Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Clin Cancer Res ; 27(20): 5669-5680, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34433651

RESUMO

PURPOSE: The epigenetic mechanisms involved in transcriptional regulation leading to malignant phenotype in gliomas remains poorly understood. Topoisomerase IIB (TOP2B), an enzyme that decoils and releases torsional forces in DNA, is overexpressed in a subset of gliomas. Therefore, we investigated its role in epigenetic regulation in these tumors. EXPERIMENTAL DESIGN: To investigate the role of TOP2B in epigenetic regulation in gliomas, we performed paired chromatin immunoprecipitation sequencing for TOP2B and RNA-sequencing analysis of glioma cell lines with and without TOP2B inhibition and in human glioma specimens. These experiments were complemented with assay for transposase-accessible chromatin using sequencing, gene silencing, and mouse xenograft experiments to investigate the function of TOP2B and its role in glioma phenotypes. RESULTS: We discovered that TOP2B modulates transcription of multiple oncogenes in human gliomas. TOP2B regulated transcription only at sites where it was enzymatically active, but not at all native binding sites. In particular, TOP2B activity localized in enhancers, promoters, and introns of PDGFRA and MYC, facilitating their expression. TOP2B levels and genomic localization was associated with PDGFRA and MYC expression across glioma specimens, which was not seen in nontumoral human brain tissue. In vivo, TOP2B knockdown of human glioma intracranial implants prolonged survival and downregulated PDGFRA. CONCLUSIONS: Our results indicate that TOP2B activity exerts a pleiotropic role in transcriptional regulation of oncogenes in a subset of gliomas promoting a proliferative phenotype.


Assuntos
Neoplasias Encefálicas/genética , DNA Topoisomerases Tipo II/fisiologia , Epigênese Genética/fisiologia , Glioma/genética , Íntrons/fisiologia , Oncogenes/fisiologia , Proteínas de Ligação a Poli-ADP-Ribose/fisiologia , Regiões Promotoras Genéticas/fisiologia , Animais , Neoplasias Encefálicas/enzimologia , Regulação Neoplásica da Expressão Gênica , Glioma/enzimologia , Humanos , Camundongos
3.
Nat Cancer ; 2(12): 1372-1386, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-35121903

RESUMO

Only a subset of recurrent glioblastoma (rGBM) responds to anti-PD-1 immunotherapy. Previously, we reported enrichment of BRAF/PTPN11 mutations in 30% of rGBM that responded to PD-1 blockade. Given that BRAF and PTPN11 promote MAPK/ERK signaling, we investigated whether activation of this pathway is associated with response to PD-1 inhibitors in rGBM, including patients that do not harbor BRAF/PTPN11 mutations. Here we show that immunohistochemistry for ERK1/2 phosphorylation (p-ERK), a marker of MAPK/ERK pathway activation, is predictive of overall survival following adjuvant PD-1 blockade in two independent rGBM patient cohorts. Single-cell RNA-sequencing and multiplex immunofluorescence analyses revealed that p-ERK was mainly localized in tumor cells and that high-p-ERK GBMs contained tumor-infiltrating myeloid cells and microglia with elevated expression of MHC class II and associated genes. These findings indicate that ERK1/2 activation in rGBM is predictive of response to PD-1 blockade and is associated with a distinct myeloid cell phenotype.


Assuntos
Glioblastoma , Glioblastoma/tratamento farmacológico , Humanos , Imunoterapia , Sistema de Sinalização das MAP Quinases , Recidiva Local de Neoplasia/tratamento farmacológico , Fosforilação
4.
Clin Cancer Res ; 26(2): 477-486, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31831565

RESUMO

PURPOSE: Paclitaxel shows little benefit in the treatment of glioma due to poor penetration across the blood-brain barrier (BBB). Low-intensity pulsed ultrasound (LIPU) with microbubble injection transiently disrupts the BBB allowing for improved drug delivery to the brain. We investigated the distribution, toxicity, and efficacy of LIPU delivery of two different formulations of paclitaxel, albumin-bound paclitaxel (ABX) and paclitaxel dissolved in cremophor (CrEL-PTX), in preclinical glioma models. EXPERIMENTAL DESIGN: The efficacy and biodistribution of ABX and CrEL-PTX were compared with and without LIPU delivery. Antiglioma activity was evaluated in nude mice bearing intracranial patient-derived glioma xenografts (PDX). Paclitaxel biodistribution was determined in sonicated and nonsonicated nude mice. Sonications were performed using a 1 MHz LIPU device (SonoCloud), and fluorescein was used to confirm and map BBB disruption. Toxicity of LIPU-delivered paclitaxel was assessed through clinical and histologic examination of treated mice. RESULTS: Despite similar antiglioma activity in vitro, ABX extended survival over CrEL-PTX and untreated control mice with orthotropic PDX. Ultrasound-mediated BBB disruption enhanced paclitaxel brain concentration by 3- to 5-fold for both formulations and further augmented the therapeutic benefit of ABX. Repeated courses of LIPU-delivered CrEL-PTX and CrEL alone were lethal in 42% and 37.5% of mice, respectively, whereas similar delivery of ABX at an equivalent dose was well tolerated. CONCLUSIONS: Ultrasound delivery of paclitaxel across the BBB is a feasible and effective treatment for glioma. ABX is the preferred formulation for further investigation in the clinical setting due to its superior brain penetration and tolerability compared with CrEL-PTX.


Assuntos
Albuminas/farmacologia , Albuminas/farmacocinética , Composição de Medicamentos/métodos , Sistemas de Liberação de Medicamentos/métodos , Glioma/tratamento farmacológico , Paclitaxel/farmacologia , Paclitaxel/farmacocinética , Polietilenoglicóis/química , Ultrassonografia/métodos , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Feminino , Glioma/patologia , Masculino , Camundongos , Camundongos Nus , Microbolhas/uso terapêutico , Nanopartículas/química , Taxa de Sobrevida , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Neurobiol ; 56(9): 6645-6653, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30911935

RESUMO

For normal functioning, the brain requires an adequate supply of blood. The components of normal brain vasculature are collectively referred to as the neurovascular unit. When the brain develops pathology, the structural and functional components of brain vasculature become compromised. This is evidenced in the case of neoplasia where the integrity of the vasculature is co-opted to further the neoplastic processes that require exponential blood resupply in order to facilitate the diffusion radius of tumor growth. Glioblastoma changes the brain vasculature in such a way that advances the tumor's progress while making it more resistant to standard modes of treatment. While the brain vasculature is changed as a result of glioblastoma growth and progression, it is also changed to advance the invasiveness of the tumor. The diagnostic criteria for glioblastoma is correlated with advanced neovascularization processes that change the previously existing vasculature into that which is morphologically atypical and in a dysfunctional state. Advancing therapies to treat glioblastoma must understand normal brain vasculature and how it is changed as a result of tumor growth.


Assuntos
Neoplasias Encefálicas/patologia , Encéfalo/irrigação sanguínea , Glioblastoma/patologia , Animais , Resistencia a Medicamentos Antineoplásicos , Humanos
6.
J Neurooncol ; 142(2): 375-384, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30706176

RESUMO

PURPOSE: To study whether the clinical outcome and molecular biology of gliomas in African-American patients fundamentally differ from those occurring in Whites. METHODS: The clinical information and molecular profiles (including gene expression array, non-silent somatic mutation, DNA methylation and protein expression) were downloaded from The Cancer genome atlas (TCGA). Electronic medical records were abstracted from Northwestern Medicine Enterprise Data Warehouse (NMEDW) for analysis as well. Grade II-IV Glioma patients were all included. RESULTS: 931 Whites and 64 African-American glioma patients from TCGA were analyzed. African-American with Karnofsky performance score (KPS) ≥ 80 have significantly lower risk of death than similar white Grade IV Glioblastoma (GBM) patients [HR (95% CI) = 0.47 (0.23, 0.98), P = 0.0444, C-index = 0.68]. Therefore, we further compared gene expression profiles between African-American GBM patients and Whites with KPS ≥ 80. Extrapolation of genes significantly associated with increased African-American patient survival revealed a set of 13 genes with a possible role in this association, including elevated expression of genes previously identified as increased in African-American breast and colon cancer patients (e.g. CRYBB2). Furthermore, gene set enrichment analysis revealed retinoic acid (RA) metabolism as a pathway significantly upregulated in African-American GBM patients who survive longer than Whites (Z-score = - 2.10, Adjusted P-value = 0.0449). CONCLUSIONS: African Americans have prolonged survival with glioma which is influenced only by initial KPS score. Genes previously associated with both racial disparities in cancer and pathways associated with RA metabolism may play an important role in glioma etiology. In the future exploration of these genes and pathways may inform novel therapies for this incurable disease.


Assuntos
Neoplasias Encefálicas/epidemiologia , Neoplasias Encefálicas/genética , Glioma/epidemiologia , Glioma/genética , Tretinoína/metabolismo , Adulto , Negro ou Afro-Americano/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Metilação de DNA , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Glioma/terapia , Humanos , Avaliação de Estado de Karnofsky , Masculino , Pessoa de Meia-Idade , Mutação , Gradação de Tumores , Análise de Sobrevida , Resultado do Tratamento , População Branca/genética
7.
Mol Neurobiol ; 56(7): 5032-5040, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30460615

RESUMO

The immunosuppressive microenvironment is one of the major factors promoting the growth of glioblastoma multiforme (GBM). Infiltration of CD4+CD25+Foxp3+ regulatory T cells (Tregs) into the tumor microenvironment plays a significant role in the suppression of the anti-tumor immunity and portends a dismal prognosis for patients. Glioma-mediated secretion of chemo-attractant C-C motif ligand 2 and 22 (CCL2/22) has previously been shown by our group to promote Treg migration in vitro. In this study, we show that a local implantation of platelet-rich fibrin patch (PRF-P) into the brain of GL261 glioma-bearing mice prolonged the survival of affected animals by 42.85% (p = 0.0011). Analysis performed on brain tumor tissue harvested from PRF-P-treated mice revealed a specific decrease in intra-tumoral lymphocytes with a preferential depletion of immunosuppressive Tregs. Importantly, co-culture of GL261 or chemo-attractants (CCL2/22) with PRF-P abrogated Treg migration. Pharmacological blockade of the CCL2/22 interaction with their receptors potentiated the inhibitory effect of PRF-P on Tregs recruitment in culture. Moreover, our findings revealed the soluble CD40 ligand (sCD40L) as a major Treg inhibitory player produced by activated platelets entrapped within the fibrin matrix of the PRF-P. Blockade of sCD40L restored the migratory capacity of Tregs, emphasizing the role of PRF-P in preventing the Treg migration to glioma tissue. Our findings highlight autologous PRF-P as a personalized, Treg-selective suppression platform that can potentially supplement and enhance the efficacy of glioma therapies.


Assuntos
Autoenxertos , Neoplasias Encefálicas/terapia , Glioma/terapia , Fibrina Rica em Plaquetas/fisiologia , Linfócitos T Reguladores/imunologia , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Craniotomia/métodos , Glioma/imunologia , Glioma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T Reguladores/metabolismo , Microambiente Tumoral/imunologia
8.
Neurotherapeutics ; 15(4): 1127-1138, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30027430

RESUMO

Antitumor immunotherapeutic strategies represent an especially promising set of approaches with rapid translational potential considering the dismal clinical context of high-grade gliomas. Dendritic cells (DCs) are the body's most professional antigen-presenting cells, able to recruit and activate T cells to stimulate an adaptive immune response. In this regard, specific loading of tumor-specific antigen onto dendritic cells potentially represents one of the most advanced strategies to achieve effective antitumor immunization. In this study, we developed a DC-specific adenoviral (Ad) vector, named Ad5scFvDEC205FF, targeting the DC surface receptor, DEC205. In vitro analysis shows that 60% of DCs was infected by this vector while the infectivity of other control adenoviral vectors was less than 10%, demonstrating superior infectivity on DCs. Moreover, an average of 14% of DCs were infected by Ad5scFvDEC205FF-GFP, while less than 3% of non-DCs were infected following in vivo administration, demonstrating highly selective in vivo DC infection. Importantly, vaccination with this vehicle expressing human glioma-specific antigen, Ad5scFvDEC205FF-CMV-IE, shows a prolonged survival benefit in GL261CMV-IE-implanted murine glioma models (p < 0.0007). Furthermore, when rechallenged, cancerous cells were completely rejected. In conclusion, our novel, viral-mediated, DC-based immunization approach has the significant therapeutic potential for patients with high-grade gliomas.


Assuntos
Imunidade Adaptativa/genética , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Neoplasias Encefálicas , Células Dendríticas/imunologia , Glioma , Lectinas Tipo C/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Receptores de Superfície Celular/metabolismo , Adenoviridae/genética , Análise de Variância , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Células Dendríticas/virologia , Modelos Animais de Doenças , Vetores Genéticos/metabolismo , Glioma/imunologia , Glioma/patologia , Glioma/terapia , Células HEK293 , Humanos , Linfonodos/citologia , Camundongos , Baço/citologia , Transdução Genética , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Prog Neurol Surg ; 32: 112-123, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29990979

RESUMO

Despite many recent advances in the management of gliomas, such as aggressive surgical resection, chemoradiotherapy, antiangiogenic therapy, and molecular targeted therapy, the survival of patients with high-grade neoplasms remains dismal. Gene therapy and oncolytic virotherapy have emerged as highly promising strategies for treatment of malignant brain tumors due to recent progress in understanding of the underlying cancer biology as well as improved techniques for genetic modification of potential therapeutics.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/terapia , Terapia Viral Oncolítica/métodos , Humanos
10.
Oncotarget ; 8(51): 89391-89405, 2017 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-29179527

RESUMO

Glioblastoma is a highly aggressive malignant brain tumor with a poor prognosis and the median survival 14.6 months. Immunomodulatory proteins and oncolytic viruses represent two treatment approaches that have recently been developed for patients with glioblastoma that could extend patient survival and result in better treatment outcomes for patients with this disease. Together, these approaches could potentially augment the treatment efficacy and strength of these anti-tumor therapies. In addition to oncolytic activities, this combinatory approach introduces immunomodulation locally only where cancerous cells are present. This thereby results in the change of the tumor microenvironment from immune-suppressive to immune-vulnerable via activation of cytotoxic T cells or through the removal of glioma cells immune-suppressive capability. This review discusses the strengths and weaknesses of adenoviral oncolytic therapy, and highlights the genetic modifications that result in more effective and targeted viral agents. Additionally, the mechanism of action of immune-activating agents is described and the results of previous clinical trials utilizing these treatments in other solid tumors are reviewed. The feasibility, synergy, and limitations for treatments that combine these two approaches are outlined and areas for which more work is needed are considered.

11.
Mol Ther Oncolytics ; 5: 97-104, 2017 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-28573184

RESUMO

Oncolytic virotherapy is a treatment approach with increasing clinical relevance, as indicated by the marked survival benefit seen in animal models and its current exploration in human patients with cancer. The use of an adenovirus vector for this therapeutic modality is common, has significant clinical benefit in animals, and its efficacy has recently been linked to an anti-tumor immune response that occurs following tumor antigen presentation. Here, we analyzed the adaptive immune system's response following viral infection by comparing replication-incompetent and replication-competent adenoviral vectors. Our findings suggest that cell death caused by replication-competent adenoviral vectors is required to induce a significant anti-tumor immune response and survival benefits in immunocompetent mice bearing intracranial glioma. We observed significant changes in the repertoire of immune cells in the brain and draining lymph nodes and significant recruitment of CD103+ dendritic cells (DCs) in response to oncolytic adenoviral therapy, suggesting the active role of the immune system in anti-tumor response. Our data suggest that the response to oncolytic virotherapy is accompanied by local and systemic immune responses and should be taken in consideration in the future design of the clinical studies evaluating oncolytic virotherapy in patients with glioblastoma multiforme (GBM).

12.
Methods Mol Biol ; 1382: 115-30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26611583

RESUMO

Adenoviral vectors have proven to be valuable resources in the development of novel therapies aimed at targeting pathological conditions of the central nervous system, including Alzheimer's disease and neoplastic brain lesions. Not only can some genetically engineered adenoviral vectors achieve remarkably efficient and specific gene delivery to target cells, but they also may act as anticancer agents by selectively replicating within cancer cells.Due to the great interest in using adenoviral vectors for various purposes, the need for a comprehensive protocol for viral vector production is especially apparent. Here, we describe the process of generating an adenoviral vector in its entirety, including the more complex process of adenoviral fiber modification to restrict viral tropism in order to achieve more efficient and specific gene delivery.


Assuntos
Adenoviridae/crescimento & desenvolvimento , Adenoviridae/genética , Adenoviridae/fisiologia , Vetores Genéticos/biossíntese , Células HEK293 , Humanos , Tropismo Viral
14.
Hum Gene Ther ; 26(9): 635-46, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26058317

RESUMO

The dismal clinical context of advanced-grade glioma demands the development of novel therapeutic strategies with direct patient impact. Adenovirus-mediated virotherapy represents a potentially effective approach for glioma therapy. In this research, we generated a novel glioma-specific adenovirus by instituting more advanced genetic modifications that can maximize the efficiency and safety of therapeutic adenoviral vectors. In this regard, a glioma-specific targeted fiber was developed through the incorporation of previously published glioma-specific, phage-panned peptide (VWT peptide) on a fiber fibritin-based chimeric fiber, designated as "GliomaFF." We showed that the entry of this virus was highly restricted to glioma cells, supporting the specificity imparted by the phage-panned peptide. In addition, the stability of the targeting moiety presented by fiber fibritin structure permitted greatly enhanced infectivity. Furthermore, the replication of this virus was restricted in glioma cells by controlling expression of the E1 gene under the activity of the tumor-specific survivin promoter. Using this approach, we were able to explore the combinatorial efficacy of various adenoviral modifications that could amplify the specificity, infectivity, and exclusive replication of this therapeutic adenovirus in glioma. Finally, virotherapy with this modified virus resulted in up to 70% extended survival in an in vivo murine glioma model. These data demonstrate that this novel adenoviral vector is a safe and efficient treatment for this difficult malignancy.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Glioma/terapia , Animais , Células CHO , Linhagem Celular Tumoral , Cricetinae , Cricetulus , Células HEK293 , Humanos , Masculino , Camundongos Nus , Transplante de Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Biblioteca de Peptídeos , Proteínas Virais/genética , Replicação Viral
15.
Curr Protoc Hum Genet ; 85: 13.11.1-13.11.9, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25827347

RESUMO

The use of stem cells (SCs) as carriers for therapeutic agents has now progressed to early clinical trials. These clinical trials exploring SC-mediated delivery of oncolytic adenoviruses will commence in the near future, hopefully yielding meritorious results that can provoke further scientific inquiry. Preclinical animal studies have demonstrated that SCs can be successfully loaded with conditionally-replicative adenoviruses and delivered to the tumor, whereupon they may evoke pronounced therapeutic efficacy. In this protocol, we describe the maintenance of SCs, provide an analysis of optimal adenoviral titers for SC loading, and evaluate the optimized viral loading on SCs.


Assuntos
Adenoviridae/metabolismo , Células-Tronco Neurais/virologia , Vírus Oncolíticos/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Técnicas Genéticas , Humanos
16.
Neuro Oncol ; 17 Suppl 2: ii24-ii36, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25746089

RESUMO

Gene therapy offers a multidimensional set of approaches intended to treat and cure glioblastoma (GBM), in combination with the existing standard-of-care treatment (surgery and chemoradiotherapy), by capitalizing on the ability to deliver genes directly to the site of neoplasia to yield antitumoral effects. Four types of gene therapy are currently being investigated for their potential use in treating GBM: (i) suicide gene therapy, which induces the localized generation of cytotoxic compounds; (ii) immunomodulatory gene therapy, which induces or augments an enhanced antitumoral immune response; (iii) tumor-suppressor gene therapy, which induces apoptosis in cancer cells; and (iv) oncolytic virotherapy, which causes the lysis of tumor cells. The delivery of genes to the tumor site is made possible by means of viral and nonviral vectors for direct delivery of therapeutic gene(s), tumor-tropic cell carriers expressing therapeutic gene(s), and "intelligent" carriers designed to increase delivery, specificity, and tumoral toxicity against GBM. These vehicles are used to carry genetic material to the site of pathology, with the expectation that they can provide specific tropism to the desired site while limiting interaction with noncancerous tissue. Encouraging preclinical results using gene therapies for GBM have led to a series of human clinical trials. Although there is limited evidence of a therapeutic benefit to date, a number of clinical trials have convincingly established that different types of gene therapies delivered by various methods appear to be safe. Due to the flexibility of specialized carriers and genetic material, the technology for generating new and more effective therapies already exists.


Assuntos
Neoplasias Encefálicas/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Glioblastoma/terapia , Animais , Neoplasias Encefálicas/genética , Terapia Genética/tendências , Vetores Genéticos/uso terapêutico , Glioblastoma/genética , Humanos , Resultado do Tratamento , Vírus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...