Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Biostatistics ; 23(3): 844-859, 2022 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-33616157

RESUMO

Validation of phenotyping models using Electronic Health Records (EHRs) data conventionally requires gold-standard case and control labels. The labeling process requires clinical experts to retrospectively review patients' medical charts, therefore is labor intensive and time consuming. For some disease conditions, it is prohibitive to identify the gold-standard controls because routine clinical assessments are performed for selective patients who are deemed to possibly have the condition. To build a model for phenotyping patients in EHRs, the most readily accessible data are often for a cohort consisting of a set of gold-standard cases and a large number of unlabeled patients. Hereby, we propose methods for assessing model calibration and discrimination using such "positive-only" EHR data that does not require gold-standard controls, provided that the labeled cases are representative of all cases. For model calibration, we propose a novel statistic that aggregates differences between model-free and model-based estimated numbers of cases across risk subgroups, which asymptotically follows a Chi-squared distribution. We additionally demonstrate that the calibration slope can also be estimated using such "positive-only" data. We propose consistent estimators for discrimination measures and derive their large sample properties. We demonstrate performances of the proposed methods through extensive simulation studies and apply them to Penn Medicine EHRs to validate two preliminary models for predicting the risk of primary aldosteronism.


Assuntos
Algoritmos , Registros Eletrônicos de Saúde , Calibragem , Humanos , Fenótipo , Estudos Retrospectivos
2.
Clin Trials ; 17(4): 402-404, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32522027

RESUMO

For utilizing electronic health records to help design and conduct clinical trials, an essential first step is to select eligible patients from electronic health records, that is, electronic health record phenotyping. We present two novel statistical methods that can be used in the context of electronic health record phenotyping. One mitigates the requirement for gold-standard control patients in developing phenotyping algorithms, and the other effectively corrects for bias in downstream analysis introduced by study samples contaminated by ineligible subjects.


Assuntos
Variação Biológica da População , Ensaios Clínicos como Assunto/métodos , Registros Eletrônicos de Saúde , Algoritmos , Humanos , Modelos Logísticos , Fenótipo , Projetos de Pesquisa
3.
Stroke ; 48(1): 187-194, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27879448

RESUMO

BACKGROUND AND PURPOSE: We sought to compare the effect of chronic treatment with commonly tolerated doses of Fasudil, a specific RhoA kinase (ROCK) inhibitor, and simvastatin (with pleiotropic effects including ROCK inhibition) on cerebral cavernous malformation (CCM) genesis and maturation in 2 models that recapitulate the human disease. METHODS: Two heterozygous murine models, Ccm1+/-Msh2-/- and Ccm2+/-Trp53-/-, were treated from weaning to 4 to 5 months of age with Fasudil (100 mg/kg per day), simvastatin (40 mg/kg per day) or with placebo. Mouse brains were blindly assessed for CCM lesion burden, nonheme iron deposition (as a quantitative measure of chronic lesional hemorrhage), and ROCK activity. RESULTS: Fasudil, but not simvastatin, significantly decreased mature CCM lesion burden in Ccm1+/-Msh2-/- mice, and in meta-analysis of both models combined, when compared with mice receiving placebo. Fasudil and simvastatin both significantly decreased the integrated iron density per mature lesion area in Ccm1+/-Msh2-/- mice, and in both models combined, compared with mice given placebo. ROCK activity in mature lesions of Ccm1+/-Msh2-/- mice was similar with both treatments. Fasudil, but not simvastatin, improved survival in Ccm1+/-Msh2-/- mice. Fasudil and simvastatin treatment did not affect survival or lesion development significantly in Ccm2+/-Trp53-/- mice alone, and Fasudil benefit seemed limited to males. CONCLUSIONS: ROCK inhibitor Fasudil was more efficacious than simvastatin in improving survival and blunting the development of mature CCM lesions. Both drugs significantly decreased chronic hemorrhage in CCM lesions. These findings justify the development of ROCK inhibitors and the clinical testing of commonly used statin agents in CCM.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Neoplasias Encefálicas/tratamento farmacológico , Modelos Animais de Doenças , Hemangioma Cavernoso do Sistema Nervoso Central/tratamento farmacológico , Sinvastatina/uso terapêutico , Quinases Associadas a rho/antagonistas & inibidores , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/uso terapêutico , Animais , Neoplasias Encefálicas/patologia , Feminino , Hemangioma Cavernoso do Sistema Nervoso Central/patologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Masculino , Camundongos , Camundongos Transgênicos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Sinvastatina/farmacologia
4.
Cerebrovasc Dis ; 43(5-6): 223-230, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28245439

RESUMO

BACKGROUND: Spontaneous intraventricular hemorrhage (IVH) is associated with high rates of morbidity and mortality despite critical care and other advances. An important step in clinical management is to confirm/rule out an underlying vascular lesion, which influences further treatment, potential for further bleeding, and prognosis. Our aim is to compare demographic and clinical characteristics between IVH patients with and without an underlying vascular lesion, and among cohorts with different vascular lesions. METHODS: We analyzed prospectively collected data of IVH patients screened for eligibility as part of the Clot Lysis: Evaluation Accelerated Resolution of IVH Phase III (CLEAR III) clinical trial. The trial adopted a structured screening process to systematically exclude patients with an underlying vascular lesion as the etiology of IVH. We collected age, sex, ethnicity, and primary diagnosis on these cases and vascular lesions were categorized prospectively as aneurysm, vascular malformation (arteriovenous malformation, dural arteriovenous fistula, and cavernoma), Moyamoya disease, or other vascular lesion. We excluded cases <18 or >80 years of age. Baseline characteristics were compared between the CLEAR group (IVH screened without vascular lesion) and the group of IVH patients screened and excluded from CLEAR because of an identified vascular lesion. We further analyzed the differential demographic and clinical characteristics among subcohorts with different vascular lesions. RESULTS: A total of 10,538 consecutive IVH cases were prospectively screened for the trial between 2011 and 2015. Out of these, 496 cases (4.7%) screened negative for underlying vascular lesion, met the inclusion criteria, and were enrolled in the trial (no vascular etiology group); and 1,205 cases (11.4%) were concurrently screened and excluded from the trial because of a demonstrated underlying vascular lesion (vascular etiology group). Cases with vascular lesion were less likely to be >45 years of age (OR 0.28, 95% CI 0.20-0.40), African-American (OR 0.23, 95% CI 0.18-0.31), or male gender (OR 0.48, 95% CI 0.38-0.60), and more likely to present with primary IVH (OR 1.85, 95% CI 1.37-2.51) compared to those with no vascular etiology (p < 0.001). Other demographic factors were associated with specific vascular lesion etiologies. A combination of demographic features increases the association with the absence of vascular lesion, but not with absolute reliability (OR 0.1, 95% CI 0.06-0.17, p < 0.001). CONCLUSION: An underlying vascular lesion as etiology of IVH cannot be excluded solely by demographic parameters in any patient. Some form of vascular imaging is necessary in screening patients before contemplating interventions like intraventricular fibrinolysis, where safety may be impacted by the presence of vascular lesion.


Assuntos
Hemorragia Cerebral Intraventricular/etiologia , Doenças Vasculares/complicações , Angiografia Cerebral , Hemorragia Cerebral Intraventricular/diagnóstico por imagem , Distribuição de Qui-Quadrado , Ensaios Clínicos Fase III como Assunto , Feminino , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Razão de Chances , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Doenças Vasculares/diagnóstico por imagem
5.
J Immunol ; 195(1): 367-76, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26026061

RESUMO

Dendritic cells (DCs) are professional APCs that are traditionally divided into two distinct subsets, myeloid DC (mDCs) and plasmacytoid DC (pDCs). pDCs are known for their ability to secrete large amounts of IFN-α. Apart from IFN-α production, pDCs can also process Ag and induce T cell immunity or tolerance. In several solid tumors, pDCs have been shown to play a critical role in promoting tumor immunosuppression. We investigated the role of pDCs in the process of glioma progression in the syngeneic murine model of glioma. We show that glioma-infiltrating pDCs are the major APC in glioma and are deficient in IFN-α secretion (p < 0.05). pDC depletion leads to increased survival of the mice bearing intracranial tumor by decreasing the number of regulatory T cells (Tregs) and by decreasing the suppressive capabilities of Tregs. We subsequently compared the ability of mDCs and pDCs to generate effective antiglioma immunity in a GL261-OVA mouse model of glioma. Our data suggest that mature pDCs and mDCs isolated from naive mice can be effectively activated and loaded with SIINFEKL Ag in vitro. Upon intradermal injection in the hindleg, a fraction of both types of DCs migrate to the brain and lymph nodes. Compared to mice vaccinated with pDC or control mice, mice vaccinated with mDCs generate a robust Th1 type immune response, characterized by high frequency of CD4(+)T-bet(+) T cells and CD8(+)SIINFEKEL(+) T cells. This robust antitumor T cell response results in tumor eradication and long-term survival in 60% of the animals (p < 0.001).


Assuntos
Neoplasias Encefálicas/terapia , Vacinas Anticâncer/administração & dosagem , Linhagem da Célula/imunologia , Células Dendríticas/imunologia , Glioma/terapia , Imunidade Adaptativa , Animais , Encéfalo/imunologia , Encéfalo/patologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Vacinas Anticâncer/imunologia , Contagem de Células , Movimento Celular , Células Dendríticas/classificação , Células Dendríticas/patologia , Células Dendríticas/transplante , Glioma/imunologia , Glioma/mortalidade , Glioma/patologia , Epitopos Imunodominantes/química , Epitopos Imunodominantes/imunologia , Interferon-alfa/biossíntese , Linfonodos/imunologia , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/imunologia , Células Mieloides/patologia , Ovalbumina/química , Ovalbumina/imunologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Análise de Sobrevida , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Células Th1/imunologia , Células Th1/patologia , Vacinação
6.
Cancer Immunol Immunother ; 65(12): 1555-1567, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27734112

RESUMO

Regulatory T cells (Tregs) are potently immunosuppressive cells that accumulate within the glioma microenvironment. The reduction in their function and/or trafficking has been previously shown to enhance survival in preclinical models of glioma. Glucocorticoid-induced TNFR-related protein (GITR) is a tumor necrosis factor superfamily receptor enriched on Tregs that has shown promise as a target for immunotherapy. An agonistic antibody against GITR has been demonstrated to inhibit Tregs in a number of models and has only been recently addressed in glioma. In this study, we examined the modality of the antibody function at the tumor site as opposed to the periphery as the blood-brain barrier prevents efficient antibody delivery to brain tumors. Mice harboring established GL261 tumors were treated with anti-GITR monotherapy and were shown to have a significant increase in overall survival (p < 0.01) when antibodies were injected directly into the glioma core, whereas peripheral antibody treatment only had a modest effect. Peripheral treatment resulted in a significant decrease in granzyme B (GrB) expression by Tregs, whereas intratumoral treatment resulted in both a decrease in GrB expression by Tregs and their selective depletion, which was largely mediated by FcγR-mediated destruction. We also discovered that anti-GITR treatment results in the enhanced survival and functionality of dendritic cells (DCs)-a previously unreported effect of this immunotherapy. In effect, this study demonstrates that the targeting of GITR is a feasible and noteworthy treatment option for glioma, but is largely dependent on the anatomical location in which the antibodies are delivered.


Assuntos
Glioma/tratamento farmacológico , Imunoterapia/métodos , Linfócitos T Reguladores/imunologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Camundongos
7.
J Transl Med ; 14(1): 134, 2016 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-27184224

RESUMO

BACKGROUND: CRAd-S-pk7 is a conditionally replicative oncolytic adenoviral vector that contains a survivin promoter and a pk7 fiber modification that confer tumor-specific transcriptional targeting and preferential replication in glioma while sparing the surrounding normal brain parenchyma. METHODS: This IND-enabling study performed under GLP conditions evaluated the toxicity and biodistribution of CRAd-S-pk7 administered as a single intracerebral dose to Syrian hamsters, a permissive model of adenoviral replication. Two hundred and forty animals were stereotactically administered either vehicle (n = 60) or CRAd-S-pk7 at 2.5 × 10(7), 2.5 × 10(8), or 2.5 × 10(9) viral particles (vp)/animal (each n = 60) on day 1. The animals were closely monitored for toxicology evaluation, assessment of viral distribution, and immunogenicity of CRAd-S-pk7. RESULTS: Changes in hematology, clinical chemistry, and coagulation parameters were minor and transient, and consistent with the inflammatory changes observed microscopically. These changes were considered to be of little toxicological significance. The vector remained localized primarily in the brain and to some degree in the tissues at the incision site. Low levels of vector DNA were detected in other tissues in a few animals suggesting systemic circulation of the virus. Viral DNA was detected in brains of hamsters for up to 62 days. However, microscopic changes and virus-related toxicity to the central nervous system were considered minor and decreased in incidence and severity over time. Such changes are not uncommon in studies using adenoviral vectors. CONCLUSION: This study provides safety and toxicology data justifying a clinical trial of CRAd-S-pk7 loaded in FDA-approved HB1.F3.CD neural stem cell carriers administered at the tumor resection bed in humans with recurrent malignant glioma.


Assuntos
Adenoviridae/genética , Vetores Genéticos/administração & dosagem , Replicação Viral , Animais , Formação de Anticorpos/imunologia , Peso Corporal , Encéfalo/patologia , Encéfalo/virologia , Cricetinae , DNA Viral/análise , Modelos Animais de Doenças , Comportamento Alimentar , Feminino , Vetores Genéticos/metabolismo , Genoma , Imunocompetência , Imunoglobulina G/imunologia , Inflamação/patologia , Injeções Intraventriculares , Masculino , Mesocricetus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Distribuição Tecidual
8.
Stem Cells ; 33(10): 2985-94, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26260958

RESUMO

The treatment of human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer has been revolutionized by trastuzumab. However, longer survival of these patients now predisposes them to forming HER2 positive brain metastases, as the therapeutic antibodies cannot cross the blood brain barrier. The current oncologic repertoire does not offer a rational, nontoxic targeted therapy for brain metastases. In this study, we used an established human neural stem cell line, HB1.F3 NSCs and generated a stable pool of cells secreting a high amount of functional full-length anti-HER2 antibody, equivalent to trastuzumab. Anti-HER2Ab secreted by the NSCs (HER2Ab-NSCs) specifically binds to HER2 overexpressing human breast cancer cells and inhibits PI3K-Akt signaling. This translates to HER2Ab-NSC inhibition of breast cancer cell growth in vitro. Preclinical in vivo experiments using HER2Ab overexpressing NSCs in a breast cancer brain metastases (BCBM) mouse model demonstrate that intracranial injection of HER2Ab-NSCs significantly improves survival. In effect, these NSCs provide tumor localized production of HER2Ab, minimizing any potential off-target side effects. Our results establish HER2Ab-NSCs as a novel, nontoxic, and rational therapeutic approach for the successful treatment of HER2 overexpressing BCBM, which now warrants further preclinical and clinical investigation.


Assuntos
Anticorpos Anti-Idiotípicos/biossíntese , Neoplasias Encefálicas/terapia , Neoplasias da Mama/tratamento farmacológico , Células-Tronco Neurais/metabolismo , Receptor ErbB-2/biossíntese , Animais , Anticorpos Anti-Idiotípicos/imunologia , Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Células-Tronco Neurais/imunologia , Células-Tronco Neurais/transplante , Receptor ErbB-2/imunologia , Trastuzumab/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Mol Pharm ; 13(6): 1843-54, 2016 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-27169484

RESUMO

As therapies continue to increase the lifespan of patients with breast cancer, the incidence of brain metastases has steadily increased, affecting a significant number of patients with metastatic disease. However, a major barrier toward treating these lesions is the inability of therapeutics to penetrate into the central nervous system and accumulate within intracranial tumor sites. In this study, we designed a cell-penetrating gold nanoparticle platform to increase drug delivery to brain metastatic breast cancer cells. TAT peptide-modified gold nanoparticles carrying doxorubicin led to improved cytotoxicity toward two brain metastatic breast cancer cell lines with a decrease in the IC50 of at least 80% compared to free drug. Intravenous administration of these particles led to extensive accumulation of particles throughout diffuse intracranial metastatic microsatellites with cleaved caspase-3 activity corresponding to tumor foci. Furthermore, intratumoral administration of these particles improved survival in an intracranial MDA-MB-231-Br xenograft mouse model. Our results demonstrate the promising application of gold nanoparticles for improving drug delivery in the context of brain metastatic breast cancer.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Peptídeos Penetradores de Células/química , Doxorrubicina/administração & dosagem , Ouro/química , Nanopartículas Metálicas/química , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/administração & dosagem , Doxorrubicina/química , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Nanopartículas Metálicas/administração & dosagem , Camundongos , Camundongos Nus
10.
Genet Med ; 17(3): 188-196, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25122144

RESUMO

PURPOSE: The phenotypic manifestations of cerebral cavernous malformation disease caused by rare PDCD10 mutations have not been systematically examined, and a mechanistic link to Rho kinase-mediated hyperpermeability, a potential therapeutic target, has not been established. METHODS: We analyzed PDCD10 small interfering RNA-treated endothelial cells for stress fibers, Rho kinase activity, and permeability. Rho kinase activity was assessed in cerebral cavernous malformation lesions. Brain permeability and cerebral cavernous malformation lesion burden were quantified, and clinical manifestations were assessed in prospectively enrolled subjects with PDCD10 mutations. RESULTS: We determined that PDCD10 protein suppresses endothelial stress fibers, Rho kinase activity, and permeability in vitro. Pdcd10 heterozygous mice have greater lesion burden than other Ccm genotypes. We demonstrated robust Rho kinase activity in murine and human cerebral cavernous malformation vasculature and increased brain vascular permeability in humans with PDCD10 mutation. Clinical phenotype is exceptionally aggressive compared with the more common KRIT1 and CCM2 familial and sporadic cerebral cavernous malformation, with greater lesion burden and more frequent hemorrhages earlier in life. We first report other phenotypic features, including scoliosis, cognitive disability, and skin lesions, unrelated to lesion burden or bleeding. CONCLUSION: These findings define a unique cerebral cavernous malformation disease with exceptional aggressiveness, and they inform preclinical therapeutic testing, clinical counseling, and the design of trials.Genet Med 17 3, 188-196.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Neoplasias do Sistema Nervoso Central/patologia , Hemangioma Cavernoso do Sistema Nervoso Central/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Mutação , Proteínas Proto-Oncogênicas/genética , Quinases Associadas a rho/metabolismo , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Adolescente , Adulto , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Transporte/genética , Células Cultivadas , Neoplasias do Sistema Nervoso Central/enzimologia , Neoplasias do Sistema Nervoso Central/genética , Criança , Pré-Escolar , Modelos Animais de Doenças , Hemangioma Cavernoso do Sistema Nervoso Central/enzimologia , Hemangioma Cavernoso do Sistema Nervoso Central/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Lactente , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Queratina-1/genética , Proteínas de Membrana/metabolismo , Camundongos , Pessoa de Meia-Idade , Estudos Prospectivos , Proteínas Proto-Oncogênicas/metabolismo , Fibras de Estresse/efeitos dos fármacos , Fibras de Estresse/metabolismo , Adulto Jovem , Quinases Associadas a rho/antagonistas & inibidores
11.
Mol Ther ; 22(1): 140-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24002694

RESUMO

Treatment options of glioblastoma multiforme are limited due to the blood-brain barrier (BBB). In this study, we investigated the utility of intranasal (IN) delivery as a means of transporting stem cell-based antiglioma therapeutics. We hypothesized that mesenchymal stem cells (MSCs) delivered via nasal application could impart therapeutic efficacy when expressing TNF-related apoptosis-inducing ligand (TRAIL) in a model of human glioma. ¹¹¹In-oxine, histology and magnetic resonance imaging (MRI) were utilized to track MSCs within the brain and associated tumor. We demonstrate that MSCs can penetrate the brain from nasal cavity and infiltrate intracranial glioma xenografts in a mouse model. Furthermore, irradiation of tumor-bearing mice tripled the penetration of (¹¹¹In)-oxine-labeled MSCs in the brain with a fivefold increase in cerebellum. Significant increase in CXCL12 expression was observed in irradiated xenograft tissue, implicating a CXCL12-dependent mechanism of MSCs migration towards irradiated glioma xenografts. Finally, MSCs expressing TRAIL improved the median survival of irradiated mice bearing intracranial U87 glioma xenografts in comparison with nonirradiated and irradiated control mice. Cumulatively, our data suggest that IN delivery of stem cell-based therapeutics is a feasible and highly efficacious treatment modality, allowing for repeated application of modified stem cells to target malignant glioma.


Assuntos
Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Rastreamento de Células , Quimiocina CXCL12/genética , Modelos Animais de Doenças , Raios gama , Expressão Gênica , Glioma/mortalidade , Glioma/patologia , Glioma/terapia , Humanos , Imageamento por Ressonância Magnética , Camundongos , Compostos Organometálicos , Oxiquinolina/análogos & derivados , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Stroke ; 45(2): 598-601, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24302484

RESUMO

BACKGROUND AND PURPOSE: Hyperpermeability and iron deposition are 2 central pathophysiological phenomena in human cerebral cavernous malformation (CCM) disease. Here, we used 2 novel MRI techniques to establish a relationship between these phenomena. METHODS: Subjects with CCM disease (4 sporadic and 17 familial) underwent MRI imaging using the dynamic contrast-enhanced quantitative perfusion and quantitative susceptibility mapping techniques that measure hemodynamic factors of vessel leak and iron deposition, respectively, previously demonstrated in CCM disease. Regions of interest encompassing the CCM lesions were analyzed using these techniques. RESULTS: Susceptibility measured by quantitative susceptibility mapping was positively correlated with permeability of lesions measured using dynamic contrast-enhanced quantitative perfusion (r=0.49; P≤0.0001). The correlation was not affected by factors, including lesion volume, contrast agent, and the use of statin medication. Susceptibility was correlated with lesional blood volume (r=0.4; P=0.0001) but not with lesional blood flow. CONCLUSIONS: The correlation between quantitative susceptibility mapping and dynamic contrast-enhanced quantitative perfusion suggests that the phenomena of permeability and iron deposition are related in CCM; hence, more leaky lesions also manifest a more cumulative iron burden. These techniques might be used as biomarkers to monitor the course of this disease and the effect of therapy.


Assuntos
Permeabilidade Capilar/fisiologia , Hemangioma Cavernoso do Sistema Nervoso Central/patologia , Adolescente , Adulto , Biomarcadores , Mapeamento Encefálico , Criança , Meios de Contraste , Interpretação Estatística de Dados , Feminino , Gadolínio , Humanos , Processamento de Imagem Assistida por Computador , Ferro/metabolismo , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Variações Dependentes do Observador , Adulto Jovem
13.
Small ; 10(24): 5137-50, 2014 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-25104165

RESUMO

The blood-brain barrier (BBB) remains a formidable obstacle in medicine, preventing efficient penetration of chemotherapeutic and diagnostic agents to malignant gliomas. Here, a transactivator of transcription (TAT) peptide-modified gold nanoparticle platform (TAT-Au NP) with a 5 nm core size is demonstrated to be capable of crossing the BBB efficiently and delivering cargoes such as the anticancer drug doxorubicin (Dox) and Gd(3+) contrast agents to brain tumor tissues. Treatment of mice bearing intracranial glioma xenografts with pH-sensitive Dox-conjugated TAT-Au NPs via a single intravenous administration leads to significant survival benefit when compared to the free Dox. Furthermore, it is demonstrated that TAT-Au NPs are capable of delivering Gd(3+) chelates for enhanced brain tumor imaging with a prolonged retention time of Gd(3+) when compared to the free Gd(3+) chelates. Collectively, these results show promising applications of the TAT-Au NPs for enhanced malignant brain tumor therapy and non-invasive imaging.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Barreira Hematoencefálica , Neoplasias Encefálicas/tratamento farmacológico , Doxorrubicina/uso terapêutico , Glioma/tratamento farmacológico , Ouro/química , Nanopartículas Metálicas , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacocinética , Neoplasias Encefálicas/patologia , Meios de Contraste , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Glioma/patologia , Imageamento por Ressonância Magnética , Camundongos
14.
Int J Biol Macromol ; 270(Pt 1): 132028, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38704066

RESUMO

Clinical therapy for widespread infections caused by Streptococcus pneumoniae (S. pneumoniae), such as community-acquired pneumonia, is highly challenging. As an important bacterial toxin, hydrogen peroxide (H2O2) secreted by S. pneumoniae can suppress the host's immune system and cause more severe disease. To address this problem, a hyaluronic acid (HA)-coated inorganic catalase-driven Janus nanomotor was developed, which can cleverly utilize and decompose H2O2 to reduce the burden of bacterial infection, and have excellent drug loading capacity. HA coating prevents rapid leakage of loaded antibiotics and improves the biocompatibility of the nanomaterials. The Janus nanomotor converted H2O2 into oxygen (O2), gave itself the capacity to move actively, and encouraged widespread dispersion in the lesion site. Encouragingly, animal experiments demonstrated that the capability of the nanomotors to degrade H2O2 contributes to diminishing the proliferation of S. pneumoniae and lung tissue damage. This self-propelled drug delivery platform provides a new therapeutic strategy for infections with toxin-secreting bacteria.


Assuntos
Catalase , Ácido Hialurônico , Peróxido de Hidrogênio , Streptococcus pneumoniae , Ácido Hialurônico/química , Catalase/metabolismo , Catalase/química , Streptococcus pneumoniae/efeitos dos fármacos , Animais , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Antibacterianos/farmacologia , Antibacterianos/química , Camundongos , Nanoestruturas/química , Humanos , Pneumonia/tratamento farmacológico
15.
Int J Biol Macromol ; 271(Pt 2): 132536, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38777021

RESUMO

The enhancement of antimicrobial wound dressings is of utmost importance in light of the escalating risk of antibiotic resistance caused by excessive antibiotic usage. Conventional antimicrobial materials eradicate pathogenic bacteria while impeding the proliferation of beneficial bacteria during the management of wound infections, thereby disturbing the equilibrium of the skin micro-ecosystem and engendering recurrent cutaneous complications. Lactobacillus rhamnosus (L.rha) is a probiotic that can inhibit the growth of certain pathogenic bacteria by secreting a large number of metabolites. In this paper, we synthesized a cross-linker (SPBA) with a boric acid molecule from succinic acid and 4-(bromomethyl)phenylboronic acid, which formed a boric acid ester bond with a diol on the natural polysaccharide sodium alginate (SA), and obtained a pH/reactive oxygen species (ROS) dual-responsive hydrogel (SA-SPBA) for loading L.rha to treat wound infections. The SA-SPBA@L.rha hydrogel improves the survival of L.rha during storage and has good injectability as well as self-healing properties. The hydrogel showed good biocompatibility, the antimicrobial effect increases in a dose-dependent manner, and it has a certain antioxidant and anti-inflammatory capacity, accelerating wound repair. The use of SA-SPBA@L.rha hydrogel provides a safe and effective strategy for the repair of skin wound infections.


Assuntos
Alginatos , Antibacterianos , Hidrogéis , Espécies Reativas de Oxigênio , Infecção dos Ferimentos , Alginatos/química , Hidrogéis/química , Hidrogéis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Concentração de Íons de Hidrogênio , Animais , Infecção dos Ferimentos/tratamento farmacológico , Infecção dos Ferimentos/microbiologia , Camundongos , Antibacterianos/farmacologia , Antibacterianos/química , Lacticaseibacillus rhamnosus/química , Cicatrização/efeitos dos fármacos , Humanos , Antioxidantes/farmacologia , Antioxidantes/química
16.
ACS Appl Mater Interfaces ; 16(15): 18400-18410, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38576193

RESUMO

Drug-resistant bacterial infection and biofilm formation are the key inhibitors of wound healing, and new strategies are urgently needed to address these issues. In this study, we designed a pH-responsive co-assembled peptide hydrogel to inhibit Methicillin-resistant Staphylococcus aureus (MRSA) infection and promote wound healing. We synthesized a cationic short peptide (Nap-FFKKK) and a co-assembled hydrogel with curcumin at pH ∼ 7.8. The loaded curcumin was continuously released in a weak acid environment (pH ∼ 5.5). The lysine-rich cationic peptide inhibited biofilm formation in MRSA via electrostatic interaction with the negatively charged bacterial cell surface and, thus, provided a reinforcing antibacterial effect with curcumin. In vitro antibacterial experiments showed that the co-assembled system considerably reduced the minimum inhibitory concentration of curcumin against MRSA by 10-fold and promoted wound healing in a mouse model of MRSA-infected wounds. This study provides a simple and promising strategy to treat drug-resistant bacterial infections in wounds.


Assuntos
Infecções Bacterianas , Curcumina , Staphylococcus aureus Resistente à Meticilina , Infecção dos Ferimentos , Animais , Camundongos , Hidrogéis , Antibacterianos , Peptídeos , Cicatrização , Concentração de Íons de Hidrogênio
17.
Small ; 9(24): 4123-9, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-23873826

RESUMO

A 3-step glioblastoma-tropic delivery and therapy method using nanoparticle programmed self-destructive neural stem cells (NSCs) is demonstrated in vivo: 1) FDA-approved NSCs for clinical trials are loaded with pH-sensitive MSN-Dox; 2) the nanoparticle conjugates provide a delayed drug-releasing mechanism and allow for NSC migration towards a distant tumor site; 3) NSCs eventually undergo cell death and release impregnated MSN-Dox, which subsequently induces toxicity towards surrounding glioma cells.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Nanopartículas/química , Células-Tronco Neurais/citologia , Animais , Apoptose , Morte Celular , Linhagem Celular Tumoral , Movimento Celular , Ensaios Clínicos como Assunto , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Humanos , Concentração de Íons de Hidrogênio , Lisossomos , Camundongos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Nanomedicina , Transplante de Neoplasias , Células-Tronco Neurais/ultraestrutura
18.
Adv Mater ; 35(38): e2302059, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37318939

RESUMO

Perovskite light-emitting diodes (PeLEDs) emerge as a promising class of optoelectronic devices for next-generation displays and lighting technology. However, the performance of blue PeLEDs lags far behind that of their green and red counterparts, including the unachieved trade-off between high efficiency and high luminance, severe efficiency roll-off, and unsatisfactory power efficiency. Here, a multi-functional chiral ligand of L-phenylalanine methyl ester hydrochloride is strategically introduced into quasi-2D perovskites, which can effectively passivate defects, modulate the phase distribution, improve photoluminescence quantum yield, guarantee high-quality film morphology, and enhance charge transport. Furthermore, ladder-like hole transport layers are established, boosting charge injection and balance. The resultant sky-blue PeLEDs (the photoluminescence peak is 493 nm and the electroluminescence peak is 497 nm) exhibit an external quantum efficiency of 12.43% at 1000 cd m-2 and a record power efficiency of 18.42 lm W-1 , rendering that the performance is among the best blue PeLEDs.

19.
J Colloid Interface Sci ; 607(Pt 2): 1919-1927, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34695740

RESUMO

Schottky-contacted nanosensors have attracted extensive attention due to their high sensitivity and fast response time. In this article, we proved that the construction of Schottky contact by Pt nanoparticles (NPs) decoration can effectively improve the performance of V2O5 nanobelts photodetectors. After modified by Pt NPs, the photocurrent of V2O5 nanobelts is increased by more than two orders of magnitude, and the photoresponse speed is improved by at least three orders of magnitude. Detailed studies have shown that the performance enhancement is attributed to the formation of the Schottky contact at the electrode-semiconductor interface due to the decrease of surface gas adsorption and the increase of V2O5 work function after Pt NPs modification. The strong built-in field in the Schottky barrier region will quickly separate photogenerated carriers, thereby reducing the electron-hole recombination rate, resulting in the fast response time and an increase in the free carrier density. Moreover, it is found that this enhancement effect can be regulated by controlling the pressure to modulating the Schottky barrier height at the interface. Overall, the Pt NPs-modified V2O5 nanobelts photodetector exhibits a broad response spectrum (visible to near infrared), fast rise/fall response time (less than 6.12/6.15 ms), high responsivity (5.6 A/W), and high specific detectivity (6.9 × 108 Jones). This study demonstrates the feasibility of building a Schottky barrier to enhance the photodetection performance, which provides a general and effective strategy towards the construction and its practical application of supersensitive and fast-response nanosensors.

20.
Nanoscale ; 14(16): 6228-6238, 2022 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-35403635

RESUMO

Over the past decade, 2D elemental semiconductors have emerged as an ever-increasingly important group in the 2D material family due to their simple crystal structures and compositions, and versatile physical properties. Taking advantage of the relatively small bandgap, outstanding carrier mobility, high air-stability and strong interactions with light, 2D tellurium (Te) has emerged as a compelling candidate for use in ultra-broadband photoelectric technologies. In this study, high-quality centimeter-scale Te nanofilms have been successfully produced by exploiting pulsed-laser deposition (PLD). By performing deposition on pre-patterned SiO2/Si substrates, a Te/Si 2D/3D heterojunction array is formed in situ. To our delight, taking advantage of the relatively small bandgap of Te, the Te/Si photodetectors demonstrate an ultra-broadband photoresponse from ultraviolet to near-infrared (370.6 nm to 2240 nm), enabling them to serve as important alternatives to conventional 2D materials such as MoS2. In addition, an outstanding on/off ratio of ∼108 and a fast response rate (a response/recovery time of 3.7 ms/4.4 ms) are achieved, which is associated with the large band offset and strong interfacial built-in electric field that contribute to suppressing the dark current and separating photocarriers. Beyond these, a 35 × 35 matrix array has been successfully constructed, where the devices exhibit comparable properties, with a production yield of 100% for 100 randomly tested devices. The average responsivity, external quantum efficiency and detectivity reach 249 A W-1, 76 350% and 1.15 × 1011 Jones, respectively, making the Te/Si devices among the best-performing 2D/3D heterojunction photodetectors. On the whole, this study has established that PLD is a promising technique for producing high-quality Te nanofilms with good scalability, and the Te/Si 2D/3D heterojunction provides a promising platform for implementing high-performance ultra-broadband photoelectronic technologies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA