Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-39179298

RESUMO

BACKGROUND AND PURPOSE: To evaluate the radiomics-based model performance for differentiation between glioblastoma (GB) and brain metastases (BM) using magnetization prepared rapid gradient echo (MPRAGE) and volumetric interpolated breath-hold examination (VIBE) T1-contrast enhanced sequences. MATERIALS AND METHODS: T1-CE MPRAGE and VIBE sequences acquired in 108 patients (31 GBs and 77 BM) during the same MRI session were retrospectively evaluated. Post standardized image pre-processing and segmentation, radiomics features were extracted from necrotic and enhancing tumor components. Pearson correlation analysis of radiomics features from tumor subcomponents was also performed. A total of 90 machine learning (ML) pipelines were evaluated using a five-fold cross validation. Performance was measured by mean AUC-ROC, Log-loss and Brier scores. RESULTS: A feature-wise comparison showed that the radiomic features between sequences were strongly correlated, with the highest correlation for shape-based features. The mean AUC across the top-ten pipelines ranged between 0.851-0.890 with T1-CE MPRAGE and between 0.869-0.907 with T1-CE VIBE sequence. Top performing models for the MPRAGE sequence commonly used support vector machines, while those for VIBE sequence used either support vector machines or random forest. Common feature reduction methods for top-performing models included linear combination filter and least absolute shrinkage and selection operator (LASSO) for both sequences. For the same ML-feature reduction pipeline, model performances were comparable (AUC-ROC difference range: [-0.078, 0.046]). CONCLUSIONS: Radiomic features derived from T1-CE MPRAGE and VIBE sequences are strongly correlated and may have similar overall classification performance for differentiating GB from BM. ABBREVIATIONS: BM: Brain metastases, GB: glioblastoma, T1-CE: T1 contrast enhanced sequence, MPRAGE: magnetization prepared rapid gradient echo, ML: machine learning, RF: random forest, VIBE: volumetric interpolated breath-hold examination.

2.
Int J Nanomedicine ; 18: 2525-2537, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37197026

RESUMO

Purpose: Hepatocellular carcinoma (HCC) has limited treatment options, and modest survival after systemic chemotherapy or procedures such as transarterial chemoembolization (TACE). There is therefore a need to develop targeted therapies to address HCC. Gene therapies hold immense promise in treating a variety of diseases, including HCC, though delivery remains a critical hurdle. This study investigated a new approach of local delivery of polymeric nanoparticles (NPs) via intra-arterial injection for targeted local gene delivery to HCC tumors in an orthotopic rat liver tumor model. Methods: Poly(beta-amino ester) (PBAE) nanoparticles were formulated and assessed for GFP transfection in N1-S1 rat HCC cells in vitro. Optimized PBAE NPs were next administered to rats via intra-arterial injection with and without orthotopic HCC tumors, and both biodistribution and transfection were assessed. Results: In vitro transfection of PBAE NPs led to >50% transfected cells in adherent and suspension culture at a variety of doses and weight ratios. Administration of NPs via intra-arterial or intravenous injection demonstrated no transfection of healthy liver, while intra-arterial NP injection led to transfection of tumors in an orthotopic rat HCC model. Conclusion: Hepatic artery injection is a promising delivery approach for PBAE NPs and demonstrates increased targeted transfection of HCC tumors compared to intravenous administration, and offers a potential alternative to standard chemotherapies and TACE. This work demonstrates proof of concept for administration of polymeric PBAE nanoparticles via intra-arterial injection for gene delivery in rats.


Assuntos
Carcinoma Hepatocelular , Quimioembolização Terapêutica , Neoplasias Hepáticas , Ratos , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Injeções Intra-Arteriais , Distribuição Tecidual , Quimioembolização Terapêutica/métodos , Polímeros
3.
Sci Adv ; 8(29): eabo6406, 2022 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-35857843

RESUMO

Hepatocellular carcinoma (HCC) develops predominantly in the inflammatory environment of a cirrhotic liver caused by hepatitis, toxin exposure, or chronic liver disease. A targeted therapeutic approach is required to enable cancer killing without causing toxicity and liver failure. Poly(beta-amino-ester) (PBAE) nanoparticles (NPs) were used to deliver a completely CpG-free plasmid harboring mutant herpes simplex virus type 1 sr39 thymidine kinase (sr39) DNA to human HCC cells. Transfection with sr39 enables cancer cell killing with the prodrug ganciclovir and accumulation of 9-(4-18F-fluoro-3-hydroxymethylbutyl)guanine (18F-FHBG) for in vivo imaging. Targeting was achieved using a CpG-free human alpha fetoprotein (AFP) promoter (CpGf-AFP-sr39). Expression was restricted to AFP-producing HCC cells, enabling selective transfection of orthotopic HCC xenografts. CpGf-AFP-sr39 NP treatment resulted in 62% reduced tumor size, and therapeutic gene expression was detectable by positron emission tomography (PET). This systemic nanomedicine achieved tumor-specific delivery, therapy, and imaging, representing a promising platform for targeted treatment of HCC.


Assuntos
Carcinoma Hepatocelular , Herpesvirus Humano 1 , Neoplasias Hepáticas , Nanopartículas , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Polímeros , Medicina de Precisão , alfa-Fetoproteínas/genética
4.
Mol Ther Oncolytics ; 21: 377-388, 2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34189258

RESUMO

Despite initial promise, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based approaches to cancer treatment have yet to yield a clinically approved therapy, due to delivery challenges, a lack of potency, and drug resistance. To address these challenges, we have developed poly(beta-amino ester) (PBAE) nanoparticles (NPs), as well as an engineered cDNA sequence encoding a secretable TRAIL (sTRAIL) protein, to enable reprogramming of liver cancer cells to locally secrete TRAIL protein. We show that sTRAIL initiates apoptosis in transfected cells and has a bystander effect to non-transfected cells. To address TRAIL resistance, NP treatment is combined with histone deacetylase inhibitors, resulting in >80% TRAIL-mediated cell death in target cancer cells and significantly slowed xenograft tumor growth. This anti-cancer effect is specific to liver cancer cells, with up to 40-fold higher cell death in HepG2 cancer cells over human hepatocytes. By combining cancer-specific TRAIL NPs with small-molecule-sensitizing drugs, this strategy addresses multiple challenges associated with TRAIL therapy and offers a new potential approach for cancer treatment.

5.
J Control Release ; 263: 18-28, 2017 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-28351668

RESUMO

Hepatocellular carcinoma (HCC) is the third most deadly cancer in the US, with a meager 5-year survival rate of <20%. Such unfavorable numbers are closely related to the heterogeneity of the disease and the unsatisfactory therapies currently used to manage patients with invasive HCC. Outside of the clinic, gene therapy research is evolving to overcome the poor responses and toxicity associated with standard treatments. The inadequacy of gene delivery vectors, including poor intracellular delivery and cell specificity, are major barriers in the gene therapy field. Herein, we described a non-viral strategy for effective and cancer-specific DNA delivery to human HCC using biodegradable poly(beta-amino ester) (PBAE) nanoparticles (NPs). Varied PBAE NP formulations were evaluated for transfection efficacy and cytotoxicity to a range of human HCC cells as well as healthy human hepatocytes. To address HCC heterogeneity, nine different sources of human HCC cells were utilized. The polymeric NPs composed of 2-((3-aminopropyl)amino) ethanol end-modified poly(1,5-pentanediol diacrylate-co-3-amino-1-propanol) ('536') at a 25 polymer-to-DNA weight-to-weight ratio led to high transfection efficacy to all of the liver cancer lines, but not to hepatocytes. Each individual HCC line had a significantly higher percentage of exogenous gene expression than the healthy liver cells (P<0.01). Notably, this biodegradable end-modified PBAE gene delivery vector was not cytotoxic and maintained the viability of hepatocytes above 80%. In a HCC/hepatocyte co-culture model, in which cancerous and healthy cells share the same micro-environment, 536 25 w/w NPs specifically transfected cancer cells. PBAE NP administration to a subcutaneous HCC mouse model, established with one of the human lines tested in vitro, confirmed effective DNA transfection in vivo. PBAE-based NPs enabled high and preferential DNA delivery to HCC cells, sparing healthy hepatocytes. These biodegradable and liver cancer-selective NPs are a promising technology to deliver therapeutic genes to liver cancer.


Assuntos
Carcinoma Hepatocelular/metabolismo , DNA/administração & dosagem , Portadores de Fármacos/administração & dosagem , Neoplasias Hepáticas/metabolismo , Nanopartículas/administração & dosagem , Polímeros/administração & dosagem , Animais , Carcinoma Hepatocelular/genética , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Feminino , Proteínas de Fluorescência Verde/genética , Hepatócitos/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/genética , Luciferases/genética , Camundongos Nus , Plasmídeos
6.
J Drug Target ; 23(7-8): 627-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26061296

RESUMO

In this article, advances in designing polymeric nanoparticles for targeted cancer gene therapy are reviewed. Characterization and evaluation of biomaterials, targeting ligands, and transcriptional elements are each discussed. Advances in biomaterials have driven improvements to nanoparticle stability and tissue targeting, conjugation of ligands to the surface of polymeric nanoparticles enable binding to specific cancer cells, and the design of transcriptional elements has enabled selective DNA expression specific to the cancer cells. Together, these features have improved the performance of polymeric nanoparticles as targeted non-viral gene delivery vectors to treat cancer. As polymeric nanoparticles can be designed to be biodegradable, non-toxic, and to have reduced immunogenicity and tumorigenicity compared to viral platforms, they have significant potential for clinical use. Results of polymeric gene therapy in clinical trials and future directions for the engineering of nanoparticle systems for targeted cancer gene therapy are also presented.


Assuntos
Terapia Genética/métodos , Nanopartículas , Neoplasias/terapia , Animais , Materiais Biocompatíveis/administração & dosagem , Regulação Neoplásica da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Humanos , Neoplasias/genética , Polímeros/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA