Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
J Appl Clin Med Phys ; 25(2): e14189, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37917885

RESUMEN

PURPOSE: We performed scalp-avoidance whole-brain irradiation with volumetric-modulated arc therapy (SAWB-VMAT) as a component of craniospinal irradiation. In SAWB-VMAT with two coplanar arcs, radiation oncologists and medical physicists sometimes experience difficulty in reducing the dose to the scalp to below the cut-off equivalent dose in 2 Gy per fraction (assuming α/ß = 2) to 50% (EQD50%scalp ). To investigate the advantage of adding coplanar or non-coplanar arcs in reducing the dose to the scalp in SAWB-VMAT, we conducted a planning study to compare the EQD50%scalp , the dose to other organs at risk (OARs), and target coverage in VMAT with two coplanar arcs (Co2arcVMAT), VMAT with three coplanar arcs (Co3arcVMAT), and VMAT with two coplanar and two non-coplanar arcs (NcVMAT). METHODS: Co2arcVMAT, Co3arcVMAT, and NcVMAT plans were created for 10 pediatric patients with medulloblastoma. The planned target volume (PTV) included the regions of the whole brain, cervical spinal cord, cerebrospinal fluid space, and intervertebral foramen. The EQD50%scalp was evaluated separately for four areas (top, back, left, and right) in each case. The prescribed dose for the PTV was 35.2 Gy in 22 fractions. RESULTS: The median EQD50%scalp of the top area was 21.9 , 22.1 , and 18.3 Gy for Co2arcVMAT, Co3arcVMAT, and NcVMAT, respectively. The EQD50%scalp of the top area was significantly reduced in NcVMAT compared to those in Co2arcVMAT and Co3arcVMAT (p < 0.05). The median EQD50%scalp of the top area for NcVMAT was < 19.9 Gy, which is the cut-off dose for severe permanent alopecia. There were no significant differences in EQD50%scalp in the three other areas, the dose to other OARs, or the dose coverage of PTV among the three techniques. CONCLUSION: NcVMAT could reduce the EQD50%scalp of the top area below the cut-off dose of 19.9 Gy. NcVMAT appears to be a promising treatment technique for SAWB-VMAT.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Radioterapia de Intensidad Modulada , Humanos , Niño , Meduloblastoma/radioterapia , Meduloblastoma/etiología , Dosificación Radioterapéutica , Reducción Gradual de Medicamentos , Cuero Cabelludo , Planificación de la Radioterapia Asistida por Computador/métodos , Radioterapia de Intensidad Modulada/métodos , Órganos en Riesgo/efectos de la radiación , Encéfalo , Neoplasias Cerebelosas/etiología
2.
JAMA Oncol ; 9(12): 1688-1695, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37883081

RESUMEN

Importance: Medulloblastoma recurrence in patients who have previously received irradiation has a dismal prognosis and lacks a standard salvage regimen. Objective: To evaluate the response rate of pediatric patients with medulloblastoma recurrence using an antiangiogenic metronomic combinatorial approach (Medulloblastoma European Multitarget Metronomic Anti-Angiogenic Trial [MEMMAT]). Design, Setting, and Participants: This phase 2, investigator-initiated, multicenter nonrandomized controlled trial assessed 40 patients with relapsed or refractory medulloblastoma without a ventriculoperitoneal shunt who were younger than 20 years at original diagnosis. Patients were enrolled between April 1, 2014, and March 31, 2021. Interventions: Treatment consisted of daily oral thalidomide, fenofibrate, celecoxib, and alternating 21-day cycles of low-dose (metronomic) oral etoposide and cyclophosphamide, supplemented by intravenous bevacizumab and intraventricular therapy consisting of alternating etoposide and cytarabine. Main Outcomes and Measures: The primary end point was response after 6 months of antiangiogenic metronomic therapy. Secondary end points included progression-free survival (PFS), overall survival (OS), and quality of life. Adverse events were monitored to assess safety. Results: Of the 40 patients (median [range] age at treatment start, 10 [4-17] years; 25 [62.5%] male) prospectively enrolled, 23 (57.5%) achieved disease control after 6 months of treatment, with a response detected in 18 patients (45.0%). Median OS was 25.5 months (range, 10.9-40.0 months), and median PFS was 8.5 months (range, 1.7-15.4 months). Mean (SD) PFS at both 3 and 5 years was 24.6% (7.9%), while mean (SD) OS at 3 and 5 years was 43.6% (8.5%) and 22.6% (8.8%), respectively. No significant differences in PFS or OS were evident based on molecular subgroup analysis or the number of prior recurrences. In patients demonstrating a response, mean (SD) overall 5-year PFS was 49.7% (14.3%), and for patients who remained progression free for the first 12 months of treatment, mean (SD) 5-year PFS was 66.7% (16.1%). Treatment was generally well tolerated. Grade 3 to 4 treatment-related adverse events included myelosuppression, infections, seizures, and headaches. One heavily pretreated patient with a third recurrence died of secondary acute myeloid leukemia. Conclusions and Relevance: This feasible and well-tolerated MEMMAT combination regimen demonstrated promising activity in patients with previously irradiated recurrent medulloblastoma. Given these results, this predominantly oral, well-tolerated, and outpatient treatment warrants further evaluation. Trial Registration: ClinicalTrials.gov Identifier: NCT01356290.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Cerebelosas , Meduloblastoma , Humanos , Masculino , Niño , Preescolar , Adolescente , Femenino , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/etiología , Etopósido , Calidad de Vida , Administración Metronómica , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/etiología , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos
3.
Curr Treat Options Oncol ; 23(6): 864-886, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35412196

RESUMEN

OPINION STATEMENT: Medulloblastoma (MB) is the most frequent pediatric brain tumor. Despite conventional therapy, MB patients have high mortality and morbidity rates mainly due to the incomplete understanding of the molecular and cellular processes involved in development of this cancer. Similar to other solid tumors, MB demonstrated high endothelial cell proliferation and angiogenic activity, wherein new blood vessels arise from the pre-existing vasculature, a process named angiogenesis. MB angiogenesis is considered a hallmark for MB development, progression, and metastasis emphasizing its potential target for antitumor therapy. However, angiogenesis is tightly regulated by a set of angiogenic factors making it a complex process to be targeted. Although agents targeting these factors and their receptors are early in development, the potential for their targeting may translate into improvement in the clinical care for MB patients. In this review, we focus on the most potent angiogenic factors and their corresponding receptors, highlighting their basic properties and expression in MB. We describe their contribution to MB tumorigenesis and angiogenesis and the potential therapeutic targeting of these factors.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Inductores de la Angiogénesis/uso terapéutico , Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/etiología , Niño , Humanos , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/etiología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo
4.
Curr Treat Options Oncol ; 22(9): 83, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34328587

RESUMEN

OPINION STATEMENT: Medulloblastoma (MB) is the most common pediatric brain malignancy, with a 5-year overall survival (OS) rate of around 65%. The conventional MB treatment, comprising surgical resection followed by irradiation and adjuvant chemotherapy, often leads to impairment in normal body functions and poor quality of life, especially with the increased risk of recurrence and subsequent development of secondary malignancies. The development and progression of MB are facilitated by a variety of immune-evading mechanisms such as the secretion of immunosuppressive molecules, activation of immunosuppressive cells, inhibition of immune checkpoint molecules, impairment of adhesive molecules, downregulation of the major histocompatibility complex (MHC) molecules, protection against apoptosis, and activation of immunosuppressive pathways. Understanding the tumor-immune relationship in MB is crucial for effective development of immune-based therapeutic strategies. In this comprehensive review, we discuss the immunological aspect of the brain, focusing on the current knowledge tackling the mechanisms of MB immune suppression and evasion. We also highlight several key immunotherapeutic approaches developed to date for the treatment of MB.


Asunto(s)
Neoplasias Cerebelosas/etiología , Susceptibilidad a Enfermedades/inmunología , Tolerancia Inmunológica , Meduloblastoma/etiología , Biomarcadores , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Cerebelosas/diagnóstico , Neoplasias Cerebelosas/epidemiología , Neoplasias Cerebelosas/terapia , Toma de Decisiones Clínicas , Terapia Combinada/efectos adversos , Terapia Combinada/métodos , Manejo de la Enfermedad , Humanos , Huésped Inmunocomprometido , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Meduloblastoma/diagnóstico , Meduloblastoma/epidemiología , Meduloblastoma/terapia , Especificidad de Órganos/inmunología , Resultado del Tratamiento , Microambiente Tumoral/inmunología
5.
Genome Med ; 13(1): 103, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34154646

RESUMEN

BACKGROUND: Medulloblastoma (MB) is the most common malignant paediatric brain tumour and a leading cause of cancer-related mortality and morbidity. Existing treatment protocols are aggressive in nature resulting in significant neurological, intellectual and physical disabilities for the children undergoing treatment. Thus, there is an urgent need for improved, targeted therapies that minimize these harmful side effects. METHODS: We identified candidate drugs for MB using a network-based systems-pharmacogenomics approach: based on results from a functional genomics screen, we identified a network of interactions implicated in human MB growth regulation. We then integrated drugs and their known mechanisms of action, along with gene expression data from a large collection of medulloblastoma patients to identify drugs with potential to treat MB. RESULTS: Our analyses identified drugs targeting CDK4, CDK6 and AURKA as strong candidates for MB; all of these genes are well validated as drug targets in other tumour types. We also identified non-WNT MB as a novel indication for drugs targeting TUBB, CAD, SNRPA, SLC1A5, PTPRS, P4HB and CHEK2. Based upon these analyses, we subsequently demonstrated that one of these drugs, the new microtubule stabilizing agent, ixabepilone, blocked tumour growth in vivo in mice bearing patient-derived xenograft tumours of the Sonic Hedgehog and Group 3 subtype, providing the first demonstration of its efficacy in MB. CONCLUSIONS: Our findings confirm that this data-driven systems pharmacogenomics strategy is a powerful approach for the discovery and validation of novel therapeutic candidates relevant to MB treatment, and along with data validating ixabepilone in PDX models of the two most aggressive subtypes of medulloblastoma, we present the network analysis framework as a resource for the field.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor , Neoplasias Cerebelosas/etiología , Desarrollo de Medicamentos , Meduloblastoma/etiología , Farmacogenética/métodos , Animales , Antineoplásicos/uso terapéutico , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/metabolismo , Biología Computacional/métodos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Humanos , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/metabolismo , Ratones , Ratones Transgénicos , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Biología de Sistemas/métodos , Transcriptoma , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Radiat Res ; 196(2): 225-234, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34046685

RESUMEN

Neutron radiation, a high-linear energy transfer radiation, has a high relative biological effectiveness (RBE) for various end points. The age at exposure is an important modifier of the effects of radiation, including carcinogenesis, with infants being generally more radiosensitive. Ptch1+/- mice offer a unique experimental system for assessing radiation carcinogenesis. Spontaneous development of medulloblastoma tumors occurs in nonirradiated animals that lose their Ptch1+ allele, most frequently by a loss of heterozygosity (LOH) of chromosome 13 via recombination or non-disjunction (referred to as S-type tumors). In contrast, tumors occur in irradiated Ptch1+/- mice as a result of chromosome 13 LOH with an interstitial deletion (R-type), making spontaneous and radiation-induced tumors discernible. To elucidate the influence of age on the effect of fast neutrons, we irradiated Ptch1+/- mice with neutrons (mean energy, ∼2 MeV) or γ rays on embryonic day (E)14 and E17 and on postnatal day (P)1, 4 or 10 and classified the resulting medulloblastomas based on chromosome 13 aberrations. Instead of LOH, some tumors harbored mutations in their Ptch1+ gene via a nonirradiation-associated mechanism such as duplication, insertion, base substitution or deletion with microhomology-mediated end joining; thus, these tumors were classified as S-type. The RBE regarding the induction of R-type tumors was 12.9 (8.6, 17.2), 9.6 (6.9, 12.3), 21.5 (17.2, 25.8), and 7.1 (4.7, 9.5) (mean and 95% confidence interval) for mice irradiated on E14, E17, P1 and P4, respectively, with the highest value seen during the most active development of the tissue and P10 being completely resistant. These results indicate that the developmental stage at exposure of the tissue influences the RBE of neutrons.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 13/efectos de la radiación , Meduloblastoma/genética , Neoplasias Inducidas por Radiación/genética , Receptor Patched-1/genética , Animales , Cromosomas Humanos Par 13/genética , Relación Dosis-Respuesta en la Radiación , Neutrones Rápidos/efectos adversos , Humanos , Pérdida de Heterocigocidad/genética , Pérdida de Heterocigocidad/efectos de la radiación , Meduloblastoma/etiología , Meduloblastoma/patología , Ratones , Neoplasias Inducidas por Radiación/patología , Tolerancia a Radiación/genética , Tolerancia a Radiación/efectos de la radiación , Efectividad Biológica Relativa
7.
Genome Biol ; 22(1): 125, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33926529

RESUMEN

BACKGROUND: Medulloblastoma (MB) is the most common malignant pediatric brain tumor that originates in the cerebellum and brainstem. Frequent somatic mutations and deregulated expression of epigenetic regulators in MB highlight the substantial role of epigenetic alterations. 5-hydroxymethylcytosine (5hmC) is a highly abundant cytosine modification in the developing cerebellum and is regulated by ten-eleven translocation (TET) enzymes. RESULTS: We investigate the alterations of 5hmC and TET enzymes in MB and their significance to cerebellar cancer formation. We show total abundance of 5hmC is reduced in MB, but identify significant enrichment of MB-specific 5hmC marks at regulatory regions of genes implicated in stem-like properties and Nanog-binding motifs. While TET1 and TET2 levels are high in MBs, only knockout of Tet1 in the smoothened (SmoA1) mouse model attenuates uncontrolled proliferation, leading to a favorable prognosis. The pharmacological Tet1 inhibition reduces cell viability and platelet-derived growth factor signaling pathway-associated genes. CONCLUSIONS: These results together suggest a potential key role of 5hmC and indicate an oncogenic nature for TET1 in MB tumorigenesis, suggesting it as a potential therapeutic target for MBs.


Asunto(s)
Susceptibilidad a Enfermedades , Meduloblastoma/etiología , Meduloblastoma/metabolismo , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/análogos & derivados , Animales , Biomarcadores de Tumor , Biología Computacional/métodos , Islas de CpG , Metilación de ADN , Bases de Datos de Ácidos Nucleicos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Meduloblastoma/mortalidad , Meduloblastoma/patología , Ratones , Ratones Transgénicos , Motivos de Nucleótidos , Pronóstico
8.
Sci Rep ; 11(1): 4259, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33608621

RESUMEN

Studying medulloblastoma, the most common malignant paediatric brain tumour, requires simple yet realistic in vitro models. In this study, we optimised a robust, reliable, three-dimensional (3D) culture method for medulloblastoma able to recapitulate the spatial conformation, cell-cell and cell-matrix interactions that exist in vivo and in patient tumours. We show that, when grown under the same stem cell enriching conditions, SHH subgroup medulloblastoma cell lines established tight, highly reproducible 3D spheroids that could be maintained for weeks in culture and formed pathophysiological oxygen gradients. 3D spheroid culture also increased resistance to standard-of-care chemotherapeutic drugs compared to 2D monolayer culture. We exemplify how this model can enhance in vitro therapeutic screening approaches through dual-inhibitor studies and continual monitoring of drug response. Next, we investigated the initial stages of metastatic dissemination using brain-specific hyaluronan hydrogel matrices. RNA sequencing revealed downregulation of cell cycle genes and upregulation of cell movement genes and key fibronectin interactions in migrating cells. Analyses of these upregulated genes in patients showed that their expression correlated with early relapse and overall poor prognosis. Our 3D spheroid model is a significant improvement over current in vitro techniques, providing the medulloblastoma research community with a well-characterised and functionally relevant culture method.


Asunto(s)
Antineoplásicos/farmacología , Técnicas de Cultivo de Célula , Proteínas Hedgehog/metabolismo , Meduloblastoma/metabolismo , Meduloblastoma/patología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Biomarcadores de Tumor , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/genética , Humanos , Meduloblastoma/etiología , Meduloblastoma/mortalidad , Pronóstico , Células Tumorales Cultivadas
9.
Oncogene ; 40(2): 396-407, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33159168

RESUMEN

The immune microenvironment of tumors can play a critical role in promoting or inhibiting tumor progression depending on the context. We present evidence that tumor-associated macrophages/microglia (TAMs) can promote tumor progression in the sonic hedgehog subgroup of medulloblastoma (SHH-MB). By combining longitudinal manganese-enhanced magnetic resonance imaging (MEMRI) and immune profiling of a sporadic mouse model of SHH-MB, we found the density of TAMs is higher in the ~50% of tumors that progress to lethal disease. Furthermore, reducing regulatory T cells or eliminating B and T cells in Rag1 mutants does not alter SHH-MB tumor progression. As TAMs are a dominant immune component in tumors and are normally dependent on colony-stimulating factor 1 receptor (CSF1R), we treated mice with a CSF1R inhibitor, PLX5622. Significantly, PLX5622 reduces a subset of TAMs, prolongs mouse survival, and reduces the volume of most tumors within 4 weeks of treatment. Moreover, concomitant with a reduction in TAMs the percentage of infiltrating cytotoxic T cells is increased, indicating a change in the tumor environment. Our studies in an immunocompetent preclinical mouse model demonstrate TAMs can have a functional role in promoting SHH-MB progression. Thus, CSF1R inhibition could have therapeutic potential for a subset of SHH-MB patients.


Asunto(s)
Neoplasias Cerebelosas/prevención & control , Modelos Animales de Enfermedad , Proteínas Hedgehog/fisiología , Meduloblastoma/prevención & control , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Macrófagos Asociados a Tumores/inmunología , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Proliferación Celular , Neoplasias Cerebelosas/etiología , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Femenino , Humanos , Masculino , Meduloblastoma/etiología , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pronóstico , Células Tumorales Cultivadas , Microambiente Tumoral
10.
Curr Treat Options Oncol ; 22(1): 6, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33245404

RESUMEN

OPINION STATEMENT: Medulloblastoma is the most frequently diagnosed primary malignant brain tumor among children. Currently available therapeutic strategies are based on surgical resection, chemotherapy, and/or radiotherapy. However, majority of patients quickly develop therapeutic resistance and are often left with long-term therapy-related side effects and sequelae. Therefore, there remains a dire need to develop more effective therapeutics to overcome the acquired resistance to currently available therapies. Unfortunately, the process of developing novel anti-neoplastic drugs from bench to bedside is highly time-consuming and very expensive. A wide range of drugs that are already in clinical use for treating non-cancerous diseases might commonly target tumor-associated signaling pathways as well and hence be of interest in treating different cancers. This is referred to as drug repurposing or repositioning. In medulloblastoma, drug repurposing has recently gained a remarkable interest as an alternative therapy to overcome therapy resistance, wherein existing non-tumor drugs are being tested for their potential anti-neoplastic effects outside the scope of their original use.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Cerebelosas/tratamiento farmacológico , Reposicionamiento de Medicamentos , Meduloblastoma/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Cerebelosas/diagnóstico , Neoplasias Cerebelosas/etiología , Toma de Decisiones Clínicas , Estudios Clínicos como Asunto , Terapia Combinada , Manejo de la Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Meduloblastoma/diagnóstico , Meduloblastoma/etiología , Pronóstico , Resultado del Tratamiento
12.
Cell Rep ; 31(2): 107511, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294450

RESUMEN

Medulloblastoma (MB) is a neoplasm linked to dysregulated cerebellar development. Previously, we demonstrated that the Sonic Hedgehog (SHH) subgroup grows hierarchically, with Sox2+ cells at the apex of tumor progression and relapse. To test whether this mechanism is rooted in a normal developmental process, we studied the role of Sox2 in cerebellar development. We find that the external germinal layer (EGL) is derived from embryonic Sox2+ precursors and that the EGL maintains a rare fraction of Sox2+ cells during the first postnatal week. Through lineage tracing and single-cell analysis, we demonstrate that these Sox2+ cells are within the Atoh1+ lineage, contribute extensively to adult granule neurons, and resemble Sox2+ tumor cells. Critically, constitutive activation of the SHH pathway leads to their aberrant persistence in the EGL and rapid tumor onset. We propose that failure to eliminate this rare but potent developmental population is the tumor initiation mechanism in SHH-subgroup MB.


Asunto(s)
Meduloblastoma/etiología , Meduloblastoma/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Linaje de la Célula/genética , Células Cultivadas , Neoplasias Cerebelosas/patología , Cerebelo/embriología , Femenino , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Ratones Noqueados , Ratones Transgénicos , Recurrencia Local de Neoplasia/patología , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/metabolismo , Factores de Transcripción SOXB1/fisiología , Transducción de Señal/fisiología , Análisis de la Célula Individual/métodos
13.
J Pediatr Hematol Oncol ; 42(5): e381-e384, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31306337

RESUMEN

Medulloblastoma is the most common malignant pediatric brain tumor. Survival rates range between 50% and 80% depending on histology and other biologic features, metastases, and treatment approach. Prader-Willi syndrome (PWS) is a genetically inherited disorder characterized by dysmorphic features, mental retardation, obesity, and hypogonadism among other features. We describe a 10.5-year-old girl with PWS and previous standard-risk medulloblastoma that relapsed in the pons 3 years after the end of treatment. Diagnosis of relapse was delayed by a preceding varicella infection, an initial clinical/radiologic response to steroids and the unusual location, and was confirmed with a stereotactic biopsy. Second-line therapy was commenced, however, the patient rapidly deteriorated and died. This is the first report of medulloblastoma in a patient with PWS.


Asunto(s)
Neoplasias Cerebelosas/diagnóstico , Meduloblastoma/diagnóstico , Recurrencia Local de Neoplasia/diagnóstico , Síndrome de Prader-Willi/complicaciones , Neoplasias Cerebelosas/etiología , Niño , Femenino , Humanos , Meduloblastoma/etiología , Recurrencia Local de Neoplasia/etiología , Pronóstico
14.
Nat Rev Cancer ; 20(1): 42-56, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31819232

RESUMEN

Medulloblastoma, a malignant brain tumour primarily diagnosed during childhood, has recently been the focus of intensive molecular profiling efforts, profoundly advancing our understanding of biologically and clinically heterogeneous disease subgroups. Genomic, epigenomic, transcriptomic and proteomic landscapes have now been mapped for an unprecedented number of bulk samples from patients with medulloblastoma and, more recently, for single medulloblastoma cells. These efforts have provided pivotal new insights into the diverse molecular mechanisms presumed to drive tumour initiation, maintenance and recurrence across individual subgroups and subtypes. Translational opportunities stemming from this knowledge are continuing to evolve, providing a framework for improved diagnostic and therapeutic interventions. In this Review, we summarize recent advances derived from this continued molecular characterization of medulloblastoma and contextualize this progress towards the deployment of more effective, molecularly informed treatments for affected patients.


Asunto(s)
Susceptibilidad a Enfermedades , Meduloblastoma/diagnóstico , Meduloblastoma/etiología , Animales , Biomarcadores de Tumor , Terapia Combinada , Manejo de la Enfermedad , Epigenómica/métodos , Estudios de Asociación Genética , Genómica/métodos , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/terapia , Técnicas de Diagnóstico Molecular , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Proteómica/métodos , Recurrencia , Resultado del Tratamiento , Microambiente Tumoral
15.
Rev Gastroenterol Peru ; 39(3): 280-283, 2019.
Artículo en Español | MEDLINE | ID: mdl-31688854

RESUMEN

Turcot syndrome is an association of primary neuroepithelial tumors of the central nervous system with adenomatous polyposis coli. It is a genetic disorder, with two forms; In type I, glioblastomas are usually associated with hereditary nonpolyposis colorectal cancer (HNPC or Lynch). In Type II, medulloblastomas are often associated with familial adenomatous polyposis coli (classical or attenuated). This patient had a medulloblastoma at seven years of age, then 20 years later developed a meningioma which recurred several times. At 36 years old he presented with anemia after digestive bleeding, and an adenomatous polyposis coli with high grade dysplasia was found at colonoscopy. As far as we know, this is the first case of Turcot syndrome described in our country.


Asunto(s)
Poliposis Adenomatosa del Colon/etiología , Neoplasias Encefálicas/complicaciones , Neoplasias Cerebelosas/etiología , Neoplasias Colorrectales/complicaciones , Meduloblastoma/etiología , Neoplasias Meníngeas/etiología , Meningioma/etiología , Recurrencia Local de Neoplasia/etiología , Síndromes Neoplásicos Hereditarios/complicaciones , Adulto , Humanos , Masculino
16.
Curr Treat Options Oncol ; 20(8): 64, 2019 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-31236711

RESUMEN

OPINION STATEMENT: Medulloblastoma (MB) is a malignant embryonal tumor of the posterior fossa and is the most common type of brain cancer in pediatric patients. In contrast, adult MB is very rare with an incidence of 0.6 per million per year and mostly affects young adults below the age of 40. Recent molecular analyses of pediatric and adult MB have classified these tumors into at least four individual molecular subgroups (SHH, WNT, group 3, and group 4) with distinct demographics, histology, and prognosis. The discrete biological composition of these tumors likely explains the marked heterogeneity in responses seen to conventional therapies such as radiation and cytotoxic chemotherapies. Given the low incidence of adult MB, prospective studies are challenging and scarce, and management guidelines are largely derived from the pediatric MB patient population and retrospective data. However, adult MB is clinically and molecularly distinct from pediatric MB and a comprehensive review of published literature on adult MB highlighting their differences is warranted. Here, we review the management of adult MB focusing on recent studies exploring the effectiveness of upfront chemotherapy, clinical trials in the context of molecular subgroup-specific therapies, and the potential role of immunotherapy in treating this disease.


Asunto(s)
Neoplasias Cerebelosas/diagnóstico , Neoplasias Cerebelosas/terapia , Meduloblastoma/diagnóstico , Meduloblastoma/terapia , Neoplasias Cerebelosas/etiología , Neoplasias Cerebelosas/mortalidad , Ensayos Clínicos como Asunto , Terapia Combinada , Diagnóstico por Imagen , Manejo de la Enfermedad , Humanos , Meduloblastoma/etiología , Meduloblastoma/mortalidad , Clasificación del Tumor , Metástasis de la Neoplasia , Estadificación de Neoplasias , Resultado del Tratamiento
17.
Sci Rep ; 8(1): 6913, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29720672

RESUMEN

Group 3 tumors account for 28% of medulloblastomas and have the worst prognosis. FTY720, an immunosuppressant currently approved for treatment of multiple sclerosis, has shown antitumor effects in several human cancer cell lines. We hypothesized that treatment with FTY720 (fingolimod) would decrease tumorigenicity in medulloblastoma patient-derived xenografts (PDXs). Three Group 3 medulloblastoma PDXs (D341, D384 and D425) were utilized. Expression of PP2A and its endogenous inhibitors I2PP2A and CIP2A was detected by immunohistochemistry and immunoblotting. PP2A activation was measured via phosphatase activation kit. Cell viability, proliferation, migration and invasion assays were performed after treatment with FTY720. Cell cycle analysis was completed using flow cytometry. A flank model using D425 human medulloblastoma PDX cells was used to assess the in vivo effects of FTY720. FTY720 activated PP2A and led to decreased medulloblastoma PDX cell viability, proliferation, migration and invasion and G1 cell cycle arrest in all three PDXs. FTY720 treatment of mice bearing D425 medulloblastoma PDX tumors resulted in a significant decrease in tumor growth compared to vehicle treated animals. FTY720 decreased viability, proliferation, and motility in Group 3 medulloblastoma PDX cells and significantly decreased tumor growth in vivo. These results suggest that FTY720 should be investigated further as a potential therapeutic agent for medulloblastoma.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Meduloblastoma/etiología , Meduloblastoma/patología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Med Genet ; 54(8): 530-536, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28596197

RESUMEN

BACKGROUND: Gorlin syndrome (GS) is an autosomal dominant syndrome characterised by multiple basal cell carcinomas (BCCs) and an increased risk of jaw cysts and early childhood medulloblastoma. Heterozygous germline variants in PTCH1 and SUFU encoding components of the Sonic hedgehog pathway explain the majority of cases. Here, we aimed to delineate genotype-phenotype correlations in GS. METHODS: We assessed genetic and phenotypic data for 182 individuals meeting the diagnostic criteria for GS (median age: 47.1; IQR: 31.1-61.1). A total of 126 patients had a heterozygous pathogenic variant, 9 had SUFU pathogenic variants and 46 had no identified mutation. RESULTS: Patients with variants were more likely to be diagnosed earlier (p=0.02), have jaw cysts (p=0.002) and have bifid ribs (p=0.003) or any skeletal abnormality (p=0.003) than patients with no identified mutation. Patients with a missense variant in PTCH1 were diagnosed later (p=0.03) and were less likely to develop at least 10 BCCs and jaw cysts than those with other pathogenic PTCH1 variants (p=0.03). Patients with SUFU pathogenic variants were significantly more likely than those with PTCH1 pathogenic variants to develop a medulloblastoma (p=0.009), a meningioma (p=0.02) or an ovarian fibroma (p=0.015), but were less likely to develop a jaw cyst (p=0.0004). CONCLUSION: We propose that the clinical heterogeneity of GS can in part be explained by the underlying or SUFU variant.


Asunto(s)
Síndrome del Nevo Basocelular/genética , Síndrome del Nevo Basocelular/patología , Receptor Patched-1/genética , Proteínas Represoras/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Síndrome del Nevo Basocelular/complicaciones , Neoplasias Cerebelosas/etiología , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Mutación de Línea Germinal , Humanos , Lactante , Masculino , Meduloblastoma/etiología , Meduloblastoma/genética , Meduloblastoma/patología , Persona de Mediana Edad
20.
Radiat Res ; 186(4): 407-414, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27690174

RESUMEN

Recently reported studies have led to a heightened awareness of the risks of cancer induced by diagnostic radiological imaging, and in particular, the risk of brain cancer after childhood CT scans. One feature of Ptch1+/- mice is their sensitivity to radiation-induced medulloblastomas (an embryonic cerebellar tumor) during a narrow window of time centered on the days around birth. Little is known about the dynamics of how dose protraction interacts with such narrow windows of sensitivity in individual tissues. Using medulloblastomas from irradiated Ptch1+/- mice with a hybrid C3H × C57BL/6 F1 genetic background, we previously showed that the alleles retained on chromosome 13 (which harbors the Ptch1 gene) reveal two major mechanisms of loss of the wild-type allele. The loss of parental alleles from the telomere extending up to or past the Ptch1 locus by recombination (spontaneous type) accounts for almost all medulloblastomas in nonirradiated mice, while tumors in irradiated mice often exhibited interstitial deletions, which start downstream of the wild-type Ptch1 and extend up varying lengths towards the centromere (radiation type). In this study, Ptch1+/- mice were exposed to an acute dose of either 100 or 500 mGy gamma rays in utero or postnatally, or the same radiation doses protracted over a four-day period, and were monitored for medulloblastoma development. The results showed dose- and age-dependent radiation-induced type tumors. Furthermore, the size of the radiation-induced deletion differed with the dose rate. The results of this work suggest that tumor latency may be related to the size of the deletion. In this study, 500 mGy exposure produced radiation-induced type tumors at all ages and dose rates, while 100 mGy exposure did not significantly produce radiation-induced type tumors. The radiation signature allows for unique mechanistic insight into the action of radiation to induce DNA lesions with known causal relationship to a specific tumor type, particularly for doses and dose rates that are relevant to both diagnostic and accidental radiological exposures.


Asunto(s)
Neoplasias Cerebelosas/genética , Rayos gamma/efectos adversos , Heterocigoto , Meduloblastoma/genética , Neoplasias Inducidas por Radiación/genética , Receptor Patched-1/genética , Animales , Neoplasias Cerebelosas/etiología , Neoplasias Cerebelosas/patología , Cromosomas de los Mamíferos/genética , Cromosomas de los Mamíferos/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Estimación de Kaplan-Meier , Pérdida de Heterocigocidad/efectos de la radiación , Meduloblastoma/etiología , Meduloblastoma/patología , Ratones , Neoplasias Inducidas por Radiación/etiología , Neoplasias Inducidas por Radiación/patología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...