Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Neurooncol ; 163(3): 635-645, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37354357

RESUMO

PURPOSE: Medulloblastomas (MBs) constitute the most common malignant brain tumor in children and adolescents. MYC-amplified Group 3 MBs are characterized by disease recurrence, specifically in the leptomeninges, whereby patients with these metastatic tumors have a mortality rate nearing 100%. Despite limited research on such tumors, studies on MB metastases at diagnosis suggest targeting kinases to be beneficial. METHODS: To identify kinase inhibitors that eradicate cells driving therapy evasion and tumor dissemination, we utilized our established patient-derived xenograft (PDX) mouse-adapted therapy platform that models human MB metastatic recurrences following standard chemoradiotherapy. High-throughput screens of 640 kinase inhibitors were conducted against cells isolated from mouse spines in the PDX model and human fetal neural stem cells to reveal compounds that targeted these treatment-refractory, metastatic cells, whilst sparing healthy cells. Blood-brain barrier permeability assays and additional in vitro experimentation helped select top candidates for in vivo studies. RESULTS: Recurrent Group 3 MB PDX spine cells were therapeutically vulnerable to a selective checkpoint kinase 1 (CHK1) inhibitor and small molecular inhibitor of platelet-derived growth factor receptor beta (PDGFRß). Inhibitor-treated cells showed a significant reduction in MB stem cell properties associated with treatment failure. Mice also demonstrated survival advantage when treated with a CHK1 inhibitor ex vivo. CONCLUSION: We identified CHK1 and PDGFRß inhibitors that effectively target MB cells fueling treatment-refractory metastases. With limited research on effective therapies for Group 3 MB metastatic recurrences, this work highlights promising therapeutic options to treat these aggressive tumors. Additional studies are warranted to investigate these inhibitors' mechanisms and recommended in vivo administration.


Assuntos
Neoplasias Encefálicas , Neoplasias Cerebelares , Meduloblastoma , Humanos , Criança , Camundongos , Animais , Adolescente , Meduloblastoma/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Recidiva Local de Neoplasia/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico , Modelos Animais de Doenças , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Neoplasias Cerebelares/patologia , Linhagem Celular Tumoral
2.
Cell Rep ; 40(13): 111420, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36170831

RESUMO

Recurrence of solid tumors renders patients vulnerable to advanced, treatment-refractory disease state with mutational and oncogenic landscape distinctive from initial diagnosis. Improving outcomes for recurrent cancers requires a better understanding of cell populations that expand from the post-therapy, minimal residual disease (MRD) state. We profile barcoded tumor stem cell populations through therapy at tumor initiation, MRD, and recurrence in our therapy-adapted, patient-derived xenograft models of glioblastoma (GBM). Tumors show distinct patterns of recurrence in which clonal populations exhibit either a pre-existing fitness advantage or an equipotency fitness acquired through therapy. Characterization of the MRD state by single-cell and bulk RNA sequencing reveals a tumor-intrinsic immunomodulatory signature with prognostic significance at the transcriptomic level and in proteomic analysis of cerebrospinal fluid (CSF) collected from patients with GBM. Our results provide insight into the innate and therapy-driven dynamics of human GBM and the prognostic value of interrogating the MRD state in solid cancers.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/patologia , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Neoplasia Residual/genética , Células-Tronco Neoplásicas/patologia , Proteômica
3.
Sci Adv ; 7(50): eabi5568, 2021 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-34878832

RESUMO

Medulloblastoma (MB) remains a leading cause of cancer-related mortality among children. The paucity of MB samples collected at relapse has hindered the functional understanding of molecular mechanisms driving therapy failure. New models capable of accurately recapitulating tumor progression in response to conventional therapeutic interventions are urgently needed. In this study, we developed a therapy-adapted PDX MB model that has a distinct advantage of generating human MB recurrence. The comparative gene expression analysis of MB cells collected throughout therapy led to identification of genes specifically up-regulated after therapy, including one previously undescribed in the setting of brain tumors, bactericidal/permeability-increasing fold-containing family B member 4 (BPIFB4). Subsequent functional validation resulted in a markedly diminished in vitro proliferation, self-renewal, and longevity of MB cells, translating into extended survival and reduced tumor burden in vivo. Targeting endothelial nitric oxide synthase, a downstream substrate of BPIFB4, impeded growth of several patient-derived MB lines at low nanomolar concentrations.

4.
STAR Protoc ; 2(2): 100436, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33912845

RESUMO

Recent data suggest that cells respond to infection by upregulating the antiviral cytokine interferon-beta (IFN-ß) in a fraction of infected cells. Approaches are thus needed to study these responses on a single-cell level rather than bulk population. Here, we describe a protocol to analyze the IFN-ß response of individual cells using flow cytometry and immunofluorescence microscopy. We show the heterogeneous IFN-ß response to inactivated Sendai virus and human cytomegalovirus, but this protocol can be adapted to other viruses. For complete details on the use and execution of this protocol, please refer to Hare et al. (2020).


Assuntos
Corantes Fluorescentes/metabolismo , Interferon beta , Imagem Individual de Molécula/métodos , Análise de Célula Única/métodos , Telomerase/metabolismo , Linhagem Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Corantes Fluorescentes/análise , Humanos , Interferon beta/análise , Interferon beta/metabolismo , Telomerase/genética
5.
Methods Mol Biol ; 1869: 69-77, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30324514

RESUMO

As a useful biotechnology, flow cytometry has revolutionized the field of cell analysis through its dynamic system that employs fluidics, optics, and electronics. It was first used to analyze DNA, but is often used to determine biomarker expression, as well as to characterize and sort cells, in accordance with various parameters. A common application of flow cytometry is the identification and isolation of a distinct cancer cell population, known as cancer stem cells (CSCs). Various biomarkers have been used to elucidate this proportion of cells within the brain, termed brain tumor initiating cells (BTICs). Here, we discuss methodology to prepare BTICs for flow cytometric analysis that includes the expression of markers.


Assuntos
Neoplasias Encefálicas/patologia , Citometria de Fluxo/métodos , Células-Tronco Neoplásicas/patologia , Adesão Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Corantes Fluorescentes/metabolismo , Humanos , Coloração e Rotulagem
6.
Methods Mol Biol ; 1869: 85-91, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30324516

RESUMO

Differentiation is a central key capability of stem cells. Their ability to be multipotent and undergo self-renewal are key identifying features of stem cells. A differentiation assay allows for study of one of the essential features of stem cells, the ability to differentiate into all of the cell types of its lineage, in order to ensure that the cells cultured and utilized in key experiments indeed have stem cell properties. Neural stem cells when plated in differentiation media, differentiate into all three neural lineages: Neurons, Astrocytes, and Oligodendrocytes. Brain tumor initiating cells (BTICs) are cells present in brain tumors that possess stem cell properties and are able to self-renew and differentiate into neural lineages. In the current chapter, we discuss protocols involved in immunofluorescence staining and identification of differentiated cells from BTIC populations.


Assuntos
Neoplasias Encefálicas/patologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Neoplásicas/patologia , Permeabilidade da Membrana Celular , Citometria de Fluxo , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Células-Tronco Neurais/metabolismo
7.
Cancer Res ; 78(17): 5124-5134, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29986997

RESUMO

Brain metastases (BM) result from the spread of primary tumors to the brain and are a leading cause of cancer mortality in adults. Secondary tissue colonization remains the main bottleneck in metastatic development, yet this "premetastatic" stage of the metastatic cascade, when primary tumor cells cross the blood-brain barrier and seed the brain before initiating a secondary tumor, remains poorly characterized. Current studies rely on specimens from fully developed macrometastases to identify therapeutic options in cancer treatment, overlooking the potentially more treatable "premetastatic" phase when colonizing cancer cells could be targeted before they initiate the secondary brain tumor. Here we use our established brain metastasis initiating cell (BMIC) models and gene expression analyses to characterize premetastasis in human lung-to-BM. Premetastatic BMIC engaged invasive and epithelial developmental mechanisms while simultaneously impeding proliferation and apoptosis. We identified the dopamine agonist apomorphine to be a potential premetastasis-targeting drug. In vivo treatment with apomorphine prevented BM formation, potentially by targeting premetastasis-associated genes KIF16B, SEPW1, and TESK2 Low expression of these genes was associated with poor survival of patients with lung adenocarcinoma. These results illuminate the cellular and molecular dynamics of premetastasis, which is subclinical and currently impossible to identify or interrogate in human patients with BM. These data present several novel therapeutic targets and associated pathways to prevent BM initiation.Significance: These findings unveil molecular features of the premetastatic stage of lung-to-brain metastases and offer a potential therapeutic strategy to prevent brain metastases. Cancer Res; 78(17); 5124-34. ©2018 AACR.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Terapia de Alvo Molecular , Metástase Neoplásica/tratamento farmacológico , Apomorfina/farmacologia , Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dopamina/metabolismo , Agonistas de Dopamina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Cinesinas/genética , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Proteínas Serina-Treonina Quinases/genética , Selenoproteína W/genética
8.
Cancer Res ; 78(17): 5023-5037, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29945963

RESUMO

Glioblastoma (GBM) carries a dismal prognosis and inevitably relapses despite aggressive therapy. Many members of the Eph receptor tyrosine kinase (EphR) family are expressed by GBM stem cells (GSC), which have been implicated in resistance to GBM therapy. In this study, we identify several EphRs that mark a therapeutically targetable GSC population in treatment-refractory, recurrent GBM (rGBM). Using a highly specific EphR antibody panel and CyTOF (cytometry by time-of-flight), we characterized the expression of all 14 EphR in primary and recurrent patient-derived GSCs to identify putative rGBM-specific EphR. EPHA2 and EPHA3 coexpression marked a highly tumorigenic cell population in rGBM that was enriched in GSC marker expression. Knockdown of EPHA2 and EPHA3 together led to increased expression of differentiation marker GFAP and blocked clonogenic and tumorigenic potential, promoting significantly higher survival in vivo Treatment of rGBM with a bispecific antibody against EPHA2/A3 reduced clonogenicity in vitro and tumorigenic potential of xenografted recurrent GBM in vivo via downregulation of AKT and ERK and increased cellular differentiation. In conclusion, we show that EPHA2 and EPHA3 together mark a GSC population in rGBM and that strategic cotargeting of EPHA2 and EPHA3 presents a novel and rational therapeutic approach for rGBM.Significance: Treatment of rGBM with a novel bispecific antibody against EPHA2 and EPHA3 reduces tumor burden, paving the way for the development of therapeutic approaches against biologically relevant targets in rGBM. Cancer Res; 78(17); 5023-37. ©2018 AACR.


Assuntos
Efrina-A2/genética , Glioblastoma/genética , Recidiva Local de Neoplasia/genética , Receptores Proteína Tirosina Quinases/genética , Animais , Biomarcadores Tumorais/genética , Carcinogênese/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Efrina-A2/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/radioterapia , Humanos , Camundongos , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/radioterapia , Células-Tronco Neoplásicas/patologia , Prognóstico , Radiação , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptor EphA3 , Receptores da Família Eph/antagonistas & inibidores , Receptores da Família Eph/genética , Temozolomida/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Neurooncol ; 126(1): 57-67, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26498281

RESUMO

Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults with average disease relapse at 9 months and median survival rarely extending beyond 15 months. Brain tumor stem cells (BTSCs) have been implicated in not only initiating GBM but also conferring resistance to therapy. However, it is not clear whether the BTSC population that initiates tumor growth is also responsible for GBM recurrence. In this study, we have developed a novel in vitro treatment model to profile the evolution of primary treatment-naïve GBM BTSCs through chemoradiotherapy. We report that our in vitro model enriched for a CD15+/CD133- BTSC population, mirroring the phenotype of BTSCs in recurrent GBM. We also show that in vitro treatment increased stem cell gene expression as well as self-renewal capacity of primary GBMs. In addition, the chemoradiotherapy-refractory gene signature obtained from gene expression profiling identified a hyper-aggressive subtype of glioma. The delivery of in vitro chemoradiotherapy to primary GBM BTSCs models several aspects of recurrent GBM biology, and could be used as a discovery and drug-screening platform to uncover new biological drivers and therapeutic targets in GBM.


Assuntos
Neoplasias Encefálicas/patologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Antígenos CD/metabolismo , Antinematódeos/farmacologia , Antineoplásicos/farmacologia , Autorrenovação Celular/fisiologia , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Masculino , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos da radiação , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA