Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cancer ; 22(1): 129, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37563568

RESUMEN

BACKGROUND: This Phase 1 study evaluates the intra- and peritumoral administration by convection enhanced delivery (CED) of human recombinant Bone Morphogenetic Protein 4 (hrBMP4) - an inhibitory regulator of cancer stem cells (CSCs) - in recurrent glioblastoma. METHODS: In a 3 + 3 dose escalation design, over four to six days, fifteen recurrent glioblastoma patients received, by CED, one of five doses of hrBMP4 ranging from 0·5 to 18 mg. Patients were followed by periodic physical, neurological, blood testing, magnetic resonance imaging (MRI) and quality of life evaluations. The primary objective of this first-in-human study was to determine the safety, dose-limiting toxicities (DLT) and maximum tolerated dose (MTD) of hrBMP4. Secondary objectives were to assess potential efficacy and systemic exposure to hrBMP4 upon intracerebral infusion. RESULTS: Intra- and peritumoral infusion of hrBMP4 was safe and well-tolerated. We observed no serious adverse events related to this drug. Neither MTD nor DLT were reached. Three patients had increased hrBMP4 serum levels at the end of infusion, which normalized within 4 weeks, without sign of toxicity. One patient showed partial response and two patients a complete (local) tumor response, which was maintained until the most recent follow-up, 57 and 30 months post-hrBMP4. Tumor growth was inhibited in areas permeated by hrBMP4. CONCLUSION: Local delivery of hrBMP4 in and around recurring glioblastoma is safe and well-tolerated. Three patients responded to the treatment. A complete response and long-term survival occurred in two of them. This warrants further clinical studies on this novel treatment targeting glioblastoma CSCs. TRIAL REGISTRATION: ClinicaTrials.gov identifier: NCT02869243.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Calidad de Vida , Proteína Morfogenética Ósea 4/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Encefálicas/patología , Dosis Máxima Tolerada
2.
Int J Cancer ; 153(3): 654-668, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37141410

RESUMEN

Glioblastoma (GB) is the most aggressive neoplasm of the brain. Poor prognosis is mainly attributed to tumor heterogeneity, invasiveness and drug resistance. Only a small fraction of GB patients survives longer than 24 months from the time of diagnosis (ie, long-term survivors [LTS]). In our study, we aimed to identify molecular markers associated with favorable GB prognosis as a basis to develop therapeutic applications to improve patients' outcome. We have recently assembled a proteogenomic dataset of 87 GB clinical samples of varying survival rates. Following RNA-seq and mass spectrometry (MS)-based proteomics analysis, we identified several differentially expressed genes and proteins, including some known cancer-related pathways and some less established that showed higher expression in short-term (<6 months) survivors (STS) compared to LTS. One such target found was deoxyhypusine hydroxylase (DOHH), which is known to be involved in the biosynthesis of hypusine, an unusual amino acid essential for the function of the eukaryotic translation initiation factor 5A (eIF5A), which promotes tumor growth. We consequently validated DOHH overexpression in STS samples by quantitative polymerase chain reaction (qPCR) and immunohistochemistry. We further showed robust inhibition of proliferation, migration and invasion of GB cells following silencing of DOHH with short hairpin RNA (shRNA) or inhibition of its activity with small molecules, ciclopirox and deferiprone. Moreover, DOHH silencing led to significant inhibition of tumor progression and prolonged survival in GB mouse models. Searching for a potential mechanism by which DOHH promotes tumor aggressiveness, we found that it supports the transition of GB cells to a more invasive phenotype via epithelial-mesenchymal transition (EMT)-related pathways.


Asunto(s)
Glioblastoma , Animales , Ratones , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/patología , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Ciclopirox , Sobrevivientes
3.
Biochem Biophys Res Commun ; 671: 124-131, 2023 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-37300942

RESUMEN

BACKGROUND: In the surgical management of glioblastoma, a highly aggressive and incurable type of brain cancer, identification and treatment of residual tissue is the most common site of disease recurrence. Monitoring and localized treatment are achieved with engineered microbubbles (MBs) by combining ultrasound and fluorescence imaging with actively targeted temozolomide (TMZ) delivery. METHODS: The MBs were conjugated with a near-infrared fluorescence probe CF790, cyclic pentapeptide bearing the RGD sequence and a carboxyl-temozolomide, TMZA. The efficiency of adhesion to HUVEC cells was assessed in vitro in realistic physiological conditions of shear rate and vascular dimensions. Cytotoxicity of TMZA-loaded MBs on U87 MG cells and IC50 were assessed by MTT tests. RESULTS: We report on the design of injectable poly(vinyl alcohol) echogenic MBs designed as a platform with active targeting ability to tumor tissues, by tethering on the surface a ligand having the tripeptide sequence, RGD. The biorecognition of RGD-MBs onto HUVEC cells is quantitatively proved. Efficient NIR emission from the CF790-decorated MBs was successfully detected. The conjugation on the MBs surface of a specific drug as TMZ is achieved. The pharmacological activity of the coupled-to-surface drug is preserved by controlling the reaction conditions. CONCLUSIONS: We present an improved formulation of PVA-MBs to achieve a multifunctional device with adhesion ability, cytotoxicity on glioblastoma cells and supporting imaging.


Asunto(s)
Glioblastoma , Glioma , Humanos , Glioblastoma/tratamiento farmacológico , Temozolomida/uso terapéutico , Medicina de Precisión , Línea Celular Tumoral , Recurrencia Local de Neoplasia , Glioma/terapia , Glioma/tratamiento farmacológico , Imagen Óptica , Oligopéptidos/uso terapéutico , Microburbujas
4.
Mol Psychiatry ; 27(3): 1848-1854, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34974525

RESUMEN

Creative thinking represents a major evolutionary mechanism that greatly contributed to the rapid advancement of the human species. The ability to produce novel and useful ideas, or original thinking, is thought to correlate well with unexpected, synchronous activation of several large-scale, dispersed cortical networks, such as the default network (DN). Despite a vast amount of correlative evidence, a causal link between default network and creativity has yet to be demonstrated. Surgeries for resection of brain tumors that lie in proximity to speech related areas are performed while the patient is awake to map the exposed cortical surface for language functions. Such operations provide a unique opportunity to explore human behavior while disrupting a focal cortical area via focal electrical stimulation. We used a novel paradigm of individualized direct cortical stimulation to examine the association between creative thinking and the DN. Preoperative resting-state fMRI was used to map the DN in individual patients. A cortical area identified as a DN node (study) or outside the DN (controls) was stimulated while the participants performed an alternate-uses-task (AUT). This task measures divergent thinking through the number and originality of different uses provided for an everyday object. Baseline AUT performance in the operating room was positively correlated with DN integrity. Direct cortical stimulation at the DN node resulted in decreased ability to produce alternate uses, but not in the originality of uses produced. Stimulation in areas that when used as network seed regions produced a network similar to the canonical DN was associated with reduction of creative fluency. Stimulation of areas that did not produce a default-like network (controls) did not alter creative thinking. This is the first study to causally link the DN and creative thinking.


Asunto(s)
Mapeo Encefálico , Creatividad , Encéfalo/fisiología , Mapeo Encefálico/métodos , Cognición/fisiología , Humanos , Imagen por Resonancia Magnética
5.
J Immunol ; 207(2): 709-719, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34215656

RESUMEN

Tumor-treating fields (TTFields) are a localized, antitumoral therapy using alternating electric fields, which impair cell proliferation. Combining TTFields with tumor immunotherapy constitutes a rational approach; however, it is currently unknown whether TTFields' locoregional effects are compatible with T cell functionality. Healthy donor PBMCs and viably dissociated human glioblastoma samples were cultured under either standard or TTFields conditions. Select pivotal T cell functions were measured by multiparametric flow cytometry. Cytotoxicity was evaluated using a chimeric Ag receptor (CAR)-T-based assay. Glioblastoma patient samples were acquired before and after standard chemoradiation or standard chemoradiation + TTFields treatment and examined by immunohistochemistry and by RNA sequencing. TTFields reduced the viability of proliferating T cells, but had little or no effect on the viability of nonproliferating T cells. The functionality of T cells cultured under TTFields was retained: they exhibited similar IFN-γ secretion, cytotoxic degranulation, and PD1 upregulation as controls with similar polyfunctional patterns. Glioblastoma Ag-specific T cells exhibited unaltered viability and functionality under TTFields. CAR-T cells cultured under TTFields exhibited similar cytotoxicity as controls toward their CAR target. Transcriptomic analysis of patients' glioblastoma samples revealed a significant shift in the TTFields-treated versus the standard-treated samples, from a protumoral to an antitumoral immune signature. Immunohistochemistry of samples before and after TTFields treatment showed no reduction in T cell infiltration. T cells were found to retain key antitumoral functions under TTFields settings. Our data provide a mechanistic insight and a rationale for ongoing and future clinical trials that combine TTFields with immunotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Glioblastoma/inmunología , Glioblastoma/terapia , Linfocitos T/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Terapia Combinada/métodos , Humanos , Inmunoterapia/métodos , Interferón gamma/metabolismo , Linfocitos T/inmunología , Transcriptoma/efectos de los fármacos
6.
J Neurooncol ; 157(1): 63-69, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35119589

RESUMEN

PURPOSE: Non-small cell lung cancer (NSCLC) tends to metastasize to the brain. Between 10 and 60% of NSCLCs harbor an activating mutation in the epidermal growth-factor receptor (EGFR), which may be targeted with selective EGFR inhibitors. However, due to a high discordance rate between the molecular profile of the primary tumor and the brain metastases (BMs), identifying an individual patient's EGFR status of the BMs necessitates tissue diagnosis via an invasive surgical procedure. We employed a deep learning (DL) method with the aim of noninvasive detection of the EGFR mutation status in NSCLC BM. METHODS: We retrospectively collected clinical, radiological, and pathological-molecular data of all the NSCLC patients who had been diagnosed with BMs and underwent resection of their BM during 2009-2019. The study population was then divided into two groups based upon EGFR mutational status. We further employed a DL technique to classify the two groups according to their preoperative magnetic resonance imaging features. Augmentation techniques, transfer learning approach, and post-processing of the predicted results were applied to overcome the relatively small cohort. Finally, we established the accuracy of our model in predicting EGFR mutation status of BM of NSCLC. RESULTS: Fifty-nine patients were included in the study, 16 patients harbored EGFR mutations. Our model predicted mutational status with mean accuracy of 89.8%, sensitivity of 68.7%, specificity of 97.7%, and a receiver operating characteristic curve value of 0.91 across the 5 validation datasets. CONCLUSION: DL-based noninvasive molecular characterization is feasible, has high accuracy and should be further validated in large prospective cohorts.


Asunto(s)
Neoplasias Encefálicas , Carcinoma de Pulmón de Células no Pequeñas , Aprendizaje Profundo , Neoplasias Pulmonares , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/patología , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación , Estudios Prospectivos , Estudios Retrospectivos
7.
Neurosurg Rev ; 43(2): 709-718, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31098789

RESUMEN

Guidelines on the administration of prophylactic antiepileptic drugs (AED), and specifically levetiracetam, for brain tumor surgery are still lacking. In this two-center matched cohort study, we aim to compare the proportion of postoperative seizures during follow-up after supratentorial tumor surgery in patients receiving no seizure prophylaxis, and those treated with levetiracetam perioperatively. Three hundred sixteen consecutive patients undergoing supratentorial tumor surgery, without history of seizures were included: 207 patients did not receive AED (no AED group), and 109 patients received levetiracetam perioperatively (levetiracetam group). The primary outcome measure was the rate of postoperative seizures. Additionally, uni- and multivariate analyses assessing possible risk factors for postoperative seizures were performed. No statistically significant difference for the occurrence of postoperative seizures was found between the two groups (10.1%, n = 21 in the no AED group vs. 9.2%, n = 10, in the levetiracetam group; p = 0.69, OR 0.9 [0.4-2.0), NNT 103 [12.9-17.1]). After propensity score matching, the primary outcome was observed in 13 patients (12.4%) from the no AED group and in 9 patients (8.6%) from the levetiracetam group (p = 0.50, OR 0.7 [0.3-1.6], NNT 26.3 [8.3-22.4]). Among all analyzed possible risk factors for postoperative seizures, only postoperative infarction showed a statistically significant association with higher seizure rates in multivariate analysis (OR 8.2 [1.1-60.6], p = 0.04). Prophylactic treatment with levetiracetam after brain tumor surgery showed no statistically significant effect in preventing postoperative seizures. However, in case a postoperative infarction occurs, its administration might be indicated.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Levetiracetam/uso terapéutico , Complicaciones Posoperatorias/prevención & control , Convulsiones/prevención & control , Neoplasias Supratentoriales/cirugía , Adulto , Anciano , Estudios de Cohortes , Craneotomía/efectos adversos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Complicaciones Posoperatorias/epidemiología , Estudios Retrospectivos , Convulsiones/epidemiología
8.
Acta Neuropathol ; 138(2): 295-308, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31069492

RESUMEN

DNA methylation patterns delineate clinically relevant subgroups of meningioma. We previously established the six meningioma methylation classes (MC) benign 1-3, intermediate A and B, and malignant. Here, we set out to identify subgroup-specific mutational patterns and gene regulation. Whole genome sequencing was performed on 62 samples across all MCs and WHO grades from 62 patients with matched blood control, including 40 sporadic meningiomas and 22 meningiomas arising after radiation (Mrad). RNA sequencing was added for 18 of these cases and chromatin-immunoprecipitation for histone H3 lysine 27 acetylation (H3K27ac) followed by sequencing (ChIP-seq) for 16 samples. Besides the known mutations in meningioma, structural variants were found as the mechanism of NF2 inactivation in a small subset (5%) of sporadic meningiomas, similar to previous reports for Mrad. Aberrations of DMD were found to be enriched in MCs with NF2 mutations, and DMD was among the most differentially upregulated genes in NF2 mutant compared to NF2 wild-type cases. The mutational signature AC3, which has been associated with defects in homologous recombination repair (HRR), was detected in both sporadic meningioma and Mrad, but widely distributed across the genome in sporadic cases and enriched near genomic breakpoints in Mrad. Compared to the other MCs, the number of single nucleotide variants matching the AC3 pattern was significantly higher in the malignant MC, which also exhibited higher genomic instability, determined by the numbers of both large segments affected by copy number alterations and breakpoints between large segments. ChIP-seq analysis for H3K27ac revealed a specific activation of genes regulated by the transcription factor FOXM1 in the malignant MC. This analysis also revealed a super enhancer near the HOXD gene cluster in this MC, which, together with general upregulation of HOX genes in the malignant MC, indicates a role of HOX genes in meningioma aggressiveness. This data elucidates the biological mechanisms rendering different epigenetic subgroups of meningiomas, and suggests leveraging HRR as a novel therapeutic target.


Asunto(s)
Metilación de ADN , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Meníngeas/clasificación , Meningioma/clasificación , Mutación , Inmunoprecipitación de Cromatina , Dosificación de Gen , Inestabilidad Genómica , Humanos , Neoplasias Meníngeas/etiología , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Meningioma/etiología , Meningioma/genética , Meningioma/patología , Proteínas de Neoplasias/genética , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/patología , Polimorfismo de Nucleótido Simple , ARN Mensajero/genética , ARN Neoplásico/genética , Reparación del ADN por Recombinación , Alineación de Secuencia , Factores de Transcripción/fisiología , Transcriptoma , Secuenciación Completa del Genoma
9.
FASEB J ; : fj201701568R, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29856660

RESUMEN

Glioblastoma is an aggressive and invasive brain malignancy with high mortality rates despite current treatment modalities. In this study, we show that a 7-gene signature, previously found to govern the switch of glioblastomas from dormancy to aggressive tumor growth, correlates with improved overall survival of patients with glioblastoma. Using glioblastoma dormancy models, we validated the role of 2 genes from the signature, thrombospondin-1 ( TSP-1) and epidermal growth factor receptor ( EGFR), as regulators of glioblastoma dormancy and explored their therapeutic potential. EGFR up-regulation was reversed using EGFR small interfering RNA polyplex, antibody, or small-molecule inhibitor. The diminished function of TSP-1 was augmented via a peptidomimetic. The combination of EGFR inhibition and TSP-1 restoration led to enhanced therapeutic efficacy in vitro, in 3-dimensional patient-derived spheroids, and in a subcutaneous human glioblastoma model in vivo. Systemic administration of the combination therapy to mice bearing intracranial murine glioblastoma resulted in marginal therapeutic outcomes, probably due to brain delivery challenges, p53 mutation status, and the aggressive nature of the selected cell line. Nevertheless, this study provides a proof of concept for exploiting regulators of tumor dormancy for glioblastoma therapy. This therapeutic strategy can be exploited for future investigations using a variety of therapeutic entities that manipulate the expression of dormancy-associated genes in glioblastoma as well as in other cancer types.-Tiram, G., Ferber, S., Ofek, P., Eldar-Boock, A., Ben-Shushan, D., Yeini, E., Krivitsky, A., Blatt, R., Almog, N., Henkin, J., Amsalem, O., Yavin, E., Cohen, G., Lazarovici, P., Lee, J. S., Ruppin, E., Milyavsky, M., Grossman, R., Ram, Z., Calderón, M., Haag, R., Satchi-Fainaro, R. Reverting the molecular fingerprint of tumor dormancy as a therapeutic strategy for glioblastoma.

10.
Eur Arch Otorhinolaryngol ; 276(9): 2491-2498, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31342144

RESUMEN

PURPOSE: The endoscopic endonasal approach is increasingly being used for resection and reconstruction of anterior skull base lesions. The vascularized nasoseptal flap (NSF) has become the workhorse for reconstruction of anterior skull base defects, resulting in a significant decrease in the incidence of cerebrospinal fluid (CSF) leak. This study presents a single center's experience with NSFs and reports associated complications. METHODS: Patients who underwent endoscopic skull base defect repair with a NSF between 2008 and 2014 were retrospectively evaluated. Complications reviewed were divided into major and minor. Major complications included new-onset and continuing CSF leak and meningitis. Minor complications included long-standing crust formation, synechia, epistaxis, septal perforation, sinusitis and anosmia. RESULTS: Of the 77 patients included in the study, 47 (61%) underwent trans-sphenoidal surgery for pituitary lesions during which CSF leak was observed. The other 30 patients underwent reconstructive surgeries for post-traumatic CSF leaks or extirpation of lesions involving the anterior skull base. A high-flow intra-operative CSF leak was observed in 25 patients (25/77, 32%). The median follow-up was 16 months (range 3-81 months). 9 patients had major complications and 27 patients had minor complications. Only high-flow intra-operative CSF leak correlated with major complications (p = 0.012). CONCLUSION: NSF is an extremely effective tool for skull base reconstruction. While it is associated with a low rate of major complications, minor complications are frequent and require local treatment, although they tend to resolve in the late postoperative period.


Asunto(s)
Pérdida de Líquido Cefalorraquídeo/etiología , Cirugía Endoscópica por Orificios Naturales/efectos adversos , Base del Cráneo/cirugía , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Niño , Preescolar , Femenino , Humanos , Incidencia , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Cirugía Endoscópica por Orificios Naturales/métodos , Procedimientos de Cirugía Plástica/efectos adversos , Procedimientos de Cirugía Plástica/métodos , Estudios Retrospectivos , Factores de Riesgo , Colgajos Quirúrgicos , Adulto Joven
11.
Isr Med Assoc J ; 21(7): 503, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31507132

RESUMEN

BACKGROUND: Tumor treating fields (TTFields) are low-intensity, intermediate frequency electric fields that affect proliferating cells. TTFields are FDA approved for treatment of newly diagnosed and recurrent glioblastoma. Combining TTFields with immunotherapy is a rational approach due to their different mechanisms of action (MOA) and to the ability of TTFields to induce immunogenic cell death. Conversely, TTFields may interfere with immune functions critical for effective T-cell responses. OBJECTIVES: To evaluate the effects of TTFields on pivotal antitumoral T-cell functions. METHODS: T-cells from healthy donor peripheral blood (PB) or from viably dissociated human glioblastoma samples were cultured under normal or TTFields conditions, with or without superantigen stimulation. Multiparametric flow cytometry (8-color) was used to assess T-cell responses by monitoring select pivotal functions: proliferation (CFSE), IFNγ secretion, cytotoxic degranulation (CD107a), and activation/exhaustion (PD-1). Cellular viability was assessed in a dedicated assay. A chimeric antigen receptor (CAR) T-cell-based assay directly evaluated cellular cytotoxicity. RESULTS: Activated PB T-cells and tumor-infiltrating T-cells (TILs) preserved all monitored anti- tumoral functions under TTFields, apart from proliferation. This finding also applied specifically to PD-1 + TILs, comprised predominantly of tumor antigen-specific cells. Activated T-cells that attempted to proliferate under TTFields demonstrated decreased viability, in line with TTField MOA. Small or no reduction in viability was found in T-cells that did not attempt to proliferate, whether activated or resting. CONCLUSIONS: All monitored anti-tumoral T cell functions, except for proliferation, were unhindered by TTFields. Our results support further investigation into combinations of TTFields with T-cell based immunotherapeutic approaches.


Asunto(s)
Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Terapia por Estimulación Eléctrica/métodos , Glioblastoma/terapia , Citometría de Flujo/métodos , Glioblastoma/patología , Humanos , Recurrencia Local de Neoplasia , Linfocitos T/citología
12.
Ann Surg Oncol ; 24(5): 1392-1398, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27896517

RESUMEN

BACKGROUND: Surgical resection and stereotactic radiosurgery (SRS) are well-established treatment options for selected patients with oligo-brain metastases (BMs). The dynamics of edema resolution with each treatment method have not been well characterized. METHODS: Of 389 patients treated for BMs between 2012 and 2014, this study retrospectively identified 107 patients (150 metastases) who underwent either surgery or SRS as a single treatment method for BMs. The two groups of patients were matched for clinical parameters. Volumetric assessments of the tumor and associated edema were performed before treatment and then 2-3 months after treatment. RESULTS: In this study, 76 surgical cases were compared with 74 cases treated with SRS. The volume of the tumor and surrounding edema was significantly greater in the surgery group than in the SRS group. However, resolution of edema was significantly more rapid in the surgical group (p < 0.0001), accompanied by faster weaning from steroids. After a matching process based on the propensity of a patient to receive SRS, a subgroup cohort was analyzed (mean maximal diameter: 21 mm in the surgical group vs 20.8 mm in the SRS group; p = 0.9). At diagnosis, edema volume, but not tumor volume, was significantly greater in the surgical group. The resolution of edema 2-3 months after treatment was better in the surgical group than in the SRS group (89.6% vs 71.1% of baseline, respectively; p = 0.09), although this difference did not reach the level of significance. CONCLUSIONS: Resolution of tumor-associated edema in BMs suitable for either surgery or SRS was significantly faster after surgical resection than after SRS. Accordingly, when both treatment options are suitable, surgery appears to induce faster resolution of the edema.


Asunto(s)
Edema Encefálico/etiología , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/terapia , Metastasectomía , Radiocirugia , Anciano , Edema Encefálico/terapia , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/patología , Cerebro , Femenino , Humanos , Masculino , Persona de Mediana Edad , Puntaje de Propensión , Estudios Retrospectivos , Factores de Tiempo , Carga Tumoral
13.
Ann Surg Oncol ; 24(3): 794-800, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27766560

RESUMEN

BACKGROUND: The extent of tumor resection (EOTR) calculated by enhanced T1 changes in glioblastomas has been previously reported to predict survival. However, fluid-attenuated inversion recovery (FLAIR) volume may better represent tumor burden. In this study, we report the first assessment of the dynamics of FLAIR volume changes over time as a predictive variable for post-resection overall survival (OS). METHODS: Contemporary data from 103 consecutive patients with complete imaging and clinical data who underwent resection of newly diagnosed glioblastoma followed by the Stupp protocol between 2010 and 2013 were analyzed. Clinical, radiographic, and outcome parameters were retrieved for each patient, including magnetic resonance imaging (MRI)-based volumetric tumor analysis before, immediately after, and 3 months post-surgery. RESULTS: OS rate was 17.6 months. A significant incremental OS advantage was noted, with as little as 85 % T1-weighted gadolinium-enhanced (T1Gd)-EOTR measured on contrast-enhanced MRI. Pre- and immediate postoperative FLAIR-based EOTR was not predictive of OS; however, abnormal FLAIR volume measured 3 months post-surgery correlated significantly with outcome when FLAIR residual tumor volume (RTV) was <19.3 cm3 and <46 % of baseline volume (p < 0.0001 for both). Age and isocitrate dehydrogenase (IDH)-1 mutation were predictive of OS (p < 0.0001, Cox proportional hazards). CONCLUSIONS: OS correlated with the immediate postoperative T1Gd-EOTR measured by enhanced T1 MRI, but not by FLAIR volume. Diminished abnormal FLAIR volume at 3 months post-surgery was associated with OS benefit when FLAIR-RTV was <19.3 cm3 or <46 % of baseline. These threshold values provide a new radiological variable that can be used for prediction of OS in patients with glioblastoma immediately after completion of standard chemoradiation.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/cirugía , Imagen de Difusión por Resonancia Magnética/métodos , Glioblastoma/diagnóstico por imagen , Glioblastoma/cirugía , Factores de Edad , Anciano , Neoplasias Encefálicas/patología , Medios de Contraste , Femenino , Gadolinio , Glioblastoma/patología , Humanos , Isocitrato Deshidrogenasa/genética , Masculino , Persona de Mediana Edad , Neoplasia Residual , Periodo Posoperatorio , Valor Predictivo de las Pruebas , Periodo Preoperatorio , Estudios Retrospectivos , Tasa de Supervivencia , Carga Tumoral
14.
Tumour Biol ; 39(4): 1010428317698357, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28443460

RESUMEN

Glioblastoma multiforme is the most common and aggressive primary brain tumor in adults. A mesenchymal phenotype was associated with tumor aggressiveness and poor prognosis in glioblastoma multiforme patients. Recently, the transcription factor RUNX1 was suggested as a driver of the glioblastoma multiforme mesenchymal gene expression signature; however, its independent role in this process is yet to be described. Here, we assessed the role of RUNX1 in U87 glioblastoma multiforme cells in correspondence to its mediated transcriptome and genome-wide occupancy pattern. Overexpression of RUNX1 led to diminished tumor growth in nude and severe combined immunodeficiency mouse xenograft tumor model. At the molecular level, RUNX1 occupied thousands of genomic regions and regulated the expression of hundreds of target genes, both directly and indirectly. RUNX1 occupied genomic regions that corresponded to genes that were shown to play a role in brain tumor progression and angiogenesis and upon overexpression led to a substantial down-regulation of their expression level. When overexpressed in U87 glioblastoma multiforme cells, RUNX1 down-regulated key pathways in glioblastoma multiforme progression including epithelial to mesenchymal transition, MTORC1 signaling, hypoxia-induced signaling, and TNFa signaling via NFkB. Moreover, master regulators of the glioblastoma multiforme mesenchymal phenotype including CEBPb, ZNF238, and FOSL2 were directly regulated by RUNX1. The data suggest a central role for RUNX1 as master regulator of gene expression in the U87 glioblastoma multiforme cell line and mark RUNX1 as a potential target for novel future therapies for glioblastoma multiforme.


Asunto(s)
Proliferación Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Transición Epitelial-Mesenquimal/genética , Glioblastoma/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/biosíntesis , Antígeno 2 Relacionado con Fos/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Humanos , Ratones , Proteínas Represoras/genética , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Childs Nerv Syst ; 33(12): 2169-2175, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28808787

RESUMEN

BACKGROUND: Incidental findings on neuroimaging in the pediatric population are an emerging treatment challenge. Treatment options for these incidental childhood brain mass lesions, which radiologically may be assumed to be low-grade gliomas (LGG), vary, ranging from careful conservative "wait and scan" treatment to surgical biopsy, gross total resection, and upfront radiation and/or chemotherapy. As malignant transformation of LGG in children is extremely rare, some series advocate careful conservative management of these lesions; however, universal treatment protocols are not totally agreed upon. ILLUSTRATIVE CASE: We present the case of a 10-year-old boy with a fronto-basal incidental cerebral mass lesion, suspected to be a low-grade glial neoplasm. Initially, magnetic resonance imaging (MRI) was done to rule out a pathology causing his growth to be delayed. A treatment with growth hormone was initiated. After close clinical and radiological follow-up of this asymptomatic lesion for 6 years, a minimal growth of the lesion was seen, which we decided to continue following. After 7 years, a clear growth with new contrast enhancement was seen on routine MRI. At this point, the lesion was surgically resected. The diagnosis was, surprisingly, glioblastoma multiforme (WHO grade IV, BRAF V-600E mutation). DISCUSSION: Malignant transformation of LGGs in children is a very rare phenomenon. This is to our knowledge the first well-documented case describing malignant transformation of a suspected benign pediatric cerebral mass lesion, which did not undergo radiation, in a patient without a cancer predisposition syndrome (e.g., neurofibromatosis), with the transformation occurring after such a long follow-up period. The management of these lesions is still controversial. Unfortunately, radiological risk factors for malignant transformation of such lesions in the pediatric age group are lacking. CONCLUSION: Conservative treatment of incidental cerebral mass lesions in children seems a valid option. These lesions should probably be followed indefinitely, while carefully watching for changes in imaging characteristics.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/terapia , Tratamiento Conservador/tendencias , Glioblastoma/diagnóstico por imagen , Glioblastoma/terapia , Hallazgos Incidentales , Niño , Tratamiento Conservador/métodos , Progresión de la Enfermedad , Humanos , Masculino , Resultado del Tratamiento
16.
JAMA ; 318(23): 2306-2316, 2017 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-29260225

RESUMEN

Importance: Tumor-treating fields (TTFields) is an antimitotic treatment modality that interferes with glioblastoma cell division and organelle assembly by delivering low-intensity alternating electric fields to the tumor. Objective: To investigate whether TTFields improves progression-free and overall survival of patients with glioblastoma, a fatal disease that commonly recurs at the initial tumor site or in the central nervous system. Design, Setting, and Participants: In this randomized, open-label trial, 695 patients with glioblastoma whose tumor was resected or biopsied and had completed concomitant radiochemotherapy (median time from diagnosis to randomization, 3.8 months) were enrolled at 83 centers (July 2009-2014) and followed up through December 2016. A preliminary report from this trial was published in 2015; this report describes the final analysis. Interventions: Patients were randomized 2:1 to TTFields plus maintenance temozolomide chemotherapy (n = 466) or temozolomide alone (n = 229). The TTFields, consisting of low-intensity, 200 kHz frequency, alternating electric fields, was delivered (≥ 18 hours/d) via 4 transducer arrays on the shaved scalp and connected to a portable device. Temozolomide was administered to both groups (150-200 mg/m2) for 5 days per 28-day cycle (6-12 cycles). Main Outcomes and Measures: Progression-free survival (tested at α = .046). The secondary end point was overall survival (tested hierarchically at α = .048). Analyses were performed for the intent-to-treat population. Adverse events were compared by group. Results: Of the 695 randomized patients (median age, 56 years; IQR, 48-63; 473 men [68%]), 637 (92%) completed the trial. Median progression-free survival from randomization was 6.7 months in the TTFields-temozolomide group and 4.0 months in the temozolomide-alone group (HR, 0.63; 95% CI, 0.52-0.76; P < .001). Median overall survival was 20.9 months in the TTFields-temozolomide group vs 16.0 months in the temozolomide-alone group (HR, 0.63; 95% CI, 0.53-0.76; P < .001). Systemic adverse event frequency was 48% in the TTFields-temozolomide group and 44% in the temozolomide-alone group. Mild to moderate skin toxicity underneath the transducer arrays occurred in 52% of patients who received TTFields-temozolomide vs no patients who received temozolomide alone. Conclusions and Relevance: In the final analysis of this randomized clinical trial of patients with glioblastoma who had received standard radiochemotherapy, the addition of TTFields to maintenance temozolomide chemotherapy vs maintenance temozolomide alone, resulted in statistically significant improvement in progression-free survival and overall survival. These results are consistent with the previous interim analysis. Trial Registration: clinicaltrials.gov Identifier: NCT00916409.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Dacarbazina/análogos & derivados , Terapia por Estimulación Eléctrica , Glioblastoma/tratamiento farmacológico , Adulto , Anciano , Antineoplásicos Alquilantes/efectos adversos , Quimioradioterapia , Dacarbazina/efectos adversos , Dacarbazina/uso terapéutico , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Glioblastoma/radioterapia , Glioblastoma/cirugía , Humanos , Quimioterapia de Mantención , Masculino , Persona de Mediana Edad , Mitosis , Análisis de Supervivencia , Temozolomida
17.
BMC Neurosci ; 17(1): 30, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27251756

RESUMEN

BACKGROUND: Conducting research on the molecular biology, immunology, and physiology of brain tumors (BTs) and primary brain tissues requires the use of viably dissociated single cells. Inadequate methods for tissue dissociation generate considerable loss in the quantity of single cells produced and in the produced cells' viability. Improper dissociation may also demote the quality of data attained in functional and molecular assays due to the presence of large quantities cellular debris containing immune-activatory danger associated molecular patterns, and due to the increased quantities of degraded proteins and RNA. RESULTS: Over 40 resected BTs and non-tumorous brain tissue samples were dissociated into single cells by mechanical dissociation or by mechanical and enzymatic dissociation. The quality of dissociation was compared for all frequently used dissociation enzymes (collagenase, DNase, hyaluronidase, papain, dispase) and for neutral protease (NP) from Clostridium histolyticum. Single-cell-dissociated cell mixtures were evaluated for cellular viability and for the cell-mixture dissociation quality. Dissociation quality was graded by the quantity of subcellular debris, non-dissociated cell clumps, and DNA released from dead cells. Of all enzymes or enzyme combinations examined, NP (an enzyme previously not evaluated on brain tissues) produced dissociated cell mixtures with the highest mean cellular viability: 93 % in gliomas, 85 % in brain metastases, and 89 % in non-tumorous brain tissue. NP also produced cell mixtures with significantly less cellular debris than other enzymes tested. Dissociation using NP was non-aggressive over time-no changes in cell viability or dissociation quality were found when comparing 2-h dissociation at 37 °C to overnight dissociation at ambient temperature. CONCLUSIONS: The use of NP allows for the most effective dissociation of viable single cells from human BTs or brain tissue. Its non-aggressive dissociative capacity may enable ambient-temperature shipping of tumor pieces in multi-center clinical trials, meanwhile being dissociated. As clinical grade NP is commercially available it can be easily integrated into cell-therapy clinical trials in neuro-oncology. The high quality viable cells produced may enable investigators to conduct more consistent research by avoiding the experimental artifacts associated with the presence dead cells or cellular debris.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/citología , Separación Celular/métodos , Proteínas Bacterianas , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Supervivencia Celular , Clostridium histolyticum , Enzimas , Congelación , Glioma/metabolismo , Glioma/patología , Humanos
18.
J Neurooncol ; 126(3): 551-7, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26603164

RESUMEN

We present a retrospective review of 55 Stereotactic Radiosurgery (SRS) procedures performed in 47 consecutive patients with high-grade glioma (HGG). Thirty-three (70.2%) patients were diagnosed with glioblastoma and 14 (29.8%) with grade III glioma. The indications for SRS were small (up to 30 mm in diameter) locally progressing lesions in 32/47 (68%) or new distant lesions in 15/47 (32%) patients. The median target volume was 2.2 cc (0.2-9.5 cc) and the median prescription dose was 18 Gy (14-24 Gy). Three patients (5.5% incidence in 55 treatments) developed radiation necrosis. In eight cases (17%) patients received a second salvage SRS treatment to nine new lesions detected during follow-up. In 22/55 SRS treatments (40.0%) patients received concurrent chemo- or biological therapy, including temozolamide (TMZ) (15 patients), bevacizumab (BVZ) (6 patients) and carboplatin in one patient. Median time to progression after SRS was 5.0 months (1.0-96.4). Median survival time after SRS was 15.9 months (2.3-109.3) overall median survival (since diagnosis) was 37.4 months (9.6-193.6 months). Long-lasting responses (>12 months) after SRS were observed in 25/46 (54.3%) patients. We compared a matched (histology, age, KPS) cohort of patients with recurrent HGG treated with BVZ alone with the current study group. Median survival was significantly longer for SRS treated patients compared to the BVZ only cohort (12.6 vs. 7.3 months, p = 0.0102). SRS may be considered an effective salvage procedure for selected patients with small volume, recurrent high-grade gliomas. Long-term radiological control was observed in more than 50% of the patients.


Asunto(s)
Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Glioma/patología , Glioma/cirugía , Radiocirugia , Terapia Recuperativa , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Carga Tumoral , Adulto Joven
19.
J Neurooncol ; 130(3): 413-422, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27573219

RESUMEN

The 54 microRNAs (miRNAs) within the DLK-DIO3 genomic region on chromosome 14q32.31 (cluster-14-miRNAs) are organized into sub-clusters 14A and 14B. These miRNAs are downregulated in glioblastomas and might have a tumor suppressive role. Any association between the expression levels of cluster-14-miRNAs with overall survival (OS) is undetermined. We randomly selected miR-433, belonging to sub-cluster 14A and miR-323a-3p and miR-369-3p, belonging to sub-cluster 14B, and assessed their role in glioblastomas in vitro and in vivo. We also determined the expression level of cluster-14-miRNAs in 27 patients with newly diagnosed glioblastoma, and analyzed the association between their level of expression and OS. Overexpression of miR-323a-3p and miR-369-3p, but not miR-433, in glioblastoma cells inhibited their proliferation and migration in vitro. Mice implanted with glioblastoma cells overexpressing miR323a-3p and miR369-3p, but not miR433, exhibited prolonged survival compared to controls (P = .003). Bioinformatics analysis identified 13 putative target genes of cluster-14-miRNAs, and real-time RT-PCR validated these findings. Pathway analysis of the putative target genes identified neuregulin as the most enriched pathway. The expression level of cluster-14-miRNAs correlated with patients' OS. The median OS was 8.5 months for patients with low expression levels and 52.7 months for patients with high expression levels (HR 0.34; 95 % CI 0.12-0.59, P = .003). The expression level of cluster-14-miRNAs correlates directly with OS, suggesting a role for this cluster in promoting aggressive behavior of glioblastoma, possibly through ErBb/neuregulin signaling.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Cromosomas Humanos Par 14 , Glioblastoma/genética , Glioblastoma/mortalidad , MicroARNs/genética , Adulto , Anciano , Animales , Encéfalo/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Estudios de Cohortes , Biología Computacional , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Glioblastoma/patología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Invasividad Neoplásica , Recurrencia Local de Neoplasia , Análisis de Supervivencia , Transfección
20.
J Neurooncol ; 130(1): 211-219, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27531351

RESUMEN

Genomic research of high grade glioma (HGG) has revealed complex biology with potential for therapeutic impact. However, the utilization of this information and impact upon patient outcome has yet to be assessed. We performed capture-based next generation sequencing (NGS) genomic analysis assay of 236/315 cancer-associated genes, with average depth of over 1000 fold, to guide treatment in HGG patients. We reviewed clinical utility and response rates in correlation to NGS results. Forty-three patients were profiled: 34 glioblastomas, 8 anaplastic astrocytomas, and one patient with anaplastic oligodendroglioma. Twenty-five patients were profiled with the 315 gene panel. The median number of identified genomic alterations (GAs) per patient was 4.5 (range 1-23). In 41 patients (95 %) at least one therapeutically-actionable GA was detected, most commonly in EGFR [17 (40 %)]. Genotype-directed treatments were prescribed in 13 patients, representing a 30 % treatment decision impact. Treatment with targeted agents included everolimus as a single agent and in combination with erlotinib; erlotinib; afatinib; palbociclib; trametinib and BGJ398. Treatments targeted various genomic findings including EGFR alterations, mTOR activation, cell cycle targets and FGFR1 mutations. None of the patients showed response to respective biologic treatments. In this group of patients with HGG, NGS revealed a high frequency of GAs that lead to targeted treatment in 30 % of the patients. The lack of response suggests that further study of mechanisms of resistance in HGG is warranted before routine use of biologically-targeted agents based on NGS results.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/genética , Genómica/métodos , Glioma/genética , Resultado del Tratamiento , Adolescente , Adulto , Anciano , Neoplasias Encefálicas/tratamiento farmacológico , Estudios de Cohortes , Receptores ErbB/genética , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib/uso terapéutico , Everolimus/uso terapéutico , Femenino , Glioma/tratamiento farmacológico , Humanos , Masculino , Persona de Mediana Edad , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA