microbeam mechanics. We envision these models and their generalizations to other fundamental 3D shapes to facilitate the predictable design of biofabricated structures using simple building blocks in the future.


Asunto(s)
Bioimpresión/métodos , Técnicas de Cultivo de Célula/métodos , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Resinas Acrílicas/química , Animales , Materiales Biocompatibles , Línea Celular Tumoral , Matriz Extracelular , Geles/química , Ensayo de Materiales , Metacrilatos/química , Ratones , Células 3T3 NIH
7.
Stem Cells ; 37(2): 166-175, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30353618

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are the progenitor cells that can regenerate the entire blood compartment, including the immune system. Recent studies have unearthed considerable immune-modulating potential of these cells. They can migrate through chemotactic gradients, differentiate into functional immune cells, and crosstalk with immune cells during infections, autoimmune diseases, and cancers. Although the primary role of HSPCs during solid malignancies is considered immunosuppressive, recent studies have discovered immune-activating HSPCs and progeny. In this review, we will discuss the recent evidence that HSPCs act as immunomodulators during solid cancers and highlight the future directions of discovery. Stem Cells 2019;37:166-175.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Inmunidad/inmunología , Inmunoterapia/métodos , Neoplasias/inmunología , Células Madre/inmunología
8.
Nat Commun ; 9(1): 4313, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30333482

RESUMEN

Immune checkpoint blockade using anti-PD-1 monoclonal antibodies has shown considerable promise in the treatment of solid tumors, but brain tumors remain notoriously refractory to treatment. In CNS malignancies that are completely resistant to PD-1 blockade, we found that bone marrow-derived, lineage-negative hematopoietic stem and progenitor cells (HSCs) that express C-C chemokine receptor type 2 (CCR2+) reverses treatment resistance and sensitizes mice to curative immunotherapy. HSC transfer with PD-1 blockade increases T-cell frequency and activation within tumors in preclinical models of glioblastoma and medulloblastoma. CCR2+HSCs preferentially migrate to intracranial brain tumors and differentiate into antigen-presenting cells within the tumor microenvironment and cross-present tumor-derived antigens to CD8+ T cells. HSC transfer also rescues tumor resistance to adoptive cellular therapy in medulloblastoma and glioblastoma. Our studies demonstrate a novel role for CCR2+HSCs in overcoming brain tumor resistance to PD-1 checkpoint blockade and adoptive cellular therapy in multiple invasive brain tumor models.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Trasplante de Células Madre Hematopoyéticas , Inmunoterapia Adoptiva , Meduloblastoma/terapia , Animales , Neoplasias Encefálicas/inmunología , Diferenciación Celular , Movimiento Celular , Células Dendríticas/inmunología , Resistencia a Antineoplásicos , Femenino , Glioblastoma/inmunología , Activación de Linfocitos , Meduloblastoma/inmunología , Ratones Transgénicos , Linfocitos T/fisiología
9.
Clin Cancer Res ; 24(16): 3955-3966, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29712687

RESUMEN

Purpose: Adoptive T-cell immunotherapy (ACT) has emerged as a viable therapeutic for peripheral and central nervous system (CNS) tumors. In peripheral cancers, optimal efficacy of ACT is reliant on dendritic cells (DCs) in the tumor microenvironment. However, the CNS is largely devoid of resident migratory DCs to function as antigen-presenting cells during immunotherapy. Herein, we demonstrate that cellular interactions between adoptively transferred tumor-reactive T cells and bone marrow-derived hematopoietic stem and progenitor cells (HSPCs) lead to the generation of potent intratumoral DCs within the CNS compartment.Experimental Design: We evaluated HSPC differentiation during ACT in vivo in glioma-bearing hosts and HSPC proliferation and differentiation in vitro using a T-cell coculture system. We utilized FACS, ELISAs, and gene expression profiling to study the phenotype and function of HSPC-derived cells ex vivo and in vivo To demonstrate the impact of HSPC differentiation and function on antitumor efficacy, we performed survival experiments.Results: Transfer of HSPCs with concomitant ACT led to the production of activated CD86+CD11c+MHCII+ cells consistent with DC phenotype and function within the brain tumor microenvironment. These intratumoral DCs largely supplanted abundant host myeloid-derived suppressor cells. We determined that during ACT, HSPC-derived cells in gliomas rely on T-cell-released IFNγ to differentiate into DCs, activate T cells, and reject intracranial tumors.Conclusions: Our data support the use of HSPCs as a novel cellular therapy. Although DC vaccines induce robust immune responses in the periphery, our data demonstrate that HSPC transfer uniquely generates intratumoral DCs that potentiate T-cell responses and promote glioma rejection in situClin Cancer Res; 24(16); 3955-66. ©2018 AACR.


Asunto(s)
Neoplasias del Sistema Nervioso Central/terapia , Glioma/terapia , Células Madre Hematopoyéticas/inmunología , Inmunoterapia Adoptiva , Animales , Antígeno B7-2/inmunología , Antígeno CD11c/inmunología , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/inmunología , Neoplasias del Sistema Nervioso Central/patología , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Glioma/genética , Glioma/inmunología , Glioma/patología , Células Madre Hematopoyéticas/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/inmunología
10.
Clin Cancer Res ; 20(4): 972-84, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24352643

RESUMEN

PURPOSE: Chimeric antigen receptor (CAR) transduced T cells represent a promising immune therapy that has been shown to successfully treat cancers in mice and humans. However, CARs targeting antigens expressed in both tumors and normal tissues have led to significant toxicity. Preclinical studies have been limited by the use of xenograft models that do not adequately recapitulate the immune system of a clinically relevant host. A constitutively activated mutant of the naturally occurring epidermal growth factor receptor (EGFRvIII) is antigenically identical in both human and mouse glioma, but is also completely absent from any normal tissues. EXPERIMENTAL DESIGN: We developed a third-generation, EGFRvIII-specific murine CAR (mCAR), and performed tests to determine its efficacy in a fully immunocompetent mouse model of malignant glioma. RESULTS: At elevated doses, infusion with EGFRvIII mCAR T cells led to cures in all mice with brain tumors. In addition, antitumor efficacy was found to be dependent on lymphodepletive host conditioning. Selective blockade with EGFRvIII soluble peptide significantly abrogated the activity of EGFRvIII mCAR T cells in vitro and in vivo, and may offer a novel strategy to enhance the safety profile for CAR-based therapy. Finally, mCAR-treated, cured mice were resistant to rechallenge with EGFRvIII(NEG) tumors, suggesting generation of host immunity against additional tumor antigens. CONCLUSION: All together, these data support that third-generation, EGFRvIII-specific mCARs are effective against gliomas in the brain and highlight the importance of syngeneic, immunocompetent models in the preclinical evaluation of tumor immunotherapies.


Asunto(s)
Traslado Adoptivo , Astrocitoma/terapia , Neoplasias Encefálicas/terapia , Receptores ErbB/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Astrocitoma/inmunología , Astrocitoma/metabolismo , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Receptores ErbB/inmunología , Humanos , Ratones , Trasplante de Neoplasias , Anticuerpos de Cadena Única/metabolismo , Linfocitos T/inmunología , Linfocitos T/trasplante
11.
J Appl Physiol (1985) ; 113(2): 263-72, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22604887

RESUMEN

The purpose of this study is to investigate the effects of exercise on cancer progression, metastasis, and underlying mechanisms in an orthotopic model of murine prostate cancer. C57BL/6 male mice (6-8 wk of age) were orthotopically injected with transgenic adenocarcinoma of mouse prostate C-1 cells (5 × 10(5)) and randomly assigned to exercise (n = 28) or a non-intervention control (n = 31) groups. The exercise group was given voluntary access to a wheel 24 h/day for the duration of the study. Four mice per group were serially killed on days 14, 31, and 36; the remaining 38 mice (exercise, n = 18; control, n = 20) were killed on day 53. Before death, MRI was performed to assess tumor blood perfusion. Primary tumor growth rate was comparable between groups, but expression of prometastatic genes was significantly modulated in exercising animals with a shift toward reduced metastasis. Exercise was associated with increased activity of protein kinases within the MEK/MAPK and PI3K/mTOR signaling cascades with subsequent increased intratumoral protein levels of HIF-1α and VEGF. This was associated with improved tumor vascularization. Multiplex ELISAs revealed distinct reductions in plasma concentrations of several angiogenic cytokines in the exercise group, which was associated with increased expression of angiogenic and metabolic genes in the skeletal muscle. Exercise-induced stabilization of HIF-1α and subsequent upregulation of VEGF was associated with "productive" tumor vascularization with a shift toward suppressed metastasis in an orthotopic model of prostate cancer.


Asunto(s)
Adenocarcinoma/fisiopatología , Adenocarcinoma/secundario , Citocinas/sangre , Terapia por Ejercicio/métodos , Neovascularización Patológica/fisiopatología , Condicionamiento Físico Animal/métodos , Neoplasias de la Próstata/fisiopatología , Adenocarcinoma/prevención & control , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales , Neovascularización Patológica/prevención & control , Neoplasias de la Próstata/prevención & control , Neoplasias de la Próstata/secundario , Resultado del Tratamiento
12.
Cancer Res ; 70(22): 9329-38, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20884629

RESUMEN

Whole-brain irradiation (WBI) therapy produces progressive learning and memory deficits in patients with primary or secondary brain tumors. Exercise enhances memory and adult hippocampal neurogenesis in the intact brain, so we hypothesized that exercise may be an effective treatment to alleviate consequences of WBI. Previous studies using animal models to address this issue have yielded mixed results and have not examined potential molecular mechanisms. We investigated the short- and long-term effects of WBI on spatial learning and memory retention and determined whether voluntary running after WBI aids recovery of brain and cognitive function. Forty adult female C57Bl/6 mice given a single dose of 5 Gy or sham WBI were trained 2.5 weeks and up to 4 months after WBI in a Barnes maze. Half of the mice received daily voluntary wheel access starting 1 month after sham or WBI. Daily running following WBI prevented the marked decline in spatial memory retention observed months after irradiation. Bromodeoxyuridine (BrdUrd) immunolabeling and enzyme-linked immunosorbent assay indicated that this behavioral rescue was accompanied by a partial restoration of newborn BrdUrd+/NeuN+ neurons in the dentate gyrus and increased hippocampal expression of brain-derived vascular endothelial growth factor and insulin-like growth factor-1, and occurred despite irradiation-induced elevations in hippocampal proinflammatory cytokines. WBI in adult mice produced a progressive memory decline consistent with what has been reported in cancer patients receiving WBI therapy. Our findings show that running can abrogate this memory decline and aid recovery of adult hippocampal plasticity, thus highlighting exercise as a potential therapeutic intervention.


Asunto(s)
Irradiación Craneana/efectos adversos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Memoria/fisiología , Neurogénesis/fisiología , Carrera/fisiología , Animales , Bromodesoxiuridina/metabolismo , Citocinas/metabolismo , Giro Dentado/citología , Giro Dentado/metabolismo , Terapia por Ejercicio , Femenino , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Hipocampo/efectos de la radiación , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/etiología , Trastornos de la Memoria/fisiopatología , Trastornos de la Memoria/prevención & control , Ratones , Ratones Endogámicos C57BL , Condicionamiento Físico Animal/fisiología , Dosis de Radiación , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...