Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Data ; 8(1): 112, 2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33875665

RESUMO

We created daily concentration estimates for fine particulate matter (PM2.5) at the centroids of each county, ZIP code, and census tract across the western US, from 2008-2018. These estimates are predictions from ensemble machine learning models trained on 24-hour PM2.5 measurements from monitoring station data across 11 states in the western US. Predictor variables were derived from satellite, land cover, chemical transport model (just for the 2008-2016 model), and meteorological data. Ten-fold spatial and random CV R2 were 0.66 and 0.73, respectively, for the 2008-2016 model and 0.58 and 0.72, respectively for the 2008-2018 model. Comparing areal predictions to nearby monitored observations demonstrated overall R2 of 0.70 for the 2008-2016 model and 0.58 for the 2008-2018 model, but we observed higher R2 (>0.80) in many urban areas. These data can be used to understand spatiotemporal patterns of, exposures to, and health impacts of PM2.5 in the western US, where PM2.5 levels have been heavily impacted by wildfire smoke over this time period.


Assuntos
Poluentes Atmosféricos/análise , Poluição do Ar/análise , Exposição Ambiental , Material Particulado/análise , Censos , Humanos , Aprendizado de Máquina , Estados Unidos
2.
Theranostics ; 9(7): 2071-2083, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31037157

RESUMO

Glioblastoma (GBM) remains the most lethal and untreatable central nervous system malignancy. The challenges to devise novel and effective anti-tumor therapies include difficulty in locating the precise tumor border for complete surgical resection, and rapid regrowth of residual tumor tissue after standard treatment. Repeatable and non-invasive intranasal application of neural stem cells (NSCs) was recently shown to enable clinically relevant delivery of therapy to tumors. Treatment with chemotactic NSCs demonstrated significant survival benefits when coupled with radiation and oncolytic virotherapy in preclinical models of GBM. In order to further augment the clinical applicability of this novel therapeutic platform, we postulate that the FDA-approved compound, methimazole (MT), can be safely utilized to delay the nasal clearance and improve the ability of NSCs to penetrate the olfactory epithelium for robust in vivo brain tumor targeting and therapeutic actions. METHODS: To examine the role of reversible reduction of the olfactory epithelial barrier in non-invasive intranasal delivery, we explored the unique pharmacologic effect of MT at a single dosage regimen. In our proof-of-concept studies, quantitative magnetic resonance imaging (MRI), immunocytochemistry, and survival analysis were performed on glioma-bearing mice treated with a single dose of MT prior to intranasal anti-GBM therapy using an oncolytic virus (OV)-loaded NSCs. RESULTS: Based on histology and in vivo imaging, we found that disrupting the olfactory epithelium with MT effectively delays clearance and allows NSCs to persist in the nasal cavity for at least 24 h. MT pretreatment amplified the migration of NSCs to the tumor. The therapeutic advantage of this enhancement was quantitatively validated by tissue analysis and MRI tracking of NSCs loaded with superparamagnetic iron oxide nanoparticles (SPIOs) in live animals. Moreover, we observed significant survival benefits in GBM-bearing mice treated with intranasal delivery of oncolytic virus-loaded NSCs following MT injection. CONCLUSION: Our work identified a novel pharmacologic strategy to accelerate the clinical application of the non-invasive NSCs-based therapeutic platform to tackle aggressive brain tumors.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Mucosa Nasal/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Adenoviridae/genética , Animais , Neoplasias Encefálicas/virologia , Linhagem Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glioblastoma/virologia , Glioma/tratamento farmacológico , Glioma/virologia , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Camundongos , Camundongos Nus , Mucosa Nasal/virologia , Células-Tronco Neurais/virologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
3.
BMC Cancer ; 18(1): 238, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29490608

RESUMO

BACKGROUND: Targeted therapy with trastuzumab has become a mainstay for HER2-positive breast cancer without a clear understanding of the mechanism of its action. While many mechanisms have been suggested for the action of trastuzumab, most of them are not substantiated by experimental data. It has been suggested that trastuzumab functions by inhibiting intracellular signaling initiated by HER2, however, the data are very controversial. A major issue is the different cellular background of various breast cancer cells lines used in these studies. Each breast cancer cell line has a unique expression profile of various HER receptors, which could significantly affect the effects of trastuzumab. METHODS: To overcome this problem, in this research we adopted a cell model that allow us to specifically examine the effects of trastuzumab on a single HER receptor without the influence of other HER receptors. Three CHO cell lines stably expressing only human EGFR (CHO-EGFR), HER2 (CHO-K6), or HER3 (CHO-HER3) were used. Various methods including cytotoxicity assay, immunoblotting, indirect immunofluorescence, cross linking, and antibody-dependent cellular cytotoxicity (ADCC) were employed in this research. RESULTS: We showed that trastuzumab did not bind EGFR and HER3, and thus did not affect the homodimerization and phosphorylation of EGFR and HER3. However, overexpression of HER2 in CHO cells, in the absence of other HER receptors, resulted in the homodimerization of HER2 and the phosphorylation of HER2 at all major pY residues. Trastuzumab bound to HER2 specifically and with high affinity. Trastuzumab inhibited neither the homodimerization of HER2, nor the phosphorylation of HER2 at most phosphotyrosine residues. Moreover, trastuzumab did not inhibit the phosphorylation of ERK and AKT in CHO-K6 cells, and did not inhibit the proliferation of CHO-K6 cells. However, trastuzumab induced strong ADCC in CHO-K6 cells. CONCLUSION: We concluded that, in the absence of other HER receptors, trastuzumab exerts its antitumor activity through the induction of ADCC, rather than the inhibition of HER2-homodimerization and phosphorylation.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Receptor ErbB-2/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Trastuzumab/farmacologia , Animais , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Células CHO , Cricetulus , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/metabolismo , Humanos , Fosforilação , Multimerização Proteica , Receptor ErbB-2/metabolismo , Receptor ErbB-3/efeitos dos fármacos , Receptor ErbB-3/metabolismo , Trastuzumab/uso terapêutico
4.
Mol Ther ; 26(4): 986-995, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29503195

RESUMO

In order to fully harness the potential of immunotherapy with chimeric antigen receptor (CAR)-modified T cells, pre-clinical studies must be conducted in immunocompetent animal models that closely mimic the immunosuppressive malignant glioma (MG) microenvironment. Thus, the goal of this project was to study the in vivo fate of T cells expressing CARs specific for the MG antigen IL13Rα2 (IL13Rα2-CARs) in immunocompetent MG models. Murine T cells expressing IL13Rα2-CARs with a CD28.ζ (IL13Rα2-CAR.CD28.ζ) or truncated signaling domain (IL13Rα2-CAR.Δ) were generated by retroviral transduction, and their effector function was evaluated both in vitro and in vivo. IL13Rα2-CAR.CD28.ζ T cells' specificity toward IL13Rα2 was confirmed through cytokine production and cytolytic activity. In vivo, a single intratumoral injection of IL13Rα2-CAR.CD28.ζ T cells significantly extended the survival of IL13Rα2-expressing GL261 and SMA560 glioma-bearing mice; long-term survivors were resistant to re-challenge with IL13Rα2-negative and IL13Rα2-positive tumors. IL13Rα2-CAR.CD28.ζ T cells proliferated, produced cytokines (IFNγ, TNF-α), and promoted a phenotypically pro-inflammatory glioma microenvironment by inducing a significant increase in the number of CD4+ and CD8+ T cells and CD8α+ dendritic cells and a decrease in Ly6G+ myeloid-derived suppressor cells (MDSCs). Our data underline the significance of CAR T cell studies in immunocompetent hosts and further validate IL13Rα2-CAR T cells as an efficacious therapeutic strategy for MG.


Assuntos
Glioblastoma/imunologia , Glioblastoma/metabolismo , Imunoterapia Adotiva , Subunidade alfa2 de Receptor de Interleucina-13/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Antígenos CD28/imunologia , Antígenos CD28/metabolismo , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Modelos Animais de Doenças , Feminino , Expressão Gênica , Vetores Genéticos/genética , Glioblastoma/genética , Glioblastoma/terapia , Humanos , Imunoterapia Adotiva/métodos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Subunidade alfa2 de Receptor de Interleucina-13/antagonistas & inibidores , Masculino , Camundongos , Receptores de Antígenos Quiméricos/genética , Resultado do Tratamento , Microambiente Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Expert Opin Drug Deliv ; 15(2): 163-172, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28895435

RESUMO

INTRODUCTION: Glioblastoma (GBM) is the most aggressive malignant brain cancer in adults, and its poor prognosis and resistance to the existing standard of care require the development of innovative therapeutic modalities. The local delivery of stem cells as therapeutic carriers against glioma has produced encouraging results, but encounters obstacles with regards to the repeatability and invasiveness of administration. Intranasal delivery of therapeutic stem cells could overcome these obstacles, among others, as a noninvasive and easily repeatable mode of administration. AREAS COVERED: This review describes nasal anatomy, routes of stem cell migration, and factors affecting stem cell delivery to hard-to-reach tumors. Furthermore, this review discusses the molecular mechanisms underlying stem cell migration following delivery, as well as possible stem cell effector functions to be considered in combination with intranasal delivery. EXPERT OPINION: Further research is necessary to elucidate the dynamics of stem cell effector functions in the context of intranasal delivery and optimize their therapeutic potency. Nonetheless, the technique represents a promising tool against brain cancer and has the potential to be expanded for use against other brain pathologies.


Assuntos
Neoplasias Encefálicas/terapia , Sistemas de Liberação de Medicamentos , Glioblastoma/terapia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Administração Intranasal , Adulto , Movimento Celular , Humanos
6.
J Vis Exp ; (124)2017 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-28605391

RESUMO

The intrinsic tropism towards brain malignancies renders stem cells as promising carriers of therapeutic agents against malignant tumors. The delivery of therapeutic stem cells via the intranasal route is a recently discovered alternative strategy, with strong potential for clinical translation, due to its non-invasive nature compared to intracranial implantation or delivery via systemic routes. The lack of blood brain barrier further strengthens the therapeutic potential of stem cells undergoing intranasal brain entry. This article summarizes the essential techniques utilized in our studies and outlines the basic principles of intranasal strategy for stem cell delivery using a mouse model of intracranial glioma xenografts. We demonstrate the optimized procedures that generate consistent and reproducible results with specific predetermined experimental parameters and offer guidelines for streamlined work flow that ensure efficient execution and reliable experimental outcome. The article is designed to serve as a baseline for further experimental customization based on hypothesis, stem cell types, or tumor specifics.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Administração Intranasal , Animais , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Receptores CXCR4/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Stem Cell Reports ; 7(3): 471-482, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27594591

RESUMO

The challenges to effective drug delivery to brain tumors are twofold: (1) there is a lack of non-invasive methods of local delivery and (2) the blood-brain barrier limits systemic delivery. Intranasal delivery of therapeutics to the brain overcomes both challenges. In mouse model of malignant glioma, we observed that a small fraction of intranasally delivered neural stem cells (NSCs) can migrate to the brain tumor site. Here, we demonstrate that hypoxic preconditioning or overexpression of CXCR4 significantly enhances the tumor-targeting ability of NSCs, but without altering their phenotype only in genetically modified NSCs. Modified NSCs deliver oncolytic virus to glioma more efficiently and extend survival of experimental animals in the context of radiotherapy. Our findings indicate that intranasal delivery of stem cell-based therapeutics could be optimized for future clinical applications, and allow for safe and repeated administration of biological therapies to brain tumors and other CNS disorders.


Assuntos
Glioma/genética , Glioma/patologia , Células-Tronco Neurais/metabolismo , Terapia Viral Oncolítica , Receptores CXCR4/genética , Administração Intranasal , Animais , Biomarcadores , Linhagem Celular Tumoral , Movimento Celular , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glioma/mortalidade , Glioma/terapia , Humanos , Hipóxia/metabolismo , Camundongos , Vírus Oncolíticos/genética , Fenótipo , Receptores CXCR4/metabolismo , Transplante de Células-Tronco , Transdução Genética , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...