Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Radiother Oncol ; 196: 110268, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38641261

RESUMEN

BACKGROUND AND PURPOSE: Multiple studies demonstrated hypothalamic-pituitary dysfunction in survivors of pediatric brain tumors. However, few studies investigated the trajectories of pituitary height in these patients and their associations with pituitary function. We aimed to evaluate longitudinal changes of pituitary height in children and adolescents with brain tumors, and their association with endocrine deficiencies. MATERIALS AND METHODS: We conducted a retrospective analysis of 193 pediatric patients (54.9% male) diagnosed with brain tumors from 2002 to 2018, with a minimum of two years of radiological follow-up. Pituitary height was measured using MRI scans at diagnosis and at 2, 5, and 10 years post-diagnosis, with clinical data sourced from patient charts. RESULTS: Average age at diagnosis was 7.6 ± 4.5 years, with a follow-up of 6.1 ± 3.4 years. 52.8% underwent radiotherapy and 37.8% experienced pituitary hormone deficiency. Radiation treatment was a significant predictor of decreased pituitary height at all observed time points (p = 0.016, p < 0.001, p = 0.008, respectively). Additionally, chemotherapy (p = 0.004) or radiotherapy (p = 0.022) history and pituitary height at 10 years (p = 0.047) were predictors of endocrine deficiencies. ANOVA revealed an expected increase in pituitary height over time in pediatric patients, but this growth was significantly impacted by radiation treatment and gender (p for interaction = 0.005 and 0.025, respectively). CONCLUSION: Cranial irradiation in pediatric patients is associated with impairment of the physiologic increase in pituitary size; in turn, decreased pituitary height is associated with endocrine dysfunction. We suggest that pituitary gland should be evaluated on surveillance imaging of pediatric brain tumor survivors, and if small for age, clinical endocrine evaluation should be pursued.


Asunto(s)
Neoplasias Encefálicas , Humanos , Niño , Masculino , Femenino , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/diagnóstico por imagen , Estudios Retrospectivos , Adolescente , Traumatismos por Radiación/etiología , Traumatismos por Radiación/diagnóstico por imagen , Hipófisis/efectos de la radiación , Hipófisis/diagnóstico por imagen , Preescolar , Imagen por Resonancia Magnética , Irradiación Craneana/efectos adversos , Hipopituitarismo/etiología
2.
Cancers (Basel) ; 15(3)2023 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-36765532

RESUMEN

Personalized vaccines against patient-unique tumor-associated antigens represent a promising new approach for cancer immunotherapy. Vaccine efficacy is assessed by quantification of changes in the frequency and/or the activity of antigen-specific T cells. Enzyme-linked immunosorbent spot (ELISpot) and flow cytometry (FCM) are methodologies frequently used for assessing vaccine efficacy. We tested these methodologies and found that both ELISpot and standard FCM [monitoring CD3/CD4/CD8/IFNγ/Viability+CD14+CD19 (dump)] demonstrate background IFNγ secretion, which, in many cases, was higher than the antigen-specific signal measured by the respective methodology (frequently ranging around 0.05-0.2%). To detect such weak T-cell responses, we developed an FCM panel that included two early activation markers, 4-1BB (CD137) and CD40L (CD154), in addition to the above-cited markers. These two activation markers have a close to zero background expression and are rapidly upregulated following antigen-specific activation. They enabled the quantification of rare T cells responding to antigens within the assay well. Background IFNγ-positive CD4 T cell frequencies decreased to 0.019% ± 0.028% and CD8 T cells to 0.009% ± 0.013%, which are 19 and 13 times lower, respectively, than without the use of these markers. The presented methodology enables highly sensitive monitoring of T-cell responses to tumor-associated antigens in the very low, but clinically relevant, frequencies.

3.
Neuro Oncol ; 25(7): 1345-1354, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-36633935

RESUMEN

BACKGROUND: Endocrine deficiencies, including hypothalamic-pituitary-gonadal axis (HPGA) impairment, are common in survivors of childhood and adolescent medulloblastoma. Still, data regarding pubertal development and fecundity are limited, and few studies assessed HPGA function in males. We aimed to describe HPGA function in a large cohort of patients with medulloblastoma. METHODS: A retrospective study comprising all 62 medulloblastoma patients treated in our center between 1987 and 2021, who were at least 2 years from completion of therapy. HPGA function was assessed based on clinical data, biochemical markers, and questionnaires. RESULTS: Overall, 76% of female patients had clinical or biochemical evidence of HPGA dysfunction. Biochemical evidence of diminished ovarian reserve was seen in all prepubertal girls (n = 4). Among the males, 34% had clinical or biochemical evidence of gonadal dysfunction, 34% had normal function, and 29% were age-appropriately clinically and biochemically prepubertal. The difference between males and females was significant (P = .003). Cyclophosphamide-equivalent dose was significantly associated with HPGA function in females, but not in males. There was no association between HPGA dysfunction and other endocrine deficiencies, length of follow-up, weight status, and radiation treatment protocol. Two female and 2 male patients achieved successful pregnancies, resulting in 6 live births. CONCLUSIONS: HPGA dysfunction is common after treatment for childhood medulloblastoma. This is seen more in females, likely due to damage to the ovaries from spinal radiotherapy. Our findings may assist in counseling patients and their families regarding risk to future fertility and need for fertility preservation.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Embarazo , Adolescente , Humanos , Masculino , Femenino , Meduloblastoma/radioterapia , Estudios Retrospectivos , Sobrevivientes , Neoplasias Cerebelosas/radioterapia , Fertilidad
4.
Front Oncol ; 12: 959133, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36091130

RESUMEN

We found that pediatric glioblastoma (PED-GBM) cell lines from diffuse intrinsic pontine glioma (DIPG) carrying the H3K27M mutation or from diffuse hemispheric glioma expressing the H3G34R mutation are sensitive to the combination of vorinostat (a histone deacetylase inhibitor) and PARP-1 inhibitors. The combined treatment increased the phosphorylation of eIF2α (P-eIF2α) relative to each drug alone and enhanced the decrease in cell survival. To explore the role played by increased P-eIF2α in modulating PED-GBM survival and response to treatments, we employed brain-penetrating inhibitors of P-eIF2α dephosphorylation: salubrinal and raphin-1. These drugs increased P-eIF2α, DNA damage, and cell death, similarly affecting the sensitivity of DIPG cells and derived neurospheres to PARP-1 inhibitors. Interestingly, these drugs also decreased the level of eIF2Bϵ (the catalytic subunit of eIF2B) and increased its phosphorylation, thereby enhancing the effect of increased P-eIF2α. Transient transfection with the S51D phosphomimetic eIF2α variant recapitulated the effect of salubrinal and raphin-1 on PED-GBM survival and sensitivity to PARP-1 inhibitors. Importantly, either salubrinal or raphin-1 dramatically increased the sensitivity of DIPG cells to radiation, the main treatment modality of PED-GBM. Finally, PED-GBM was more sensitive than normal human astrocytes to salubrinal, raphin-1, and the treatment combinations described herein. Our results indicate that combinations of histone deacetylase inhibitors and PARP-1 inhibitors should be evaluated for their toxicity and efficacy in PED-GBM patients and point to drugs that increase P-eIF2α or modulate its downstream effectors as a novel means of treating PED-GBM.

5.
Acta Derm Venereol ; 101(7): adv00501, 2021 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-34230982

RESUMEN

Targeted medications and immunotherapies are being developed to specifically target the pathways involved in tumours. There is limited experience with these new medications and their cutaneous side-effects in the paediatric population. A retrospective study of all paediatric oncological patients treated with targeted therapies and immunotherapies between 1 January 2013 and 1 August 2020 was carried out in 2 haemato-oncological referral centres. A total of 103 children were included in the study. The median (interquartile range) age was 13 years (8.4-16.9), male:female ratio 1.5:1, median (interquartile range) follow-up was 7 months (2-18). Fifty (48%) of the children developed cutaneous adverse events. Treatment was discontinued in only 3 (6%) cases and was altered in only (2%) 1 case due to a cutaneous adverse event. When targeted therapies and immunotherapies for tumours in children are used, there is an increased incidence of cutaneous adverse events. Nevertheless, treatment modification or discontinuation due to cutaneous side-effects is rarely needed.


Asunto(s)
Terapia Molecular Dirigida , Piel , Niño , Femenino , Humanos , Inmunoterapia/efectos adversos , Lactante , Masculino , Terapia Molecular Dirigida/efectos adversos , Derivación y Consulta , Estudios Retrospectivos
6.
Childs Nerv Syst ; 37(5): 1449-1459, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33821340

RESUMEN

Traditional management of newly diagnosed pediatric brain tumors (PBTs) consists of cranial imaging, typically magnetic resonance imaging (MRI), and is frequently followed by tissue diagnosis, through either surgical biopsy or tumor resection. Therapy regimes are typically dependent on histological diagnosis. To date, many treatment regimens are based on molecular biology. The scope of this article is to discuss the role of diagnosis and further treatment of PBTs based solely on MRI features, in light of the latest treatment protocols. Typical MRI findings and indications for surgical biopsy of these lesions are described.


Asunto(s)
Neoplasias Encefálicas , Biopsia , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/terapia , Niño , Humanos , Imagen por Resonancia Magnética
7.
Acta Neuropathol ; 140(5): 765-776, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32895736

RESUMEN

Replication repair deficiency (RRD) leading to hypermutation is an important driving mechanism of high-grade glioma (HGG) occurring predominantly in the context of germline mutations in RRD-associated genes. Although HGG presents specific patterns of DNA methylation corresponding to oncogenic mutations, this has not been well studied in replication repair-deficient tumors. We analyzed 51 HGG arising in the background of gene mutations in RRD utilizing either 450 k or 850 k methylation arrays. These were compared with HGG not known to be from patients with RRD. RRD HGG harboring secondary mutations in glioma genes such as IDH1 and H3F3A displayed a methylation pattern corresponding to these methylation subgroups. Strikingly, RRD HGG lacking these known secondary mutations clustered together with an incompletely described group of HGG previously labeled "Wild type-C" or "Paediatric RTK 1". Independent analysis of two comparator HGG cohorts showed that other RRD/hypermutant tumors clustered within these subgroups, suggesting that undiagnosed RRD may be driving some HGG clustering in this location. RRD HGG displayed a unique CpG Island Demethylator Phenotype in contrast to the CpG Island Methylator Phenotype described in other cancers. Hypomethylation was enriched at gene promoters with prominent demethylation in genes and pathways critical to cellular survival including cell cycle, gene expression, cellular metabolism, and organization. These data suggest that methylation arrays may provide diagnostic information for the detection of RRD HGG. Furthermore, our findings highlight the unique natural selection pressures in these highly dysregulated, hypermutant cancers and provide the novel impact of hypermutation and RRD on the cancer epigenome.


Asunto(s)
Neoplasias Encefálicas/genética , Metilación de ADN/genética , Trastornos por Deficiencias en la Reparación del ADN/genética , Reparación del ADN/genética , Glioma/genética , Adolescente , Adulto , Niño , Preescolar , Trastornos por Deficiencias en la Reparación del ADN/complicaciones , Femenino , Mutación de Línea Germinal , Humanos , Masculino , Adulto Joven
8.
Front Oncol ; 9: 327, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31114757

RESUMEN

Pediatric brain tumors are the most common solid tumor type and the leading cause of cancer-related death in children. The immune system plays an important role in cancer pathogenesis and in the response to immunotherapy treatments. T lymphocytes are key elements for the response of the immune system to cancer cells and have been associated with prognosis of different cancers. Neutrophils on the other hand, which secrete pro-angiogenic and anti-apoptotic factors, enhance the ability of tumor cells to grow and develop into metastases. We conducted a retrospective study of 120 pediatric brain cancer patients and 171 elective pediatric patients hospitalized in Dana Children's Hospital and Sheba Medical Center. Data on age, sex, treatment, lymphocyte, neutrophil, and monocyte count were collected from routinely performed preoperative blood tests. Neutrophil-to-lymphocyte ratio (NLR), and the lymphocyte-to-monocyte ratio (LMR) were calculated and significance was determined by paired T test. p < 0.05 was considered as statistically significant. NLR was significantly higher in the pediatric brain cancer patients. The high NLR in pediatric brain cancer patients is the result of a combination of low lymphocytes and high neutrophils. Both of these factors can have a role in cancer development and propagation and also in response to therapy.

9.
Acta Haematol ; 141(2): 119-127, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30726835

RESUMEN

BACKGROUND: The WHO defined myeloid and lymphoid neoplasms (MLN) with eosinophilia associated with PDGFRB, PDGFRA, FGFR1 rearrangements as a new entity in 2016. PDGFRB-rearranged MLN sensitive to imatinib were described in adult patients. We report the first pediatric patient with PDGFRB-rearranged myeloproliferative disorder associated with T-lymphoblastic lymphoma bearing the t(5; 14)(q33;q32) translocation who was successfully treated with imatinib only. Methods/Aims: Analysis of bone marrow and peripheral blood cells by fluorescent in situ hybridization identified the PDGFRB partner as CCDC88C. Whole genome sequencing of the patient's DNA identified the exact junction site, confirmed by PCR amplification and Sanger sequencing. A real-time quantitative PCR assay was designed to quantify the fused CCDC88C-PDGFRB product. RESULTS: A 2.5-year-old boy was diagnosed with myeloproliferative disorder and eosinophilia associated with lymphoblastic lymphoma both bearing the CCDC88C-PDGFRB fusion. Imatinib therapy resulted in rapid clinical, hematological, and cytogenetic response. Molecular response to treatment was monitored by a real-time PCR assay specific for the CCDC88C- PDGFRB fusion. CONCLUSION: This is the first description of MLN with eosinophilia in the pediatric age group. Response to treatment with imatinib only was monitored by specific quantitative PCR assay with sustained remission lasting 5.5 years from diagnosis.


Asunto(s)
Mesilato de Imatinib/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de Microfilamentos/genética , Trastornos Mieloproliferativos/tratamiento farmacológico , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Secuencia de Bases , Preescolar , Humanos , Hibridación Fluorescente in Situ , Cariotipo , Masculino , Trastornos Mieloproliferativos/complicaciones , Trastornos Mieloproliferativos/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Secuenciación Completa del Genoma
10.
Neuroradiology ; 60(9): 913-919, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30030548

RESUMEN

PURPOSE: The aim of this retrospective study is to investigate the value of the susceptibility-weighted imaging (SWI) sequence compared to gradient echo (GRE) in the detection and follow-up of cavernous malformations in patients who underwent whole-brain irradiation as part of their medulloblastoma treatment. METHODS: We retrospectively examined MRI studies of 28 subjects (16 males, 12 females) who received whole-brain irradiation as part of their treatment. Ages at irradiation ranged from 2 to 38 years. All patients were periodically followed up with MR imaging (ranging from 9 to 336 months). Two neuroradiologists reviewed studies of the same patients, comparing the number of suspected cavernomas detected on GRE and SWI sequences performed at different times (median time between studies, 10 months). RESULTS: Hypointense lesions were detected in 24 subjects on SWI sequences and in 19 subjects on GRE sequences. More lesions were seen on SWI than on GRE (p = 0.006). Four patients had no detectable lesions. The minimal period from irradiation to first lesion detection was 14 months. Cavernomas larger than 3 mm were detected in 14 subjects by both GRE and SWI. None of the subjects had symptoms related to cavernomas. CONCLUSIONS: The sensitivity of SWI in the detection of hypointense lesions in patients after whole-brain irradiation is significantly higher than that of the GRE sequence. It appears that almost all subjects eventually develop small hypointense lesions after radiotherapy, and some of them progress to cavernous malformations. The clinical significance of the increased sensitivity of SWI in this group of patients is not entirely certain.


Asunto(s)
Hemangioma Cavernoso del Sistema Nervioso Central/diagnóstico por imagen , Hemangioma Cavernoso del Sistema Nervioso Central/etiología , Meduloblastoma/diagnóstico por imagen , Meduloblastoma/radioterapia , Tumores Neuroectodérmicos Primitivos/diagnóstico por imagen , Tumores Neuroectodérmicos Primitivos/radioterapia , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Sensibilidad y Especificidad
11.
Oncotarget ; 9(44): 27547-27563, 2018 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-29938005

RESUMEN

Glioblastoma, a fatal disease in both adult and pediatric patients, currently has limited treatment options that offer no more than temporary relief. Our experiments with adult and pediatric glioblastoma cell lines showed that radiation induces a dose-dependent increase in the level of MutT homolog 1 (MTH1) - an enzyme that hydrolyzes oxidized purine nucleoside triphosphates. Similarly, the combination of vorinostat, which is a histone deacetylase inhibitor, and ABT-888, which is a PARP-1 inhibitor, enhanced clonogenic death and increased the MTH1 level, relative to each treatment alone. This result suggests that the MTH1 level is directly related to the damage that is inflicted upon the cells, and its activity protects them against anti-neoplastic therapy. Indeed, the MTH1 inhibitor TH588 and MTH1 siRNA increased glioblastoma's response to both radiation and the combination of vorinostat and ABT-888. TH588 also inhibited glioblastoma's capacity for migration and invasion. In normal fibroblasts, low radiation doses and the combination of vorinostat and ABT-888 decreased the level of the enzyme. TH588 did not alter the fibroblasts' response to radiation and only mildly affected their response to the combination of vorinostat and ABT-888. In summary, the inhibition of MTH1 is required to better realize the therapeutic potential of anti-neoplastic treatments in glioblastoma.

12.
Cell Res ; 28(2): 187-203, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29327725

RESUMEN

Neural progenitor cells undergo somatic retrotransposition events, mainly involving L1 elements, which can be potentially deleterious. Here, we analyze the whole genomes of 20 brain samples and 80 non-brain samples, and characterized the retrotransposition landscape of patients affected by a variety of neurodevelopmental disorders including Rett syndrome, tuberous sclerosis, ataxia-telangiectasia and autism. We report that the number of retrotranspositions in brain tissues is higher than that observed in non-brain samples and even higher in pathologic vs normal brains. The majority of somatic brain retrotransposons integrate into pre-existing repetitive elements, preferentially A/T rich L1 sequences, resulting in nested insertions. Our findings document the fingerprints of encoded endonuclease independent mechanisms in the majority of L1 brain insertion events. The insertions are "non-classical" in that they are truncated at both ends, integrate in the same orientation as the host element, and their target sequences are enriched with a CCATT motif in contrast to the classical endonuclease motif of most other retrotranspositions. We show that L1Hs elements integrate preferentially into genes associated with neural functions and diseases. We propose that pre-existing retrotransposons act as "lightning rods" for novel insertions, which may give fine modulation of gene expression while safeguarding from deleterious events. Overwhelmingly uncontrolled retrotransposition may breach this safeguard mechanism and increase the risk of harmful mutagenesis in neurodevelopmental disorders.


Asunto(s)
Encéfalo/fisiopatología , Elementos de Nucleótido Esparcido Largo/genética , Trastornos del Neurodesarrollo/genética , Nucleótidos de Adenina/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Daño del ADN , Bases de Datos Genéticas , Endonucleasas/genética , Exones , Regulación de la Expresión Génica , Genes/genética , Genómica/métodos , Humanos , MicroARNs/genética , Mutación , Neuronas/metabolismo , Estadísticas no Paramétricas , Nucleótidos de Timina/genética , Secuenciación Completa del Genoma
13.
J Neurooncol ; 136(1): 189-195, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29143272

RESUMEN

Diffuse intrinsic pontine glioma (DIPG) is an incurable disease with a median overall survival of 10 months. Immune modulating antibodies have recently emerged as a highly promising treatment modality in multiple cancer types. We present results from the first study to evaluate the immune modulating antibody MDV9300 (pidilizumab) in pediatric patients with DIPG. All patients aged 3 years and older, diagnosed with DIPG between February 2014 and June 2015 in Israel, were offered to participate in the study. Enrolled patients were started on biweekly 6 mg/kg MDV9300 after radiation completion. Treatment was continued until disease progression on imaging. Patients were followed biweekly for the occurrence of neurological deficit toxicities and side effects. Secondary endpoints were event free survival and overall survival. Of 13 children diagnosed with DIPG during the study period, nine were enrolled in the study. The patients underwent radiotherapy and none had chemotherapy. A total of 83 cycles of MDV9300 (range 2-16) were applied. The main side effects were neutropenia (CTCAE grade 1-3), mild to moderate fatigue, and acute elevation of blood pressure. Four patients died within 1 year of the diagnosis, another three died within 2 years and two children are still alive nearly 30 months from diagnosis, with stable disease. The median event free survival is 9.3 months (range 6.8-24) and the median overall survival is 15.6 months (range 6.9-28). Preliminary results demonstrate that MDV9300 treatment is safe and may be effective in the treatment of children with DIPG.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Glioma/tratamiento farmacológico , Adolescente , Neoplasias del Tronco Encefálico/inmunología , Neoplasias del Tronco Encefálico/radioterapia , Niño , Preescolar , Terapia Combinada , Progresión de la Enfermedad , Femenino , Glioma/inmunología , Glioma/radioterapia , Humanos , Masculino , Supervivencia sin Progresión , Estudios Prospectivos , Análisis de Supervivencia
14.
Cell ; 171(5): 1042-1056.e10, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29056344

RESUMEN

We present an extensive assessment of mutation burden through sequencing analysis of >81,000 tumors from pediatric and adult patients, including tumors with hypermutation caused by chemotherapy, carcinogens, or germline alterations. Hypermutation was detected in tumor types not previously associated with high mutation burden. Replication repair deficiency was a major contributing factor. We uncovered new driver mutations in the replication-repair-associated DNA polymerases and a distinct impact of microsatellite instability and replication repair deficiency on the scale of mutation load. Unbiased clustering, based on mutational context, revealed clinically relevant subgroups regardless of the tumors' tissue of origin, highlighting similarities in evolutionary dynamics leading to hypermutation. Mutagens, such as UV light, were implicated in unexpected cancers, including sarcomas and lung tumors. The order of mutational signatures identified previous treatment and germline replication repair deficiency, which improved management of patients and families. These data will inform tumor classification, genetic testing, and clinical trial design.


Asunto(s)
Neoplasias/genética , Adulto , Niño , Análisis por Conglomerados , ADN Polimerasa II/genética , ADN Polimerasa III/genética , Replicación del ADN , Humanos , Mutación , Neoplasias/clasificación , Neoplasias/patología , Neoplasias/terapia , Proteínas de Unión a Poli-ADP-Ribosa/genética
15.
Childs Nerv Syst ; 33(12): 2169-2175, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28808787

RESUMEN

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


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/terapia , Tratamiento Conservador/tendencias , Glioblastoma/diagnóstico por imagen , Glioblastoma/terapia , Hallazgos Incidentales , Niño , Tratamiento Conservador/métodos , Progresión de la Enfermedad , Humanos , Masculino , Resultado del Tratamiento
16.
Oncotarget ; 7(46): 74860-74871, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27556862

RESUMEN

Temozolomide (TMZ) is an alkylating agent that has become the mainstay treatment of the most malignant brain cancer, glioblastoma multiforme (GBM). Unfortunately only a limited number of patients positively respond to it. It has been shown that zinc metal reestablishes chemosensitivity but this effect has not been tested with TMZ. Using both in vitro and in vivo experimental approaches, we investigated whether addition of zinc to TMZ enhances its cytotoxicity against GBM. In vitro cell viability analysis showed that the cytotoxic activity of TMZ was substantially increased with addition of zinc and this response was accompanied by an elevation of p21, PUMA, BAX and Caspase-3 expression and a decrease in growth fraction as manifested by low ki67 and lower colony formation. Analysis of GBM as intracranial xenografts in athymic mice and administration of concurrent TMZ and zinc yielded results consistent with those of the in vitro analyses. The co-treatment resulted in significant reduction in tumor volume in TMZ/zinc treated mice relative to treatment with TMZ alone. Our results suggest that zinc may serve as a potentiator of TMZ therapy in GBM patients.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/patología , Dacarbazina/análogos & derivados , Glioblastoma/patología , Zinc/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Caspasa 3/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Dacarbazina/farmacología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Ratones , Temozolomida , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
18.
J Neurooncol ; 129(3): 453-460, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27377654

RESUMEN

Patients with progressive primary brain tumors (PBT) are attracted to promising new treatments, even prior to convincing data. Anti-PD1 immunotherapies have been in the spotlight since publication of groundbreaking results for metastatic melanoma with pembrolizumab (PBL). Our objective was to report on the response and toxicity of PBL in patients with advanced PBT. We retrospectively reviewed the charts of 22 patients (17 adults and 5 children) with recurrent central nervous system tumors treated with PBL. We analyzed prior antineoplastic therapies, steroid usage, and outcomes. Patients received a median of two neoplastic therapies prior to PBL, and a median of three infusions of PBL in adults and four in children. Twelve patients (9 adults and 3 children) started PBL on steroids (median dose in adults 4 mg; range 2-8, and in children 1.5 mg, range 0.5-4) and five patients received steroids later during PBL treatment. Twelve patients (10 adults and 2 children) received concomitant bevacizumab with PBL. Side effects were minimal. All patients showed progressive tumor growth during therapy. Median OS from the start of PBL was 2.6 months in adults and 3.2 months in children. Two GB patients underwent tumor resection following treatment with PBL. Tumor-lymphocytic response in these cases was unremarkable, and PD-L1 immuno-staining was negative. In this series of 22 patients with recurrent primary brain tumors, PBL showed no clinical or histologic efficacy. We do not recommend further use of PBL for recurrent PBT unless convincing prospective clinical trial data are published.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Resultado del Tratamiento , Adulto , Anciano , Neoplasias del Sistema Nervioso Central/diagnóstico por imagen , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad
19.
PLoS One ; 11(5): e0155711, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27196668

RESUMEN

Inhibitors of poly[ADP-ribose] polymerase 1 (PARPis) show promise for treatment of cancers which lack capacity for homologous recombination repair (HRR). However, new therapeutic strategies are required in order to overcome innate and acquired resistance to these drugs and thus expand the array of cancers that could benefit from them. We show that human cancer cell lines which respond poorly to ABT-888 (a PARPi), become sensitive to it when co-treated with vorinostat (a histone deacetylase inhibitor (HDACi)). Vorinostat also sensitized PARPis insensitive cancer cell lines to 6-thioguanine (6-TG)-a drug that targets PARPis sensitive cells. The sensitizing effect of vorinostat was associated with increased phosphorylation of eukaryotic initiation factor (eIF) 2α which in and of itself increases the sensitivity of cancer cells to ABT-888. Importantly, these drug combinations did not affect survival of normal fibroblasts and breast cells, and significantly increased the inhibition of xenograft tumor growth relative to each drug alone, without affecting the mice weight or their liver and kidney function. Our results show that combination of vorinostat and ABT-888 could potentially prove useful for treatment of cancer with innate resistance to PARPis due to active HRR machinery, while the combination of vorinostat and 6-TG could potentially overcome innate or acquired resistance to PARPis due to secondary or reversal BRCA mutations, to decreased PARP-1 level or to increased expression of multiple drug resistant proteins. Importantly, drugs which increase phosphorylation of eIF2α may mimic the sensitizing effect of vorinostat on cellular response to PARPis or to 6-TG, without activating all of its downstream effectors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bencimidazoles/administración & dosificación , Resistencia a Antineoplásicos , Ácidos Hidroxámicos/administración & dosificación , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Animales , Proteína BRCA1/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Senescencia Celular , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación , Plásmidos/metabolismo , Recombinasa Rad51/metabolismo , Recombinación Genética , Tioguanina/administración & dosificación , Vorinostat , Pérdida de Peso
20.
J Clin Oncol ; 34(19): 2206-11, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27001570

RESUMEN

PURPOSE: Recurrent glioblastoma multiforme (GBM) is incurable with current therapies. Biallelic mismatch repair deficiency (bMMRD) is a highly penetrant childhood cancer syndrome often resulting in GBM characterized by a high mutational burden. Evidence suggests that high mutation and neoantigen loads are associated with response to immune checkpoint inhibition. PATIENTS AND METHODS: We performed exome sequencing and neoantigen prediction on 37 bMMRD cancers and compared them with childhood and adult brain neoplasms. Neoantigen prediction bMMRD GBM was compared with responsive adult cancers from multiple tissues. Two siblings with recurrent multifocal bMMRD GBM were treated with the immune checkpoint inhibitor nivolumab. RESULTS: All malignant tumors (n = 32) were hypermutant. Although bMMRD brain tumors had the highest mutational load because of secondary polymerase mutations (mean, 17,740 ± standard deviation, 7,703), all other high-grade tumors were hypermutant (mean, 1,589 ± standard deviation, 1,043), similar to other cancers that responded favorably to immune checkpoint inhibitors. bMMRD GBM had a significantly higher mutational load than sporadic pediatric and adult gliomas and all other brain tumors (P < .001). bMMRD GBM harbored mean neoantigen loads seven to 16 times higher than those in immunoresponsive melanomas, lung cancers, or microsatellite-unstable GI cancers (P < .001). On the basis of these preclinical data, we treated two bMMRD siblings with recurrent multifocal GBM with the anti-programmed death-1 inhibitor nivolumab, which resulted in clinically significant responses and a profound radiologic response. CONCLUSION: This report of initial and durable responses of recurrent GBM to immune checkpoint inhibition may have implications for GBM in general and other hypermutant cancers arising from primary (genetic predisposition) or secondary MMRD.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias Encefálicas/genética , Neoplasias Colorrectales/genética , Glioblastoma/genética , Mutación , Síndromes Neoplásicos Hereditarios/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Adulto , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/tratamiento farmacológico , Niño , Preescolar , Femenino , Glioblastoma/diagnóstico por imagen , Glioblastoma/tratamiento farmacológico , Humanos , Imagen por Resonancia Magnética , Masculino , Nivolumab
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...