Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Protoc ; 18(7): 2143-2180, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37248391

RESUMEN

Medulloblastoma and high-grade glioma represent the most aggressive and frequent lethal solid tumors affecting individuals during pediatric age. During the past years, several models have been established for studying these types of cancers. Human organoids have recently been shown to be a valid alternative model to study several aspects of brain cancer biology, genetics and test therapies. Notably, brain cancer organoids can be generated using genetically modified cerebral organoids differentiated from human induced pluripotent stem cells (hiPSCs). However, the protocols to generate them and their downstream applications are very rare. Here, we describe the protocols to generate cerebellum and forebrain organoids from hiPSCs, and the workflow to genetically modify them by overexpressing genes found altered in patients to finally produce cancer organoids. We also show detailed protocols to use medulloblastoma and high-grade glioma organoids for orthotopic transplantation and co-culture experiments aimed to study cell biology in vivo and in vitro, for lineage tracing to investigate the cell of origin and for drug screening. The protocol takes 60-65 d for cancer organoids generation and from 1-4 weeks for downstream applications. The protocol requires at least 3-6 months to become proficient in culturing hiPSCs, generating organoids and performing procedures on immunodeficient mice.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Cerebelosas , Glioma , Células Madre Pluripotentes Inducidas , Meduloblastoma , Humanos , Niño , Animales , Ratones , Meduloblastoma/genética , Meduloblastoma/patología , Técnicas de Cocultivo , Evaluación Preclínica de Medicamentos , Glioma/patología , Organoides , Prosencéfalo , Diferenciación Celular , Neoplasias Cerebelosas/patología
2.
Methods Mol Biol ; 2423: 95-101, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34978691

RESUMEN

Conventional chemotherapies for medulloblastoma are restricted to only proliferative population leaving the cancer stem cells unscathed. This shortcoming of the traditional therapies is attributed to the relapse and metastasis of the cancer. The current research is entirely focused on the screening of therapeutic agents that can restrict and target the self-renewal potential of the cancer stem cells. The advances in drug screening strategies have led to high-throughput screening which provide a robust and expeditious platform to screen potential compounds against cancer stem cells. In this book chapter, we describe two in vitro assays that are routinely used to measure the cell killing and anti-self-renewal activity of the compounds against the cancer stem cells. Combining these assays with high-throughput screening offers a rapid, reliable, and inexpensive approach to screen potential compounds against cancer stem cells and to overcome the limitation of conventional chemotherapeutic agents.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/patología , Evaluación Preclínica de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Meduloblastoma/patología , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Bibliotecas de Moléculas Pequeñas/farmacología
3.
J Adolesc Young Adult Oncol ; 11(1): 68-77, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33891492

RESUMEN

Purpose: Medulloblastomas, comprising 20%-25% of all primary brain tumors in children are much rarer in adulthood. Disease biology varies substantially across different age groups; however, owing to rarity, adults with medulloblastoma are traditionally treated using pediatric protocols. This is a retrospective audit of adolescent and adult medulloblastoma from a comprehensive cancer center. Methods: Data regarding demography, clinical presentation, imaging characteristics, histopathological features, molecular profiling, risk stratification, treatment details, and outcomes were retrieved from medical records. All time-to-event outcomes were analyzed using Kaplan-Meier method and compared with the log-rank test. Univariate and multivariate analysis of relevant prognostic factors was done with p value <0.05 being considered statistically significant. Results: A total of 162 patients ≥15 years of age with medulloblastoma were included. The median age was 25 years (range: 15-59 years) with leptomeningeal metastases seen in 31 (19%) patients at initial diagnosis. Following surgery, patients were treated with appropriate risk-stratified adjuvant therapy comprising of craniospinal irradiation plus boost with or without systemic chemotherapy. At a median follow-up of 50 months, 5-year Kaplan-Meier estimates of progression-free survival and overall survival were 53.5% and 59.5%, respectively. The addition of adjuvant systemic chemotherapy did not impact upon survival in standard-risk medulloblastoma. High-risk (HR) disease and anaplastic histology emerged as significant and independent predictors of poor survival on multivariate analysis. Conclusion: Medulloblastoma is a rare tumor in adolescents and adults with key differences in disease biology and resultant outcomes compared with the pediatric population. Contemporary management comprising maximal safe resection followed by appropriate risk-stratified adjuvant therapy provides acceptable survival outcomes.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Adolescente , Adulto , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/terapia , Niño , Auditoría Clínica , Humanos , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/patología , Pronóstico , Estudios Retrospectivos
4.
Biochem Pharmacol ; 190: 114593, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33964282

RESUMEN

Aberrant activation of the Hedgehog (Hh) pathway is implicated in the pathogenesis and development of multiple cancers, especially Hh-driven medulloblastoma (MB). Smoothened (SMO) is a promising therapeutic target of the Hh pathway in clinical cancer treatment. However, SMO mutations frequently occur, which leads to drug resistance and tumor relapse. Novel inhibitors that target both the wild-type and mutant SMO are in high demand. In this study, we identified a novel Hh pathway inhibitor, pseudolaric acid B (PAB), which significantly inhibited the expression of Gli1 and its transcriptional target genes, such as cyclin D1 and N-myc, thus inhibiting the proliferation of DAOY and Ptch1+/- primary MB cells. Mechanistically, PAB can potentially bind to the extracellular entrance of the heptahelical transmembrane domain (TMD) of SMO, based on molecular docking and the BODIPY-cyclopamine binding assay. Further, PAB also efficiently blocked ciliogenesis, demonstrating the inhibitory effects of PAB on the Hh pathway at multiple levels. Thus, PAB may overcome drug-resistance induced by SMO mutations, which frequently occurs in clinical setting. PAB markedly suppressed tumor growth in the subcutaneous allografts of Ptch1+/- MB cells. Together, our results identified PAB as a potent Hh pathway inhibitor to treat Hh-dependent MB, especially cases resistant to SMO antagonists.


Asunto(s)
Neoplasias Cerebelosas/tratamiento farmacológico , Diterpenos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Proteínas Hedgehog/antagonistas & inhibidores , Meduloblastoma/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Células A549 , Animales , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Diterpenos/química , Diterpenos/uso terapéutico , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Células HEK293 , Células HeLa , Proteínas Hedgehog/química , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Células 3T3 NIH , Estructura Secundaria de Proteína , Transducción de Señal/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Cancer Res ; 80(13): 2818-2832, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32371475

RESUMEN

Aberrant activation of the Hedgehog (HH) signaling pathway underlines the initiation and progression of a multitude of cancers. The effectiveness of the leading drugs vismodegib (GDC-0449) and sonidegib (LDE225), both Smoothened (SMO) antagonists, is compromised by acquisition of mutations that alter pathway components, notably secondary mutations in SMO and amplification of GLI2, a transcriptional mediator at the end of the pathway. Pharmacologic blockade of GLI2 activity could ultimately overcome these diversified refractory mechanisms, which would also be effective in a broader spectrum of primary tumors than current SMO antagonists. To this end, we conducted a high-content screening directly analyzing the ciliary translocation of GLI2, a key event for GLI2 activation in HH signal transduction. Several prostaglandin compounds were shown to inhibit accumulation of GLI2 within the primary cilium (PC). In particular, prostaglandin E1 (PGE1), an FDA-approved drug, is a potent GLI2 antagonist that overcame resistance mechanisms of both SMO mutagenesis and GLI2 amplification. Consistent with a role in HH pathway regulation, EP4 receptor localized to the PC. Mechanistically, PGE1 inhibited HH signaling through the EP4 receptor, enhancing cAMP-PKA activity, which promoted phosphorylation and degradation of GLI2 via the ubiquitination pathway. PGE1 also effectively inhibited the growth of drug refractory human medulloblastoma xenografts. Together, these results identify PGE1 and other prostaglandins as potential templates for complementary therapeutic development to circumvent resistance to current generation SMO antagonists in use in the clinic. SIGNIFICANCE: These findings show that PGE1 exhibits pan-inhibition against multiple drug refractory activities for Hedgehog-targeted therapies and elicits significant antitumor effects in xenograft models of drug refractory human medulloblastoma mimicking GLI2 amplification.


Asunto(s)
Alprostadil/farmacología , Neoplasias Cerebelosas/tratamiento farmacológico , Resistencia a Antineoplásicos , Amplificación de Genes , Proteínas Hedgehog/antagonistas & inhibidores , Meduloblastoma/tratamiento farmacológico , Proteínas Nucleares/genética , Proteína Gli2 con Dedos de Zinc/genética , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Ratones Endogámicos NOD , Inhibidores de Agregación Plaquetaria/farmacología , Subtipo EP4 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Biochem Mol Toxicol ; 34(5): e22471, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32057176

RESUMEN

BACKGROUND: Medulloblastoma is a common tumor originates from central nervous system in children with metastatic potential. Geniposide is the major active ingredient separated from the fruit of Gardenia jasminoides Ellis. Herein, we tested the possible anticancer activity of geniposide on human medulloblastoma cells, as well as the potential underlying molecular mechanisms. METHODS: Firstly, followed by geniposide incubation, cell viability, proliferation, apoptosis, migration, and invasion of medulloblastoma Daoy cells, along with microRNA-373 (miR-373) expression were tested, respectively. Then, the influences of miR-373 overexpression in the reduction of medulloblastoma cell proliferation, migration, and invasion and the elevation of apoptosis, triggered by geniposide treatment, were re-investigated. Finally, the Ras/Raf/MEK/ERK pathway activity was analyzed. RESULTS: Geniposide treatment inhibited medulloblastoma cell viability, proliferation, migration, and invasion, but promoted cell apoptosis. Surprisingly, miR-373 expression in medulloblastoma cells was obviously downregulated by geniposide treatment. miR-373 overexpression reversed the effects of geniposide on Daoy cells. Furthermore, geniposide hindered the Ras/Raf/MEK/ERK pathway by downregulating miR-373 expression. CONCLUSION: Geniposide exhibited anticancer activity on human medulloblastoma cells and blocked Ras/Raf/MEK/ERK pathway by downregulating miR-373 expression.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Cerebelosas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Frutas/química , Gardenia/química , Iridoides/farmacología , Meduloblastoma/metabolismo , MicroARNs/metabolismo , Extractos Vegetales/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Cerebelosas/patología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Meduloblastoma/patología , MicroARNs/genética , Invasividad Neoplásica/genética , Transfección
7.
Biomed Pharmacother ; 124: 109887, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31986414

RESUMEN

PURPOSE: Long non-coding RNA (LncRNA) HOX transcript antisense RNA (HOTAIR) and Yin Yang 1 (YY1) are reported to be involved in tumorigenesis. However, the effect and molecular mechanism of HOTAIR on YY1 expression remains poorly understood. The study aimed to investigate the functions and molecular mechanism of LncRNA HOTAIR in medulloblastoma progression. METHODS: qPCR was performed to detect HOTAIR and YY1 mRNA in tissues and cells, as well as that of miR-1 and miR-206 expression levels. Western blot assay was used to test YY1 and EMT-related biomarkers' protein levels. Cell proliferation was tested with CCK-8 assay and colony formation assay. Migration and invasion abilities were tested with Transwell migration and invasion assays. Tumor growth was tested with an in vivo animal study. Cell apoptosis was tested with an Annexin V-FITC/PI kit. Luciferase assay was used to test the luciferase intensity of YY1 and HOTAIR. RNA pull down assay was used to detect the combination between HOTAIR and miR-1/miR-206. RESULTS: In this study, we found that HOTAIR and YY1 were up-regulated in medulloblastoma tissues and cell lines, and HOTAIR increased YY1 expression. The molecular mechanism demonstrated that HOTAIR negatively regulated miR-1 and miR-206 expression, which can directly target YY1 in medulloblastoma cells. Moreover, HOTAIR increased YY1 expression through binding to miR-1 and miR-206. The functional experiments showed that HOTAIR knockdown suppressed medulloblastoma cell proliferation, tumor growth, migration and invasion, and promoted cell apoptosis via the modulation of the miR-1/miR-206-YY1 axis, as well as epithelial to mesenchymal transition (EMT). CONCLUSION: These data indicate that HOTAIR promotes medulloblastoma progression via acting as a competing endogenous RNA (ceRNA) to regulate YY1 expression through binding to miR-1 and miR-206.


Asunto(s)
Neoplasias Cerebelosas/genética , Meduloblastoma/genética , ARN Largo no Codificante/genética , Factor de Transcripción YY1/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Cerebelosas/patología , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Meduloblastoma/patología , Ratones , Ratones Desnudos , MicroARNs/genética , Invasividad Neoplásica/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Lancet Oncol ; 19(6): 785-798, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29753700

RESUMEN

BACKGROUND: Medulloblastoma is associated with rare hereditary cancer predisposition syndromes; however, consensus medulloblastoma predisposition genes have not been defined and screening guidelines for genetic counselling and testing for paediatric patients are not available. We aimed to assess and define these genes to provide evidence for future screening guidelines. METHODS: In this international, multicentre study, we analysed patients with medulloblastoma from retrospective cohorts (International Cancer Genome Consortium [ICGC] PedBrain, Medulloblastoma Advanced Genomics International Consortium [MAGIC], and the CEFALO series) and from prospective cohorts from four clinical studies (SJMB03, SJMB12, SJYC07, and I-HIT-MED). Whole-genome sequences and exome sequences from blood and tumour samples were analysed for rare damaging germline mutations in cancer predisposition genes. DNA methylation profiling was done to determine consensus molecular subgroups: WNT (MBWNT), SHH (MBSHH), group 3 (MBGroup3), and group 4 (MBGroup4). Medulloblastoma predisposition genes were predicted on the basis of rare variant burden tests against controls without a cancer diagnosis from the Exome Aggregation Consortium (ExAC). Previously defined somatic mutational signatures were used to further classify medulloblastoma genomes into two groups, a clock-like group (signatures 1 and 5) and a homologous recombination repair deficiency-like group (signatures 3 and 8), and chromothripsis was investigated using previously established criteria. Progression-free survival and overall survival were modelled for patients with a genetic predisposition to medulloblastoma. FINDINGS: We included a total of 1022 patients with medulloblastoma from the retrospective cohorts (n=673) and the four prospective studies (n=349), from whom blood samples (n=1022) and tumour samples (n=800) were analysed for germline mutations in 110 cancer predisposition genes. In our rare variant burden analysis, we compared these against 53 105 sequenced controls from ExAC and identified APC, BRCA2, PALB2, PTCH1, SUFU, and TP53 as consensus medulloblastoma predisposition genes according to our rare variant burden analysis and estimated that germline mutations accounted for 6% of medulloblastoma diagnoses in the retrospective cohort. The prevalence of genetic predispositions differed between molecular subgroups in the retrospective cohort and was highest for patients in the MBSHH subgroup (20% in the retrospective cohort). These estimates were replicated in the prospective clinical cohort (germline mutations accounted for 5% of medulloblastoma diagnoses, with the highest prevalence [14%] in the MBSHH subgroup). Patients with germline APC mutations developed MBWNT and accounted for most (five [71%] of seven) cases of MBWNT that had no somatic CTNNB1 exon 3 mutations. Patients with germline mutations in SUFU and PTCH1 mostly developed infant MBSHH. Germline TP53 mutations presented only in childhood patients in the MBSHH subgroup and explained more than half (eight [57%] of 14) of all chromothripsis events in this subgroup. Germline mutations in PALB2 and BRCA2 were observed across the MBSHH, MBGroup3, and MBGroup4 molecular subgroups and were associated with mutational signatures typical of homologous recombination repair deficiency. In patients with a genetic predisposition to medulloblastoma, 5-year progression-free survival was 52% (95% CI 40-69) and 5-year overall survival was 65% (95% CI 52-81); these survival estimates differed significantly across patients with germline mutations in different medulloblastoma predisposition genes. INTERPRETATION: Genetic counselling and testing should be used as a standard-of-care procedure in patients with MBWNT and MBSHH because these patients have the highest prevalence of damaging germline mutations in known cancer predisposition genes. We propose criteria for routine genetic screening for patients with medulloblastoma based on clinical and molecular tumour characteristics. FUNDING: German Cancer Aid; German Federal Ministry of Education and Research; German Childhood Cancer Foundation (Deutsche Kinderkrebsstiftung); European Research Council; National Institutes of Health; Canadian Institutes for Health Research; German Cancer Research Center; St Jude Comprehensive Cancer Center; American Lebanese Syrian Associated Charities; Swiss National Science Foundation; European Molecular Biology Organization; Cancer Research UK; Hertie Foundation; Alexander and Margaret Stewart Trust; V Foundation for Cancer Research; Sontag Foundation; Musicians Against Childhood Cancer; BC Cancer Foundation; Swedish Council for Health, Working Life and Welfare; Swedish Research Council; Swedish Cancer Society; the Swedish Radiation Protection Authority; Danish Strategic Research Council; Swiss Federal Office of Public Health; Swiss Research Foundation on Mobile Communication; Masaryk University; Ministry of Health of the Czech Republic; Research Council of Norway; Genome Canada; Genome BC; Terry Fox Research Institute; Ontario Institute for Cancer Research; Pediatric Oncology Group of Ontario; The Family of Kathleen Lorette and the Clark H Smith Brain Tumour Centre; Montreal Children's Hospital Foundation; The Hospital for Sick Children: Sonia and Arthur Labatt Brain Tumour Research Centre, Chief of Research Fund, Cancer Genetics Program, Garron Family Cancer Centre, MDT's Garron Family Endowment; BC Childhood Cancer Parents Association; Cure Search Foundation; Pediatric Brain Tumor Foundation; Brainchild; and the Government of Ontario.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Cerebelosas/genética , Metilación de ADN , Pruebas Genéticas/métodos , Mutación de Línea Germinal , Meduloblastoma/genética , Modelos Genéticos , Adolescente , Adulto , Neoplasias Cerebelosas/mortalidad , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/terapia , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Herencia , Humanos , Lactante , Masculino , Meduloblastoma/mortalidad , Meduloblastoma/patología , Meduloblastoma/terapia , Linaje , Fenotipo , Valor Predictivo de las Pruebas , Supervivencia sin Progresión , Estudios Prospectivos , Reproducibilidad de los Resultados , Estudios Retrospectivos , Factores de Riesgo , Transcriptoma , Secuenciación del Exoma , Adulto Joven
9.
World Neurosurg ; 93: 485.e1-5, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27450977

RESUMEN

BACKGROUND: Medulloblastoma is the most common malignant brain tumor in children. Infants are in the high-risk category. Complete surgical resection is the single most important determinant of prognosis and survival in nonmetastatic disease. Infants with large primaries after incomplete resection/biopsy and poor general condition have bad prognosis. They are considered poor candidates for intensive chemotherapy involving high dose methotrexate/autologous stem cell transplantation as they are often unable to tolerate these aggressive regimens. CASE DESCRIPTION: The patient, withinfantile medulloblastoma, was supposed to have complete resection but only a biopsy could be attempted because of increased tumor vascularity. He was in very poor general condition after surgery and his parents declined aggressive chemotherapy and shunt surgery. He was given dose dense neo-adjuvant chemotherapy along with the histone deactylase inhibitor valproate for 5 cycles, with minimal toxicity, after which the tumor was resected. The examination of the resected specimen revealed a complete pathologic response. He then received a total of 18 cycles of chemotherapy and valproate to complete 1 year of systemic treatment. The child is now 6.5 years of age, disease-free, without evidence of any neurocognitive or developmental abnormalities. CONCLUSIONS: We suggest that the role of neoadjuvant chemotherapy should be explored in patients with infantile medulloblastoma in whom upfront complete resection is not possible, considering the gratifying results obtained in our case.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/patología , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/patología , Trastornos del Neurodesarrollo/inducido químicamente , Ácido Valproico/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carboplatino/administración & dosificación , Carboplatino/efectos adversos , Neoplasias Cerebelosas/complicaciones , Niño , Preescolar , Cuidados Críticos/métodos , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Esquema de Medicación , Etopósido/administración & dosificación , Etopósido/efectos adversos , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/efectos adversos , Humanos , Lactante , Estudios Longitudinales , Masculino , Meduloblastoma/complicaciones , Terapia Neoadyuvante/efectos adversos , Terapia Neoadyuvante/métodos , Trastornos Neurocognitivos/inducido químicamente , Trastornos Neurocognitivos/prevención & control , Trastornos del Neurodesarrollo/prevención & control , Resultado del Tratamiento , Ácido Valproico/efectos adversos , Vincristina/administración & dosificación , Vincristina/efectos adversos
10.
Cancer Radiother ; 20(2): 104-8, 2016 Apr.
Artículo en Francés | MEDLINE | ID: mdl-26996790

RESUMEN

PURPOSE: Retrospective analysis of the results of 52 children irradiated for a medulloblastoma. PATIENTS AND METHODS: Between 1974 and 2012, 52 children with an average age of 6 years and a half (11 months-17 years and a half) were treated with surgery then with radiotherapy at the Comprehensive Cancer Centre of Strasbourg (France). For 44 children, the treatment consisted of a chemotherapy. RESULTS: After a mean follow-up of 106.6 months (7-446 months), 13 relapses and 24 deaths were observed. Overall survival at 5 years and 10 years were 62% and 57%, respectively. Disease-free survival at 5 years and 10 years were 80% and 63%, respectively. Univariate analysis found the following adverse prognostic factors: the existence of a postoperative residue, the positivity of the cerebrospinal fluid, the metastatic status and medulloblastoma of high-risk. Positivity of the cerebrospinal fluid remains a negative factor in multivariate analysis. CONCLUSION: These results confirm the survival rate obtained by a conventional approach (surgery then irradiation). Insufficiency of results and rarity of medulloblastoma require the establishment of international protocols.


Asunto(s)
Neoplasias Cerebelosas/mortalidad , Neoplasias Cerebelosas/terapia , Meduloblastoma/mortalidad , Meduloblastoma/terapia , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Cerebelosas/líquido cefalorraquídeo , Neoplasias Cerebelosas/patología , Quimioterapia Adyuvante , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Francia/epidemiología , Humanos , Lactante , Masculino , Meduloblastoma/líquido cefalorraquídeo , Meduloblastoma/patología , Metotrexato/administración & dosificación , Neoplasia Residual/patología , Procarbazina/administración & dosificación , Pronóstico , Radioterapia Adyuvante , Estudios Retrospectivos , Vincristina/administración & dosificación
11.
Cancer Radiother ; 20(1): 14-7, 2016 Feb.
Artículo en Francés | MEDLINE | ID: mdl-26706607

RESUMEN

PURPOSE: Retrospective analysis of the results of 21 adults treated for medulloblastoma. PATIENTS AND METHODS: Between 1978 and 2011, 21 adults with an average age of 31 years (18.3-50) were treated with surgery then with radiotherapy (n=20) at the Comprehensive Cancer Center of Strasbourg. For some (n=12), treatment consisted of chemotherapy. RESULTS: After a mean follow-up of 122 months (19-423), six relapses and seven deaths were observed. Overall survival at 5 years and 10 years was 89.4 ± 7.1% for both. Disease-free survival at 5 years and 10 years was 79.6 ± 9.2% and 85.7 ± 7.6% and 60.6 ± 17.7%, respectively. CONCLUSION: The rarity of medulloblastoma, especially in adults and these results confirm the necessity of international protocols.


Asunto(s)
Neoplasias Cerebelosas/mortalidad , Neoplasias Cerebelosas/terapia , Meduloblastoma/mortalidad , Meduloblastoma/terapia , Adolescente , Adulto , Neoplasias Cerebelosas/patología , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Meduloblastoma/patología , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Radioterapia Adyuvante , Estudios Retrospectivos , Adulto Joven
12.
J Neurooncol ; 121(3): 433-40, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25411097

RESUMEN

Decreasing oxidative damage with the antioxidant agent N-acetylcysteine (NAC) can block the side effects of chemotherapy, but may diminish anti-tumor efficacy. We tested the potential for interactions of high dose NAC against a minimally effective cisplatin chemotherapy regimen in rat models of human pediatric cancers. Athymic rats received subcutaneous implantation of human SK-N-AS neuroblastoma cells or intra-cerebellar implantation of human D283-MED medulloblastoma cells. Rats were untreated or treated with cisplatin (3 or 4 mg/kg IV) with or without NAC (1,000 mg/kg IV) 30 min before or 4 h after cisplatin treatment. Blood urea nitrogen (BUN) and tumor volumes were measured. Cisplatin decreased the growth of SK-N-AS neuroblastoma subcutaneous tumors from 17.7 ± 4.9 to 6.4 ± 2.5 fold over baseline 2 weeks after treatment (P < 0.001). Pretreatment with NAC decreased cisplatin efficacy, while 4 h delayed NAC did not significantly affect cisplatin anti-tumor effects (relative tumor volume 6.8 ± 2.0 fold baseline, P < 0.001). In D283-MED medulloblastoma brain tumors, cisplatin decreased final tumor volume to 3.9 ± 2.3 mm(3) compared to untreated tumor volume of 45.9 ± 38.7 (P = 0.008). Delayed NAC did not significantly alter cisplatin efficacy (tumor volume 6.8 ± 8.1 mm(3), P = 0.014 versus control). Cisplatin was minimally nephrotoxic in these models. NAC decreased cisplatin-induced elevations in BUN (P < 0.02). NAC chemoprotection did not alter cisplatin therapy, if delayed until 4 h after chemotherapy. These data support a Phase I/II clinical trial of delayed NAC to reduce ototoxicity in children with localized pediatric cancers.


Asunto(s)
Acetilcisteína/administración & dosificación , Antineoplásicos/toxicidad , Antioxidantes/administración & dosificación , Cisplatino/toxicidad , Neoplasias Experimentales/tratamiento farmacológico , Animales , Neoplasias Encefálicas/patología , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Meduloblastoma/patología , Trasplante de Neoplasias , Neuroblastoma/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Desnudas
13.
Oncotarget ; 5(16): 7149-61, 2014 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-25216529

RESUMEN

Regardless of the recent advances in cytotoxic therapies, 30% of children diagnosed with medulloblastoma. succumb to the disease. Therefore, novel therapeutic approaches are warranted. Here we demonstrate that Pazopanib a clinically approved multi-kinase angiogenesis inhibitor (MKI) inhibits proliferation and apoptosis in medulloblastoma cell lines. Moreover, Pazopanib profoundly attenuates medulloblastoma cell migration, a prerequisite for tumor invasion and metastasis. In keeping with the observed anti-neoplastic activity of Pazopanib, we also delineate reduced phosphorylation of the STAT3 protein, a key regulator of medulloblastoma proliferation and cell survival. Finally, we document profound in vivo activity of Pazopanib in an orthotopic mouse model of the most aggressive c-myc amplified human medulloblastoma variant. Pazopanib reduced the growth rate of intracranial growing medulloblastoma and significantly prolonged the survival. Furthermore, to put these results into a broader perspective we analysed Pazopanib side by side with the MKI Sorafenib. Both compounds share a similar target profile but display different pharmacodynamics and pharmacokinetics with distinct cytotoxic activity in different tumor entities. Thus, we identified Pazopanib as a new promising candidate for a rational clinical assessment for targeted paediatric medulloblastoma therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Cerebelosas/tratamiento farmacológico , Meduloblastoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Cerebelosas/enzimología , Neoplasias Cerebelosas/patología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Indazoles , Masculino , Meduloblastoma/enzimología , Meduloblastoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Niacinamida/administración & dosificación , Niacinamida/análogos & derivados , Niacinamida/farmacología , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Sorafenib , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Mol Cancer Res ; 12(5): 694-702, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24505127

RESUMEN

UNLABELLED: Targeting amino acid metabolism has therapeutic implications for aggressive brain tumors. Asparagine is an amino acid that is synthesized by normal cells. However, some cancer cells lack asparagine synthetase (ASNS), the key enzyme for asparagine synthesis. Asparaginase (ASNase) contributes to eradication of acute leukemia by decreasing asparagine levels in serum and cerebrospinal fluid. However, leukemic cells may become ASNase-resistant by upregulating ASNS. High expression of ASNS has also been associated with biologic aggressiveness of other cancers, including gliomas. Here, the impact of enzymatic depletion of asparagine on proliferation of brain tumor cells was determined. ASNase was used as monotherapy or in combination with conventional chemotherapeutic agents. Viability assays for ASNase-treated cells demonstrated significant growth reduction in multiple cell lines. This effect was reversed by glutamine in a dose-dependent manner--as expected, because glutamine is the main amino group donor for asparagine synthesis. ASNase treatment also reduced sphere formation by medulloblastoma and primary glioblastoma cells. ASNase-resistant glioblastoma cells exhibited elevated levels of ASNS mRNA. ASNase cotreatment significantly enhanced gemcitabine or etoposide cytotoxicity against glioblastoma cells. Xenograft tumors in vivo showed no significant response to ASNase monotherapy and little response to temozolomide alone. However, combinatorial therapy with ASNase and temozolomide resulted in significant growth suppression for an extended duration of time. Taken together, these findings indicate that amino acid depletion warrants further investigation as adjunctive therapy for brain tumors. IMPLICATIONS: Findings have potential impact for providing adjuvant means to enhance brain tumor chemotherapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Asparaginasa/farmacología , Asparagina/deficiencia , Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Animales , Asparaginasa/administración & dosificación , Asparaginasa/metabolismo , Asparagina/metabolismo , Aspartatoamoníaco Ligasa/metabolismo , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Daño del ADN , Dacarbazina/administración & dosificación , Dacarbazina/farmacología , Sinergismo Farmacológico , Glioblastoma/tratamiento farmacológico , Glioblastoma/enzimología , Glioblastoma/metabolismo , Glioblastoma/patología , Glutamina/farmacología , Humanos , Masculino , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/enzimología , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Ratones Desnudos , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Res ; 73(23): 7034-42, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24154871

RESUMEN

Medulloblastoma is a cancer of the cerebellum, for which there is currently no approved targeted therapy. Recent transcriptomics approaches have demonstrated that medulloblastoma is composed of molecularly distinct subgroups, one of which is characterized by activation of the Hedgehog pathway, which in mouse models is sufficient to drive medulloblastoma development. There is thus considerable interest in targeting the Hedgehog pathway for therapeutic benefit in medulloblastoma, particularly given the recent approval of the Hedgehog pathway inhibitor vismodegib for metastatic and locally advanced basal cell carcinoma. Like other molecularly targeted therapies, however, there have been reports of acquired resistance to vismodegib, driven by secondary Hedgehog pathway mutations and potentially by activation of the phosphatidylinositol 3-kinase (PI3K) pathway. Given that acquired resistance to vismodegib may occur as a result of inappropriate PI3K pathway activation, we asked if loss of the PI3K pathway regulator, phosphatase and tensin homologue (Pten), which has been reported to occur in patients within the Hedgehog subgroup, would constitute a mechanism of innate resistance to vismodegib in Hedgehog-driven medulloblastoma. We find that Hedgehog pathway inhibition successfully restrains growth of Pten-deficient medulloblastoma in this mouse model, but does not drive tumor regression, as it does in Pten-wild-type medulloblastoma. Combined inhibition of the Hedgehog and PI3K pathways may lead to superior antitumor activity in PTEN-deficient medulloblastoma in the clinic.


Asunto(s)
Anilidas/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Cerebelosas/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Meduloblastoma/tratamiento farmacológico , Fosfohidrolasa PTEN/fisiología , Piridinas/uso terapéutico , Anilidas/farmacología , Animales , Antineoplásicos/farmacología , Transformación Celular Neoplásica/genética , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Evaluación Preclínica de Medicamentos , Femenino , Eliminación de Gen , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/genética , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Embarazo , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
16.
J Exp Clin Cancer Res ; 32: 27, 2013 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-23672687

RESUMEN

BACKGROUND: Medulloblastoma (MB) is the most common malignant brain tumor in childhood with a 5-year survival of approximately 60%. We have recently shown that treatment of human MB cells with 5-aza-2'-deoxycytidine (5-aza-dC) reduces the clonogenic survival significantly. Here, we tested combinatorial effects of 5-aza-dC with other epigenetic (valproic acid, SAHA) and differentiation-inducing drugs (resveratrol, abacavir, retinoic acid) on human MB cells in vitro to intensify the antitumor therapy further. METHODS: Three human MB cell lines were treated with 5-aza-dC alone or in combination for three or six days. Metabolic activity was measured by WST-1 assay. To determine long-term reproductive survival, clonogenic assays were performed. Induction of DNA double-strand break (DSB) repair was measured by γH2AX assay. RESULTS: The applied single drugs, except for ATRA, reduced the metabolic activity dose-dependently in all MB cell lines. Longer treatment times enhanced the reduction of metabolic activity by 5-aza-dC. Combinatorial treatments showed differential, cell line-dependent responses indicating an important impact of the genetic background. 5-Aza-dC together with resveratrol was found to exert the most significant inhibitory effects on metabolic activity in all cell lines. 5-aza-dC alone reduced the clonogenicity of MB cells significantly and induced DSB with no further changes after adjuvant administration of resveratrol. CONCLUSION: The observed significant decrease in metabolic activity by combinatorial treatment of MB cells with 5-aza-dC and resveratrol does not translate into long-term reproductive survival deficiency in vitro. Further studies in animal models are needed to clarify the resveratrol-mediated anticancer mechanisms in vivo.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Epigénesis Genética/efectos de los fármacos , Meduloblastoma/genética , Meduloblastoma/patología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Cerebelosas/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Decitabina , Relación Dosis-Respuesta a Droga , Humanos , Meduloblastoma/metabolismo , Clasificación del Tumor , Tretinoina/farmacología , Ensayo de Tumor de Célula Madre , Ácido Valproico/farmacología
17.
Anticancer Res ; 32(7): 2689-96, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22753727

RESUMEN

BACKGROUND/AIM: Zeng Sheng Ping (ZSP) is a traditional herbal remedy used to prevent progression and growth of neoplastic lesions. It has been shown to inhibit Notch2 expression in a murine lung cancer model, leading us to investigate its therapeutic potential in Notch-dependent brain tumors. MATERIALS AND METHODS: 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS), apoptosis, and quantitative real-time polymerase chain reaction (RT-PCR) analyses were performed in glioma and medulloblastoma cell lines, and morphological analyses in DAOY flank xenografts. RESULTS: ZSP inhibited brain tumor growth in vitro, in part, by apoptotic induction. Down-regulation of the Notch2 receptor, the pathway target Hairy/Enhancer of Split homolog 1 (Hes1), and of the stem cell markers Nestin and CD133 was also observed. Reductions in tumor mass and increases in the necrotic fraction of DAOY xenografts, in mice treated with oral ZSP were also observed, but these were not significant. CONCLUSION: ZSP can block brain tumor growth and the expression of Notch pathway members and stem cell markers in vitro.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Receptores Notch/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Ratones Desnudos , Fitoterapia , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
PLoS One ; 6(6): e21370, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21712951

RESUMEN

Platelet-derived growth factor receptors (PDGFRs) have been implicated in a wide array of human malignancies, including medulloblastoma (MB), the most common brain tumor of childhood. Although significant progress in MB biology and therapeutics has been achieved during the past decades, MB remains a horrible challenge to the physicians and researchers. Therefore, novel inhibitors targeting PDGFR signaling pathway may offer great promise for the treatment of MB. In the present study, we investigated the cytotoxicity and mechanisms of cambogin in Daoy MB cells. Our results show that cambogin triggers significant S phase cell cycle arrest and apoptosis via down regulation of cyclin A and E, and activation of caspases. More importantly, further mechanistic studies demonstrated that cambogin inhibits PDGFR signaling in Daoy and genetically defined mouse embryo fibroblast (MEF) cell lines. These results suggest that cambogin is preferentially cytotoxic to cells expressing PDGFR. Our findings may provide a novel approach by targeting PDGFR signaling against MB.


Asunto(s)
Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Fase S/efectos de los fármacos , Terpenos/toxicidad , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Ciclina A/metabolismo , Ciclina E/metabolismo , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Ratones Noqueados , Estructura Molecular , Extractos Vegetales/química , Extractos Vegetales/toxicidad , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/efectos de los fármacos , Terpenos/química
20.
J Neuropathol Exp Neurol ; 70(6): 430-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21572341

RESUMEN

To determine whether the zinc finger transcription factors GLI1 to GLI3 and suppressor of fused (SUFU) components of the Sonic hedgehog signaling pathway may be prognostic markers and potential therapeutic targets in pediatric medulloblastoma (MB), we investigated the relationship of the expression of these proteins to prognosis in the MB of 124 patients who had undergone surgery at the Hospital for Sick Children (Toronto, Ontario, Canada). The expressions of GLI1 (p = 0.011) and GLI2 (p = 0.003), but not of GLI3 (p = 0.774) or SUFU (p = 0.137), in the MB were associated with a worse overall survival by Kaplan-Meier analysis. Overall survival of patients positive for GLI1 and GLI2 was 6.01 ± 0.85 years and 5.27 ± 1.44 years, respectively, versus 10.11 ± 1.52 years and 10.18 ± 0.22 years for patients negative for GLI1 and GLI2, respectively. Knockdown of GLI2 in 3 MB cell lines resulted in decreased cell number and viability, as determined by the MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay; knockdown of GLI1 had no effect. The decrease in cell number with GLI2 knockdown was caused by G0 cell cycle arrest; there was no induction of apoptosis. These results suggest that targeting the Sonic hedgehog pathway in positive patients may be a useful adjuvant therapeutic strategy for MB.


Asunto(s)
Apoptosis/fisiología , Neoplasias Cerebelosas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Meduloblastoma/metabolismo , Proteínas Nucleares/metabolismo , Adolescente , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Cerebelosas/patología , Niño , Preescolar , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Etiquetado Corte-Fin in Situ/métodos , Lactante , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Meduloblastoma/patología , Proteínas Nucleares/genética , Pediatría , ARN Interferente Pequeño/farmacología , Estudios Retrospectivos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transducción Genética/métodos , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA