Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 386(26): 2471-2481, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35767439

RESUMEN

BACKGROUND: Pediatric patients with diffuse intrinsic pontine glioma (DIPG) have a poor prognosis, with a median survival of less than 1 year. Oncolytic viral therapy has been evaluated in patients with pediatric gliomas elsewhere in the brain, but data regarding oncolytic viral therapy in patients with DIPG are lacking. METHODS: We conducted a single-center, dose-escalation study of DNX-2401, an oncolytic adenovirus that selectively replicates in tumor cells, in patients with newly diagnosed DIPG. The patients received a single virus infusion through a catheter placed in the cerebellar peduncle, followed by radiotherapy. The primary objective was to assess the safety and adverse-event profile of DNX-2401. The secondary objectives were to evaluate the effect of DNX-2401 on overall survival and quality of life, to determine the percentage of patients who have an objective response, and to collect tumor-biopsy and peripheral-blood samples for correlative studies of the molecular features of DIPG and antitumor immune responses. RESULTS: A total of 12 patients, 3 to 18 years of age, with newly diagnosed DIPG received 1×1010 (the first 4 patients) or 5×1010 (the subsequent 8 patients) viral particles of DNX-2401, and 11 received subsequent radiotherapy. Adverse events among the patients included headache, nausea, vomiting, and fatigue. Hemiparesis and tetraparesis developed in 1 patient each. Over a median follow-up of 17.8 months (range, 5.9 to 33.5), a reduction in tumor size, as assessed on magnetic resonance imaging, was reported in 9 patients, a partial response in 3 patients, and stable disease in 8 patients. The median survival was 17.8 months. Two patients were alive at the time of preparation of the current report, 1 of whom was free of tumor progression at 38 months. Examination of a tumor sample obtained during autopsy from 1 patient and peripheral-blood studies revealed alteration of the tumor microenvironment and T-cell repertoire. CONCLUSIONS: Intratumoral infusion of oncolytic virus DNX-2401 followed by radiotherapy in pediatric patients with DIPG resulted in changes in T-cell activity and a reduction in or stabilization of tumor size in some patients but was associated with adverse events. (Funded by the European Research Council under the European Union's Horizon 2020 Research and Innovation Program and others; EudraCT number, 2016-001577-33; ClinicalTrials.gov number, NCT03178032.).


Asunto(s)
Neoplasias del Tronco Encefálico , Glioma Pontino Intrínseco Difuso , Viroterapia Oncolítica , Virus Oncolíticos , Adenoviridae , Adolescente , Astrocitoma/radioterapia , Astrocitoma/terapia , Neoplasias del Tronco Encefálico/mortalidad , Neoplasias del Tronco Encefálico/patología , Neoplasias del Tronco Encefálico/radioterapia , Neoplasias del Tronco Encefálico/terapia , Niño , Preescolar , Glioma Pontino Intrínseco Difuso/mortalidad , Glioma Pontino Intrínseco Difuso/radioterapia , Glioma Pontino Intrínseco Difuso/terapia , Glioma/radioterapia , Glioma/terapia , Humanos , Infusiones Intralesiones , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Calidad de Vida , Microambiente Tumoral
2.
Childs Nerv Syst ; 36(4): 705-711, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32020269

RESUMEN

INTRODUCTION: The prognosis of diffuse intrinsic pontine glioma (DIPG) is poor. The role of biopsy in DIPG remains controversial since the diagnosis may be established with imaging alone. Recent advances in understanding molecular biology and targeting of brain tumors have created a renewed interest in biopsy for DIPG. The Neurosurgery Working Group (NWG) of the SIOP-Europe Brain Tumor Group (BTG) undertook a survey among international pediatric neurosurgeons to define their current perceptions and practice regarding DIPG biopsy. METHODS: The NWG developed a 20-question survey which was emailed to neurosurgeons in the International Society for Pediatric Neurosurgery (ISPN). The questionnaire included questions on diagnosis, indications, and techniques for biopsy, clinical trials, and healthcare infrastructure. RESULTS: The survey was sent to 202 neurosurgeons and 73 (36%) responded. Consensus of > 75% agreement was reached for 12/20 questions, which included (1) radiological diagnosis of DIPG is sufficient outside a trial, (2) clinical trial-based DIPG biopsy is justified if molecular targets are investigated and may be used for treatment, and (3) morbidity/mortality data must be collected to define the risk:benefit ratio. The remaining 8/20 questions proved controversial and failed to reach consensus. CONCLUSIONS: Routine DIPG biopsy continues to be debated. Most neurosurgeons agreed that DIPG biopsy within a clinical trial should be supported, with the aims of defining the procedure risks, improving understanding of tumor biology, and evaluating new treatment targets. Careful family counseling and consent remain important.


Asunto(s)
Neoplasias del Tronco Encefálico , Glioma , Biopsia , Neoplasias del Tronco Encefálico/diagnóstico por imagen , Neoplasias del Tronco Encefálico/cirugía , Niño , Europa (Continente) , Glioma/diagnóstico por imagen , Glioma/cirugía , Humanos , Neurocirujanos , Encuestas y Cuestionarios
3.
Childs Nerv Syst ; 34(4): 617-626, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29460065

RESUMEN

BACKGROUND: The intraoperative magnetic resonance scanner (ioMR) was introduced in our unit in 2009, and has been used routinely since then. OBJECTIVE: This study aims to describe indications, radiological features, and clinical outcomes of the patients operated on with ioMRI and analyze our experience. METHODS: A retrospective analysis of a prospective surgical database has been performed, including surgical procedure, intent, radiological reports, need for second-look surgery, and complications, supplemented by further review of the clinical notes and the scans. RESULTS: From 2009 to 2015, 255 surgical procedures with ioMR were performed: 175 were craniotomies for tumor excision, 65 were epilepsy related, and 15 were biopsies or cyst drainages. The mean age was 9.4 years. One ioMR was performed in 79.5% patients; the mean duration of the MR was 41 min. In 172 cases (67.4%), no actions followed the ioMR. When the aim of the surgery was debulking of the tumor, the percentage of patients in which the ioMR was followed by resection was higher than when complete resection was the aim (56 vs 27.5%). The complication rate was not increased when compared with our previous results (infection 1%, neurological deficits 12%). CONCLUSION: This is the largest published series of ioMRI-aided pediatric neurosurgery to date. We have demonstrated that it can be used safely and routinely in pediatric neurosurgical procedures at any age, assisting the surgeon in achieving the best extent of resection and aiding in intra-operative decision-making for tumor- and non-tumor-related intracranial pathology.


Asunto(s)
Neoplasias Encefálicas/cirugía , Epilepsia/cirugía , Imagen por Resonancia Magnética/efectos adversos , Monitoreo Intraoperatorio/efectos adversos , Procedimientos Neuroquirúrgicos/efectos adversos , Pediatría , Adolescente , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Estudios Longitudinales , Pediatría/métodos , Pediatría/normas , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven
4.
J Transl Med ; 15(1): 104, 2017 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-28499389

RESUMEN

BACKGROUND: Prognosis of patients with glioblastoma multiforme (GBM) remains dismal, with median overall survival (OS) of about 15 months. It is therefore crucial to search alternative strategies that improve these results obtained with conventional treatments. In this context, immunotherapy seems to be a promising therapeutic option. We hypothesized that the addition of tumor lysate-pulsed autologous dendritic cells (DCs) vaccination to maximal safe resection followed by radiotherapy and concomitant and adjuvant temozolomide could improve patients' survival. METHODS: We conducted a phase-II clinical trial of autologous DCs vaccination in patients with newly diagnosed patients GBM who were candidates to complete or near complete resection. Candidates were finally included if residual tumor volume was lower than 1 cc on postoperative radiological examination. Autologous DCs were generated from peripheral blood monocytes and pulsed with autologous whole tumor lysate. The vaccination calendar started before radiotherapy and was continued during adjuvant chemotherapy. Progression free survival (PFS) and OS were analyzed with the Kaplan-Meier method. Immune response were assessed in blood samples obtained before each vaccines. RESULTS: Thirty-two consecutive patients were screened, one of which was a screening failure due to insufficient resection. Median age was 61 years (range 42-70). Karnofsky performance score (KPS) was 90-100 in 29%, 80 in 35.5% and 60-70 in 35.5% of cases. MGMT (O6-methylguanine-DNA-methyltransferase) promoter was methylated in 45.2% of patients. No severe adverse effects related to immunotherapy were registered. Median PFS was 12.7 months (CI 95% 7-16) and median OS was 23.4 months (95% CI 16-33.1). Increase in post-vaccination tumor specific immune response after vaccines (proliferation or cytokine production) was detected in 11/27 evaluated patients. No correlation between immune response and survival was found. CONCLUSIONS: Our results suggest that the addition of tumor lysate-pulsed autologous DCs vaccination to tumor resection and combined radio-chemotherapy is feasible and safe. A multicenter randomized clinical trial is warranted to evaluate the potential survival benefit of this therapeutic approach. Trial registration This phase-II trial was registered as EudraCT: 2009-009879-35 and ClinicalTrials.gov Identifier: NCT01006044 retrospectively registered.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Quimioradioterapia , Células Dendríticas/inmunología , Glioblastoma/inmunología , Glioblastoma/terapia , Vacunación , Adulto , Anciano , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/cirugía , Quimioradioterapia/efectos adversos , Terapia Combinada , Citocinas/sangre , Supervivencia sin Enfermedad , Estudios de Factibilidad , Femenino , Fluorescencia , Glioblastoma/sangre , Glioblastoma/cirugía , Humanos , Inflamación/patología , Masculino , Persona de Mediana Edad , Análisis de Supervivencia , Trasplante Autólogo , Vacunación/efectos adversos
5.
J Neurooncol ; 116(1): 169-75, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24135848

RESUMEN

Our purpose was to analyze the pattern of failure in glioblastoma (GBM) patients at first recurrence after radiotherapy and temozolomide and its relationship with different factors. From 77 consecutive GBM patients treated at our institution with fluorescence guided surgery and standard radiochemotherapy, 58 first recurrences were identified and included in a retrospective review. Clinical data including age, Karnofsky performance score, preoperative tumor volume and location, extend of resection, MGMT promoter methylation status, time to progression (PFS), overall survival (OS) and adjuvant therapies were reviewed for every patient. Recurrent tumor location respect the original lesion was the end point of the study. The recurrence pattern was local only in 65.5% of patients and non-local in 34.5%. The univariate and multivariate analysis showed that greater preoperative tumor volume in T1 gadolinium enhanced sequences, was the only variable with statistical signification (p < 0.001) for increased rate of non-local recurrences, although patients with MGMT methylation and complete resection of enhancing tumor presented non-local recurrences more frequently. PFS was longer in patients with non-local recurrences (13.8 vs. 6.4 months; p = 0.019, log-rank). However, OS was not significantly different in both groups (24.0 non-local vs. 19.3 local; p = 0.9). Rate of non-local recurrences in our series of patients treated with fluorescence guided surgery and standard radiochemotherapy was higher than previously published in GBM, especially in patients with longer PFS. Greater preoperative enhancing tumor volume was associated with increased rate of non-local recurrences.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Recurrencia Local de Neoplasia/etiología , Antineoplásicos Alquilantes/efectos adversos , Neoplasias Encefálicas/genética , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Dacarbazina/efectos adversos , Dacarbazina/análogos & derivados , Progresión de la Enfermedad , Femenino , Glioblastoma/genética , Humanos , Estado de Ejecución de Karnofsky , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Análisis Multivariante , Recurrencia Local de Neoplasia/genética , Regiones Promotoras Genéticas/genética , Radioterapia/efectos adversos , Temozolomida , Proteínas Supresoras de Tumor/genética
6.
Neurocirugia (Astur : Engl Ed) ; 33(6): 261-268, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34625382

RESUMEN

BACKGROUND AND AIMS: Intraoperative MRI (ioMRI) consists of performing a MRI during brain or spinal surgery. Although it is a safe and useful technique, it is available in a few hospitals. This means some aspects are not perfectly defined or standardized, forcing each center to develop its own solutions. Our goal is to describe the technique, evaluate the changes made to optimize its use and thus be able to facilitate the intraoperative resonance implementation in other neurosurgery departments. METHODS: A prospective analysis of patients consecutively operated using high-field ioMRI guidance was carried out, describing the type of tumor, clinical data, time and sequences of ioMR, use of intraoperative neurophysiology, preoperative tumor volume, after ioMR, and postoperative, as well as complications. RESULTS: ioMR was performed in 38 patients selected from among 425 brain tumors (9%) operated on in this interval. The tumor types were: 11 glioblastomas, 8 anaplastic astrocytomas, 5 diffuse astrocytomas, 4 meningiomas, 3 oligodendrogliomas, 2 metastases, 2 epidermoid cysts, 1 astroblastoma, 1 arachnoid cyst and 1 pituitary adenoma. The mean age was 45 years. The mean preoperative tumor volume was 45.22cc, after the ioMR 5.08cc and postoperative 1.28cc. Resection was extended after ioMR in 76%. Gross total resection was achieved in 15 patients and residual tumor of less than 1cc was observed in 8. An intentional tumor tissue was left in an eloquent brain region (mean volume 7cc) in 13 patients. Bleeding and ischemia complications were detected early on ioMR in 5%. MRI length was 47 min on average. CONCLUSIONS: Intraoperative MRI was a useful and safe technique, and no associated complications were registered.


Asunto(s)
Astrocitoma , Neoplasias Encefálicas , Glioblastoma , Humanos , Persona de Mediana Edad , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/cirugía , Neoplasias Encefálicas/patología , Imagen por Resonancia Magnética/métodos , Procedimientos Neuroquirúrgicos/métodos , Glioblastoma/cirugía , Atención a la Salud
7.
Eur J Trauma Emerg Surg ; 48(3): 2189-2198, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34401937

RESUMEN

BACKGROUND: COVID-19 has overloaded health care systems, testing the capacity and response in every European region. Concerns were raised regarding the impact of resources' reorganization on certain emergency pathology management. The aim of the present study was to assess the impact of the outbreak (in terms of reduction of neurosurgical emergencies) during lockdown in different regions of Spain. METHODS: We analyzed the impact of the outbreak in four different affected regions by descriptive statistics and univariate comparison with same period of two previous years. These regions differed in their incidence level (high/low) and in the time of excess mortality with respect to lockdown declaration. That allowed us to analyze their influence on the characteristics of neurosurgical emergencies registered for every region. RESULTS: 1185 patients from 18 neurosurgical centers were included. Neurosurgical emergencies that underwent surgery dropped 24.41% and 28.15% in 2020 when compared with 2019 and 2018, respectively. A higher reduction was reported for the most affected regions by COVID-19. Non-traumatic spine experienced the most significant decrease in number of cases. Life-threatening conditions did not suffer a reduction in any health care region. CONCLUSIONS: COVID-19 affected dramatically the neurosurgical emergency management. The most significant reduction in neurosurgical emergencies occurred on those regions that were hit unexpectedly by the pandemic, as resources were focused on fighting the virus. As a consequence, life-threating and non-life-threatening conditions' mortality raised. Results in regions who had time to prepare for the hit were congruent with an organized and sensible neurosurgical decision-making.


Asunto(s)
COVID-19 , COVID-19/epidemiología , Control de Enfermedades Transmisibles , Atención a la Salud , Urgencias Médicas , Humanos , Procedimientos Neuroquirúrgicos , España/epidemiología
8.
Neuropathology ; 31(6): 575-82, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21355891

RESUMEN

Thirty consecutive surgical patients with glioblastoma, were operated upon using fluorescence induced by 5-aminolevulinic acid as guidance. The fluorescent quality of the tissue was used to take biopsies from the tumor center, from the invasive area around it and from adjacent normal-looking tissue. These samples were analyzed with HE, Ki-67 and nestin. Nestin expression in tissue surrounding glioblastoma cases was compared to tissue surrounding vascular lesions, metastasis and hippocampal sclerosis. The rate of gross total resection assessed by volumetric MRI was 83%. Using HE examination as the gold standard, fluorescence identified solid tumor with 100% positive predictive value, invasive areas with 97%, and normal tissue with 67% negative predictive value. Ki67 stained some cells in 69% of the non-fluorescent samples around the tumor. There was always strong nestin expression around the tumor but it was similar to control cases in non-glioma lesions with subacute expansion. 5-aminolevulinic acid fluorescence guidance is very reliable and can help to study the tumor-brain interface. Nestin expression is strong and constant in the tissue around the tumor, but is mostly an acute glial reaction, not specific of the neoplasm. Nestin staining is not recommended as a tumor stem cell marker.


Asunto(s)
Ácido Aminolevulínico , Neoplasias Encefálicas/patología , Colorantes Fluorescentes , Glioblastoma/patología , Proteínas de Filamentos Intermediarios/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/cirugía , Glioblastoma/metabolismo , Glioblastoma/cirugía , Humanos , Inmunohistoquímica , Proteínas de Filamentos Intermediarios/análisis , Proteínas del Tejido Nervioso/análisis , Nestina
9.
J Neurol Surg A Cent Eur Neurosurg ; 82(1): 53-58, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33260244

RESUMEN

INTRODUCTION: The infiltrative margin of glioblastomas (GBM) contains proliferative tumor cells difficult to estimate radiologically as they are included in the hyperintense signal of T2 sequences and they remain in the cavity margin after tumor resection. The amount of these cells could determine overall survival (OS) of these patients. MATERIAL AND METHODS: From October 2007 to January 2010, patients whose MRI were suggestive of newly diagnosed, resectable high-grade glioma were operated using fluorescence-guided surgery (FGS). Separate samples were selectively taken from nonfluorescent white matter areas just adjacent to the border of the pale fluorescence and staining was made for Ki-67. OS was analyzed with Kaplan-Meier and Cox regression. Multivariate analysis included the following prognosis variables: age, extent of resection (EOR), O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation, and performance status index. RESULTS: Sample included 65 patients, comprising 37 men and 28 women, with a median Karnofsky Performance Score (KPS) of 80 (40-100) and mean age of 60 (34-78) years. Mean preoperative tumor volume was 35.8 mL. EOR was 100% in 52 patients (80%), with the lower EOR being 88%. For Ki-67, 39 patients had <5% and 26 had ≥5%. OS was 26.8 months (95% confidence interval [CI]: 18.9-28.2) for the Ki-67 low group versus 15.8 months (95% CI: 7.7-18.2) for the Ki-67 high group (p = 0.002). CONCLUSION: Proliferative activity in the normal-looking brain around the resection cavity measured with Ki-67 immunostaining is an important independent prognostic factor for GBM cases with complete resection of enhancing tumor. When complete resection is not reached, this factor is not relevant for prognosis.


Asunto(s)
Neoplasias Encefálicas/mortalidad , Glioblastoma/mortalidad , Antígeno Ki-67/análisis , Adulto , Anciano , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Proliferación Celular , Femenino , Glioblastoma/diagnóstico por imagen , Glioblastoma/patología , Humanos , Estado de Ejecución de Karnofsky , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Índice Mitótico , Pronóstico , Estudios Retrospectivos
10.
J Histochem Cytochem ; 57(5): 503-12, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19188486

RESUMEN

Increased protein synthesis is regulated, in part, by two eukaryotic translation initiation factors (eIFs): eIF4E and eIF2alpha. One or both of these factors are often overexpressed in several types of cancer cells; however, no data are available at present regarding eIF4E and eIF2alpha levels in brain tumors. In this study, we analyzed the expression, subcellular localization and phosphorylation states of eIF4E and eIF2alpha in 64 brain tumors (26 meningiomas, 16 oligodendroglial tumors, and 22 astrocytomas) and investigated the correlation with the expression of MIB-1, p53, and cyclin D1 proteins as well. There are significant differences in the phosphorylated eIF4E levels between the tumors studied, being the highest in meningiomas and the lowest in the oligodendroglial tumors. Relative to subcellular localization, eIF4E is frequently found in the nucleus of the oligodendroglial tumors and rarely in the same compartment of the meningiomas, whereas eIF2alpha showed an inverse pattern. Finally, cyclin D1 levels directly correlate with the phosphorylation status of both factors. The different expression, phosphorylation, or/and subcellular distribution of eIF2alpha and eIF4E within the brain types of tumors studied could indicate that different pathways are activated for promoting cell cycle proliferation, for instance, leading to increased cyclin D1 expression.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular , Ciclina D1/metabolismo , Femenino , Humanos , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Fosforilación , Proteína p53 Supresora de Tumor/metabolismo , Adulto Joven
12.
PLoS One ; 14(6): e0217881, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31170245

RESUMEN

BACKGROUND: Hypofractionated radiation therapy is a feasible and safe treatment option in elderly and frail patients with glioblastoma. The aim of this study was to evaluate the effectiveness of hypofractionated radiation therapy with concurrent temozolomide in terms of feasibility and disease control in primary glioblastoma patients with poor prognostic factors other than advanced age, such as post-surgical neurological complications, high tumor burden, unresectable or multifocal lesions, and potential low treatment compliance due to social factors or rapidly progressive disease. MATERIAL AND METHODS: GTV included the surgical cavity plus disease visible in T1WI-MRI, FLAIR-MRI and in the MET-uptake. The CTV was defined as the GTV plus 1.5-2 cm margin; the PTV was the CTV+0.3 cm margin. Forty, fourty-five, and fifty grays in 15 fractions were prescribed to 95% of PTV, CTV, and GTV, respectively. Treatment was delivered using IMRT or the VMAT technique. Simultaneously, 75 mg/m2/day of temozolomide were administered. RESULTS: Between January 2010 and November 2017, we treated a total of 17 patients. The median age at diagnosis was 68-years; median KPS was 50-70%. MGMT-methylation status was negative in 5 patients, and 8 patients were IDH-wildtype. Eight of 18 patients were younger than 65-years. Median tumor volume was 26.95cc; median PTV volume was 322cc. Four lesions were unresectable; 6 patients underwent complete surgical resection. Median residual volume was 1.14cc. Progression-free survival was 60% at 6 months, 33% at 1-year and 13% at 2-years (median OS = 7 months). No acute grade 3-5 toxicities were documented. Symptomatic grade 3 radiation necrosis was observed in one patient. CONCLUSIONS: Patients with poor clinical factors other than advanced age can be selected for hypofractionated radiotherapy. The OS and PFS rates obtained in our series are similar to those in patients treated with standard fractionation, assuring good treatment adherence, low rates of toxicity and probable improved cost-effectiveness.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Hipofraccionamiento de la Dosis de Radiación , Temozolomida/uso terapéutico , Anciano , Neoplasias Encefálicas/diagnóstico por imagen , Análisis Factorial , Femenino , Glioblastoma/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Necrosis , Recurrencia Local de Neoplasia/patología , Pronóstico , Supervivencia sin Progresión , Estudios Prospectivos , Temozolomida/efectos adversos
14.
Acta Neuropathol Commun ; 7(1): 64, 2019 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-31036068

RESUMEN

Pediatric high grade gliomas (pHGG), including diffuse intrinsic pontine gliomas (DIPGs), are aggressive tumors with a dismal outcome. Radiotherapy (RT) is part of the standard of care of these tumors; however, radiotherapy only leads to a transient clinical improvement. Delta-24-RGD is a genetically engineered tumor-selective adenovirus that has shown safety and clinical efficacy in adults with recurrent gliomas. In this work, we evaluated the feasibility, safety and therapeutic efficacy of Delta-24-RGD in combination with radiotherapy in pHGGs and DIPGs models. Our results showed that the combination of Delta-24-RGD with radiotherapy was feasible and resulted in a synergistic anti-glioma effect in vitro and in vivo in pHGG and DIPG models. Interestingly, Delta-24-RGD treatment led to the downregulation of relevant DNA damage repair proteins, further sensitizing tumors cells to the effect of radiotherapy. Additionally, Delta-24-RGD/radiotherapy treatment significantly increased the trafficking of immune cells (CD3, CD4+ and CD8+) to the tumor niche compared with single treatments. In summary, administration of the Delta-24-RGD/radiotherapy combination to pHGG and DIPG models is safe and significantly increases the overall survival of mice bearing these tumors. Our data offer a rationale for the combination Delta-24-RGD/radiotherapy as a therapeutic option for children with these tumors. SIGNIFICANCE: Delta-24-RGD/radiotherapy administration is safe and significantly increases the survival of treated mice. These positive data underscore the urge to translate this approach to the clinical treatment of children with pHGG and DIPGs.


Asunto(s)
Neoplasias Encefálicas/terapia , Neoplasias del Tronco Encefálico/terapia , Glioma Pontino Intrínseco Difuso/terapia , Viroterapia Oncolítica/métodos , Adenoviridae/fisiología , Animales , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/radioterapia , Neoplasias del Tronco Encefálico/complicaciones , Neoplasias del Tronco Encefálico/radioterapia , Línea Celular Tumoral , Terapia Combinada/métodos , Daño del ADN , Glioma Pontino Intrínseco Difuso/complicaciones , Glioma Pontino Intrínseco Difuso/radioterapia , Vectores Genéticos , Humanos , Ratones
15.
Nat Med ; 25(3): 470-476, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30742120

RESUMEN

Glioblastoma is the most common primary central nervous system malignancy and has a poor prognosis. Standard first-line treatment, which includes surgery followed by adjuvant radio-chemotherapy, produces only modest benefits to survival1,2. Here, to explore the feasibility, safety and immunobiological effects of PD-1 blockade in patients undergoing surgery for glioblastoma, we conducted a single-arm phase II clinical trial (NCT02550249) in which we tested a presurgical dose of nivolumab followed by postsurgical nivolumab until disease progression or unacceptable toxicity in 30 patients (27 salvage surgeries for recurrent cases and 3 cases of primary surgery for newly diagnosed patients). Availability of tumor tissue pre- and post-nivolumab dosing and from additional patients who did not receive nivolumab allowed the evaluation of changes in the tumor immune microenvironment using multiple molecular and cellular analyses. Neoadjuvant nivolumab resulted in enhanced expression of chemokine transcripts, higher immune cell infiltration and augmented TCR clonal diversity among tumor-infiltrating T lymphocytes, supporting a local immunomodulatory effect of treatment. Although no obvious clinical benefit was substantiated following salvage surgery, two of the three patients treated with nivolumab before and after primary surgery remain alive 33 and 28 months later.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Linfocitos Infiltrantes de Tumor/inmunología , Terapia Neoadyuvante , Procedimientos Neuroquirúrgicos , Nivolumab/uso terapéutico , Microambiente Tumoral/inmunología , Adulto , Anciano , Neoplasias Encefálicas/inmunología , Quimiocinas/genética , Quimiocinas/inmunología , Femenino , Glioblastoma/inmunología , Humanos , Masculino , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/inmunología , Transcriptoma
16.
Presse Med ; 47(11-12 Pt 2): e199-e212, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30385181

RESUMEN

Diffuse gliomas constitute a diverse group of malignant brain tumors with varying aggressive course and heterogeneous survival. Although the mainstay of treatment of these distinct tumors is still based on the combination of surgery and classical therapeutic weapons such as radiotherapy and chemotherapy, important advances have been achieved over the past decades leading to meaningful improvements in survival times. In addition, recent progress in molecular profiling has allowed the identification of patients with better prognosis and more likely to respond to specific antitumor treatment. This is particularly true for grade II and III 1p/19q-codeleted gliomas, a subset of tumors in which data maturation after long-term follow-up have proved extremely important for accurate assessment of efficacy. This article aims at providing a review of the current specific antitumor treatment of diffuse glioma, particularly grade II and III glioma and glioblastoma, with special emphasis on the most relevant clinical trials conducted in these populations of patients.


Asunto(s)
Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Glioma/patología , Glioma/terapia , Antineoplásicos/clasificación , Antineoplásicos/uso terapéutico , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Clasificación del Tumor , Procedimientos Neuroquirúrgicos/métodos , Radioterapia/métodos
17.
Neurosurgery ; 83(5): 1050-1056, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29088386

RESUMEN

BACKGROUND: There are no effective treatments for diffuse intrinsic pontine gliomas (DIPGs); these tumors cannot be surgical resected, and diagnosis is based on magnetic resonance imaging. As a result, tumor tissues for molecular studies and pathologic diagnosis are infrequent. New clinical trials are investigating novel medications and therapeutic techniques in an effort to improve treatment of patients with DIPGs. OBJECTIVE: To determine the safety, tolerability, and toxicity of an oncolytic adenovirus, DNX-2401, injected into the cerebellar peduncle in pediatric subjects with DIPG and to collect tumor samples of this type of tumor. METHODS: Phase I, single-center, uncontrolled trial. A tumor biopsy will be performed through the cerebellar peduncle, and DNX-2401 will be injected immediately after the biopsy. Standard therapy consisting of radiotherapy and chemotherapy will follow in 2 to 6 wk. EXPECTED OUTCOMES: Improvement of overall survival and quality of life in patients with DIPG and collection of tumor specimens to study the molecular profiling of these tumors. DISCUSSION: The aims of this trial are to contribute to the sample collection of DIPG and to offer treatment during the tumor tissue biopsy using the virus. If this virus works as expected, it could kill the tumor cells with no damage to healthy tissue, functioning as a targeted therapy. It is important to note that edema has not been observed with this virus in all trials performed to date. The information obtained through this and other similar studies may be useful for developing or improving new therapies in the battle against DIPG.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias del Tronco Encefálico/terapia , Glioma/terapia , Viroterapia Oncolítica/métodos , Proyectos de Investigación , Niño , Humanos , Imagen por Resonancia Magnética , Masculino , Viroterapia Oncolítica/efectos adversos
18.
Front Oncol ; 8: 61, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29594041

RESUMEN

Diffuse intrinsic pontine gliomas (DIPGs) are aggressive glial brain tumors that primarily affect children, for which there is no curative treatment. Median overall survival is only one year. Currently, the scientific focus is on expanding the knowledge base of the molecular biology of DIPG, and identifying effective therapies. Oncolytic adenovirus DNX-2401 is a replication-competent, genetically modified virus capable of infecting and killing glioma cells, and stimulating an anti-tumor immune response. Clinical trials evaluating intratumoral DNX-2401 in adults with recurrent glioblastoma have demonstrated that the virus has a favorable safety profile and can prolong survival. Subsequently, these results have encouraged the transition of this biologically active therapy from adults into the pediatric population. To this aim, we have designed a clinical Phase I trial for newly diagnosed pediatric DIPG to investigate the feasibility, safety, and preliminary efficacy of delivering DNX-2401 into tumors within the pons following biopsy. This case report presents a pediatric patient enrolled in this ongoing Phase I trial for children and adolescents with newly diagnosed DIPG. The case involves an 8-year-old female patient with radiologically diagnosed DIPG who underwent stereotactic tumor biopsy immediately followed by intratumoral DNX-2401 in the same biopsy track. Because there were no safety concerns or new neurological deficits, the patient was discharged 3 days after the procedures. To our knowledge, this is the first report of intratumoral DNX-2401 for a patient with DIPG in a clinical trial. We plan to demonstrate that intratumoral delivery of an oncolytic virus following tumor biopsy for pediatric patients with DIPG is a novel and feasible approach and that DNX-2401 represents an innovative treatment for the disease.

19.
J Clin Oncol ; 36(14): 1419-1427, 2018 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-29432077

RESUMEN

Purpose DNX-2401 (Delta-24-RGD; tasadenoturev) is a tumor-selective, replication-competent oncolytic adenovirus. Preclinical studies demonstrated antiglioma efficacy, but the effects and mechanisms of action have not been evaluated in patients. Methods A phase I, dose-escalation, biologic-end-point clinical trial of DNX-2401 was conducted in 37 patients with recurrent malignant glioma. Patients received a single intratumoral injection of DNX-2401 into biopsy-confirmed recurrent tumor to evaluate safety and response across eight dose levels (group A). To investigate the mechanism of action, a second group of patients (group B) underwent intratumoral injection through a permanently implanted catheter, followed 14 days later by en bloc resection to acquire post-treatment specimens. Results In group A (n = 25), 20% of patients survived > 3 years from treatment, and three patients had a ≥ 95% reduction in the enhancing tumor (12%), with all three of these dramatic responses resulting in > 3 years of progression-free survival from the time of treatment. Analyses of post-treatment surgical specimens (group B, n = 12) showed that DNX-2401 replicates and spreads within the tumor, documenting direct virus-induced oncolysis in patients. In addition to radiographic signs of inflammation, histopathologic examination of immune markers in post-treatment specimens showed tumor infiltration by CD8+ and T-bet+ cells, and transmembrane immunoglobulin mucin-3 downregulation after treatment. Analyses of patient-derived cell lines for damage-associated molecular patterns revealed induction of immunogenic cell death in tumor cells after DNX-2401 administration. Conclusion Treatment with DNX-2401 resulted in dramatic responses with long-term survival in recurrent high-grade gliomas that are probably due to direct oncolytic effects of the virus followed by elicitation of an immune-mediated antiglioma response.


Asunto(s)
Adenoviridae , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Viroterapia Oncolítica/métodos , Virus Oncolíticos , Adulto , Biopsia , Femenino , Humanos , Inyecciones Intralesiones , Masculino , Persona de Mediana Edad , Tasa de Supervivencia , Resultado del Tratamiento
20.
Neuro Oncol ; 20(7): 930-941, 2018 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-29373718

RESUMEN

Background: Glioblastoma, the most aggressive primary brain tumor, is genetically heterogeneous. Alternative splicing (AS) plays a key role in numerous pathologies, including cancer. The objectives of our study were to determine whether aberrant AS could play a role in the malignant phenotype of glioma and to understand the mechanism underlying its aberrant regulation. Methods: We obtained surgical samples from patients with glioblastoma who underwent 5-aminolevulinic fluorescence-guided surgery. Biopsies were taken from the tumor center as well as from adjacent normal-appearing tissue. We used a global splicing array to identify candidate genes aberrantly spliced in these glioblastoma samples. Mechanistic and functional studies were performed to elucidate the role of our top candidate splice variant, BAF45d, in glioblastoma. Results: BAF45d is part of the switch/sucrose nonfermentable complex and plays a key role in the development of the CNS. The BAF45d/6A isoform is present in 85% of over 200 glioma samples that have been analyzed and contributes to the malignant glioma phenotype through the maintenance of an undifferentiated cellular state. We demonstrate that BAF45d splicing is mediated by polypyrimidine tract-binding protein 1 (PTBP1) and that BAF45d regulates PTBP1, uncovering a reciprocal interplay between RNA splicing regulation and transcription. Conclusions: Our data indicate that AS is a mechanism that contributes to the malignant phenotype of glioblastoma. Understanding the consequences of this biological process will uncover new therapeutic targets for this devastating disease.


Asunto(s)
Empalme Alternativo , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Factores de Transcripción/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Movimiento Celular , Proliferación Celular , Glioblastoma/metabolismo , Glioblastoma/patología , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Isoformas de Proteínas , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA