Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Lancet Oncol ; 19(6): 785-798, 2018 06.
Article in English | MEDLINE | ID: mdl-29753700

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/genetics , Cerebellar Neoplasms/genetics , DNA Methylation , Genetic Testing/methods , Germ-Line Mutation , Medulloblastoma/genetics , Models, Genetic , Adolescent , Adult , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/therapy , Child , Child, Preschool , DNA Mutational Analysis , Female , Gene Expression Profiling , Genetic Predisposition to Disease , Heredity , Humans , Infant , Male , Medulloblastoma/mortality , Medulloblastoma/pathology , Medulloblastoma/therapy , Pedigree , Phenotype , Predictive Value of Tests , Progression-Free Survival , Prospective Studies , Reproducibility of Results , Retrospective Studies , Risk Factors , Transcriptome , Exome Sequencing , Young Adult
2.
Cancer Cell ; 31(6): 737-754.e6, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28609654

ABSTRACT

While molecular subgrouping has revolutionized medulloblastoma classification, the extent of heterogeneity within subgroups is unknown. Similarity network fusion (SNF) applied to genome-wide DNA methylation and gene expression data across 763 primary samples identifies very homogeneous clusters of patients, supporting the presence of medulloblastoma subtypes. After integration of somatic copy-number alterations, and clinical features specific to each cluster, we identify 12 different subtypes of medulloblastoma. Integrative analysis using SNF further delineates group 3 from group 4 medulloblastoma, which is not as readily apparent through analyses of individual data types. Two clear subtypes of infants with Sonic Hedgehog medulloblastoma with disparate outcomes and biology are identified. Medulloblastoma subtypes identified through integrative clustering have important implications for stratification of future clinical trials.


Subject(s)
Medulloblastoma/classification , Precision Medicine , Cluster Analysis , Cohort Studies , DNA Copy Number Variations , DNA Methylation , Gene Expression Profiling , Genomics , Humans , Medulloblastoma/genetics , Medulloblastoma/therapy
3.
PLoS One ; 12(2): e0172552, 2017.
Article in English | MEDLINE | ID: mdl-28231263

ABSTRACT

Medulloblastoma comprises four main subgroups (WNT, SHH, Group 3 and Group 4) originally defined by transcriptional profiling. In primary medulloblastoma tissues, these groups are thought to be distinguishable using the immunohistochemical detection of ß-catenin, filamin A, GAB1 and YAP1 protein markers. To investigate the utility of these markers for in vitro studies using medulloblastoma cell lines, immunoblotting and indirect immunofluorescence were employed for the detection of ß-catenin, filamin A, GAB1 and YAP1 in both DAOY and D283 Med reference cell lines and the panel of six medulloblastoma cell lines derived in our laboratory from the primary tumor tissues of known molecular subgroups. Immunohistochemical detection of these markers was performed on formalin-fixed paraffin-embedded tissue of the matching primary tumors. The results revealed substantial divergences between the primary tumor tissues and matching cell lines in the immunoreactivity pattern of medulloblastoma-subgroup-specific protein markers. Regardless of the molecular subgroup of the primary tumor, all six patient-derived medulloblastoma cell lines exhibited a uniform phenotype: immunofluorescence showed the nuclear localization of YAP1, accompanied by strong cytoplasmic positivity for ß-catenin and filamin A, as well as weak positivity for GAB1. The same immunoreactivity pattern was also found in both DAOY and D283 Med reference medulloblastoma cell lines. Therefore, we can conclude that various medulloblastoma cell lines tend to exhibit the same characteristics of protein marker expression under standard in vitro conditions. Such a finding emphasizes the importance of the analyses of primary tumors in clinically oriented medulloblastoma research and the urgent need to develop in vitro models of improved clinical relevance, such as 3D cultures and organotypic slice cultures.


Subject(s)
Cerebellar Neoplasms/pathology , Cerebellum/pathology , Medulloblastoma/pathology , Adolescent , Adult , Biomarkers, Tumor/analysis , Child , Child, Preschool , Female , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunohistochemistry , Infant , Male , Tumor Cells, Cultured , Young Adult
4.
Cancer Biomark ; 17(1): 107-16, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27314299

ABSTRACT

BACKGROUND: Nestin, CD133 and ABCG2 are recently discussed as putative markers, co-expression of which might determine a cancer stem cell (CSC) phenotype in sarcomas. OBJECTIVE: Our study is focused on immunohistochemical analysis of nestin, CD133 and ABCG2 expression in rhabdomyosarcoma, Ewing sarcoma and osteosarcoma. Furthermore, we also analyzed the possible correlation of nestin, CD133 and ABCG2 expression levels with the patient outcome to identify potential prognostic values of these three putative CSC markers in the same cohorts. METHODS: Using immunohistochemistry, expression of nestin, CD133 and ABCG2 was analyzed in 24 rhabdomyosarcoma, 22 Ewing sarcoma and 10 osteosarcoma tissue samples and expression levels of these markers were correlated with clinical outcome. RESULTS: High nestin levels indicate poor prognosis in patients with Ewing sarcoma (P = 0.001), and high CD133 expression is associated with shorter survival in rhabdomyosarcoma patients (P = 0.002). In contrast, no significant relationship was found between ABCG2 expression and the clinical outcome. CONCLUSIONS: Our analysis represents the first complex study of these three putative CSCs markers together in three different types of pediatric sarcomas and showed their possible prognostic values in these tumors.


Subject(s)
AC133 Antigen/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Biomarkers, Tumor , Nestin/metabolism , Sarcoma/metabolism , Sarcoma/mortality , AC133 Antigen/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Gene Expression , Humans , Immunohistochemistry , Infant , Infant, Newborn , Kaplan-Meier Estimate , Male , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Neoplastic Stem Cells/metabolism , Nestin/genetics , Prognosis , Sarcoma/diagnosis , Sarcoma/genetics , Young Adult
5.
Lancet Oncol ; 17(4): 484-495, 2016 04.
Article in English | MEDLINE | ID: mdl-26976201

ABSTRACT

BACKGROUND: Patients with incomplete surgical resection of medulloblastoma are controversially regarded as having a marker of high-risk disease, which leads to patients undergoing aggressive surgical resections, so-called second-look surgeries, and intensified chemoradiotherapy. All previous studies assessing the clinical importance of extent of resection have not accounted for molecular subgroup. We analysed the prognostic value of extent of resection in a subgroup-specific manner. METHODS: We retrospectively identified patients who had a histological diagnosis of medulloblastoma and complete data about extent of resection and survival from centres participating in the Medulloblastoma Advanced Genomics International Consortium. We collected from resections done between April, 1997, and February, 2013, at 35 international institutions. We established medulloblastoma subgroup affiliation by gene expression profiling on frozen or formalin-fixed paraffin-embedded tissues. We classified extent of resection on the basis of postoperative imaging as gross total resection (no residual tumour), near-total resection (<1·5 cm(2) tumour remaining), or sub-total resection (≥1·5 cm(2) tumour remaining). We did multivariable analyses of overall survival and progression-free survival using the variables molecular subgroup (WNT, SHH, group 4, and group 3), age (<3 vs ≥3 years old), metastatic status (metastases vs no metastases), geographical location of therapy (North America/Australia vs rest of the world), receipt of chemotherapy (yes vs no) and receipt of craniospinal irradiation (<30 Gy or >30 Gy vs no craniospinal irradiation). The primary analysis outcome was the effect of extent of resection by molecular subgroup and the effects of other clinical variables on overall and progression-free survival. FINDINGS: We included 787 patients with medulloblastoma (86 with WNT tumours, 242 with SHH tumours, 163 with group 3 tumours, and 296 with group 4 tumours) in our multivariable Cox models of progression-free and overall survival. We found that the prognostic benefit of increased extent of resection for patients with medulloblastoma is attenuated after molecular subgroup affiliation is taken into account. We identified a progression-free survival benefit for gross total resection over sub-total resection (hazard ratio [HR] 1·45, 95% CI 1·07-1·96, p=0·16) but no overall survival benefit (HR 1·23, 0·87-1·72, p=0·24). We saw no progression-free survival or overall survival benefit for gross total resection compared with near-total resection (HR 1·05, 0·71-1·53, p=0·8158 for progression-free survival and HR 1·14, 0·75-1·72, p=0·55 for overall survival). No significant survival benefit existed for greater extent of resection for patients with WNT, SHH, or group 3 tumours (HR 1·03, 0·67-1·58, p=0·89 for sub-total resection vs gross total resection). For patients with group 4 tumours, gross total resection conferred a benefit to progression-free survival compared with sub-total resection (HR 1·97, 1·22-3·17, p=0·0056), especially for those with metastatic disease (HR 2·22, 1·00-4·93, p=0·050). However, gross total resection had no effect on overall survival compared with sub-total resection in patients with group 4 tumours (HR 1·67, 0·93-2·99, p=0·084). INTERPRETATION: The prognostic benefit of increased extent of resection for patients with medulloblastoma is attenuated after molecular subgroup affiliation is taken into account. Although maximum safe surgical resection should remain the standard of care, surgical removal of small residual portions of medulloblastoma is not recommended when the likelihood of neurological morbidity is high because there is no definitive benefit to gross total resection compared with near-total resection. FUNDING: Canadian Cancer Society Research Institute, Terry Fox Research Institute, Canadian Institutes of Health Research, National Institutes of Health, Pediatric Brain Tumor Foundation, and the Garron Family Chair in Childhood Cancer Research.


Subject(s)
Brain Neoplasms/classification , Brain Neoplasms/surgery , Medulloblastoma/classification , Medulloblastoma/surgery , Prognosis , Adult , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Canada , Child , Child, Preschool , Combined Modality Therapy , Disease Progression , Disease-Free Survival , Female , Humans , Infant , Magnetic Resonance Imaging , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Retrospective Studies
6.
Tumour Biol ; 37(7): 9535-48, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26790443

ABSTRACT

The three most frequent pediatric sarcomas, i.e., Ewing's sarcoma, osteosarcoma, and rhabdomyosarcoma, were examined in this study: three cell lines derived from three primary tumor samples were analyzed from each of these tumor types. Detailed comparative analysis of the expression of three putative cancer stem cell markers related to sarcomas-ABCG2, CD133, and nestin-was performed on both primary tumor tissues and corresponding cell lines. The obtained results showed that the frequency of ABCG2-positive and CD133-positive cells was predominantly increased in the respective cell lines but that the high levels of nestin expression were reduced in both osteosarcomas and rhabdomyosarcomas under in vitro conditions. These findings suggest the selection advantage of cells expressing ABCG2 or CD133, but the functional tests in NOD/SCID gamma mice did not confirm the tumorigenic potential of cells harboring this phenotype. Subsequent analysis of the expression of common stem cell markers revealed an evident relationship between the expression of the transcription factor Sox2 and the tumorigenicity of the cell lines in immunodeficient mice: the Sox2 levels were highest in the two cell lines that were demonstrated as tumorigenic. Furthermore, Sox2-positive cells were found in the respective primary tumors and all xenograft tumors showed apparent accumulation of these cells. All of these findings support our conclusion that regardless of the expression of ABCG2, CD133 and nestin, only cells displaying increased Sox2 expression are directly involved in tumor initiation and growth; therefore, these cells fit the definition of the cancer stem cell phenotype.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Sarcoma/metabolism , Sarcoma/pathology , AC133 Antigen/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adolescent , Adult , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Child , Child, Preschool , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/pathology , Nestin/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology
7.
Cancer Cell ; 27(5): 728-43, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25965575

ABSTRACT

Ependymal tumors across age groups are currently classified and graded solely by histopathology. It is, however, commonly accepted that this classification scheme has limited clinical utility based on its lack of reproducibility in predicting patients' outcome. We aimed at establishing a uniform molecular classification using DNA methylation profiling. Nine molecular subgroups were identified in a large cohort of 500 tumors, 3 in each anatomical compartment of the CNS, spine, posterior fossa, supratentorial. Two supratentorial subgroups are characterized by prototypic fusion genes involving RELA and YAP1, respectively. Regarding clinical associations, the molecular classification proposed herein outperforms the current histopathological classification and thus might serve as a basis for the next World Health Organization classification of CNS tumors.


Subject(s)
Age Factors , Central Nervous System Neoplasms/pathology , Ependymoma/pathology , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Adult , Aged , Central Nervous System Neoplasms/classification , Central Nervous System Neoplasms/genetics , Child , Child, Preschool , DNA Methylation , Ependymoma/classification , Ependymoma/genetics , Female , Gene Dosage , Gene Expression Profiling , Gene Fusion , Humans , Infant , Male , Middle Aged , Phosphoproteins/genetics , Transcription Factors , Transcription, Genetic , YAP-Signaling Proteins , Young Adult
8.
Tumour Biol ; 36(10): 7483-91, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25910708

ABSTRACT

The p73 protein is a member of the p53 family, and this protein is known to be essential for the maintenance of genomic stability, DNA repair, and apoptosis regulation. Transcription from two promoters leads to two main N-terminal isoforms: the TAp73 isoform is reported to have tumor suppressor function, whereas the ΔNp73 isoform likely has oncogenic potential. The present study is focused on the investigation of a possible role of both these p73 N-terminal isoforms in the process of centrosome amplification. HGG-02 and GM7 glioblastoma cell lines and the Daoy medulloblastoma cell line were used in this study. The cells were analyzed using indirect immunofluorescence to determine TAp73 and ΔNp73 expression patterns and possible co-localization with the BubR1 protein, as well as the number of centrosomes. A transiently transfected GM7 cell line was used to verify the results concerning the N-terminal isoforms in relation to centrosome amplification. We found that increased immunoreactivity for the ΔNp73 isoform is associated with the occurrence of an abnormal number of centrosomes in particular cells. Using the transiently transfected GM7 cell line, we confirmed that centrosome amplification is present in cells with overexpression of the ΔNp73 isoform. In contrast, the immunoreactivity for the TAp73 isoform was weak or medium in most of the cells with an aberrant number of centrosomes. To determine the putative counterpart of the p73 N-terminal isoforms among spindle assembly checkpoint (SAC) proteins, we also evaluated possible interactions between the N-terminal isoforms and BubR1 protein, but no co-localization of these proteins was observed.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Centrosome/physiology , Chromosome Aberrations , DNA-Binding Proteins/genetics , Gene Amplification , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , DNA Repair , Fluorescent Antibody Technique, Indirect , Humans , Promoter Regions, Genetic , Protein Isoforms , Protein Serine-Threonine Kinases/genetics , Tumor Cells, Cultured , Tumor Protein p73
9.
Cancer Cell Int ; 14: 51, 2014.
Article in English | MEDLINE | ID: mdl-24959102

ABSTRACT

BACKGROUND: A detailed analysis of the expression of 440 cancer-related genes was performed after the combined treatment of medulloblastoma cells with all-trans retinoic acid (ATRA) and inhibitors of lipoxygenases (LOX) and cyclooxygenases (COX). The combinations of retinoids and celecoxib as a COX-2 inhibitor were reported to be effective in some regimens of metronomic therapy of relapsed solid tumors with poor prognosis. Our previous findings on neuroblastoma cells using expression profiling showed that LOX/COX inhibitors have the capability of enhancing the differentiating action of ATRA. Presented study focused on the continuation of our previous work to confirm the possibility of enhancing ATRA-induced cell differentiation in these cell lines via the application of LOX/COX inhibitors. This study provides more detailed information concerning the mechanisms of the enhancement of the ATRA-induced differentiation of medulloblastoma cells. METHODS: The Daoy and D283 Med medulloblastoma cell lines were chosen for this study. Caffeic acid (an inhibitor of 5-LOX) and celecoxib (an inhibitor on COX-2) were used in combined treatment with ATRA. The expression profiling was performed using Human Cancer Oligo GEArray membranes, and the most promising results were verified using RT-PCR. RESULTS: The expression profiling of the selected cancer-related genes clearly confirmed that the differentiating effects of ATRA should be enhanced via its combined administration with caffeic acid or celecoxib. This effect was detected in both cell lines. An increased expression of the genes that encoded the proteins participating in induced differentiation and cytoskeleton remodeling was detected in both cell lines in a concentration-dependent manner. This effect was also observed for the CDKN1A gene encoding the p21 protein, which is an important regulator of the cell cycle, and for the genes encoding proteins that are associated with proteasome activity. Furthermore, our results showed that D283 Med cells are significantly more sensitive to treatment with ATRA alone than Daoy cells. CONCLUSIONS: The obtained results on medulloblastoma cell lines are in accordance with our previous findings on neuroblastoma cells and confirm our hypothesis concerning the common mechanism of the enhancement of ATRA-induced cell differentiation in various types of pediatric solid tumors.

10.
Tumour Biol ; 35(8): 7617-27, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24798977

ABSTRACT

The induced differentiation of tumor cells into mature phenotypes is a promising strategy in cancer therapy. In this study, the effects of combined treatment with all-trans retinoic acid (ATRA) and lipoxygenase/cyclooxygenase inhibitors were examined in two osteosarcoma cell lines, Saos-2 and OSA-01. Caffeic acid and celecoxib were used as inhibitors of 5-lipoxygenase and of cyclooxygenase-2, respectively. Changes in the cell proliferation, matrix mineralization, and occurrence of differentiation markers were evaluated in treated cell populations at intervals. The results confirmed the capability of caffeic acid to enhance the antiproliferative effect of ATRA in both cell lines. In contrast, celecoxib showed the same effect in Saos-2 cells only. Furthermore, the extension of matrix mineralization was observed after combined treatment with ATRA and celecoxib or caffeic acid. The increased expression of osteogenic differentiation markers was observed in both cell lines after the combined application of ATRA and inhibitors. The obtained results clearly demonstrate the capability of lipoxygenase/cyclooxygenase inhibitors to enhance the antiproliferative and differentiating effect of ATRA in osteosarcoma cells, although some of these effects are specific and depend on the biological features of the respective tumor or cell line.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Cyclooxygenase Inhibitors/pharmacology , Lipoxygenase Inhibitors/pharmacology , Osteosarcoma/drug therapy , Tretinoin/pharmacology , Bone Neoplasms/pathology , Caffeic Acids/pharmacology , Celecoxib , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Osteosarcoma/pathology , Pyrazoles/pharmacology , Sulfonamides/pharmacology
11.
J Clin Oncol ; 32(9): 886-96, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24493713

ABSTRACT

PURPOSE: Medulloblastoma comprises four distinct molecular subgroups: WNT, SHH, Group 3, and Group 4. Current medulloblastoma protocols stratify patients based on clinical features: patient age, metastatic stage, extent of resection, and histologic variant. Stark prognostic and genetic differences among the four subgroups suggest that subgroup-specific molecular biomarkers could improve patient prognostication. PATIENTS AND METHODS: Molecular biomarkers were identified from a discovery set of 673 medulloblastomas from 43 cities around the world. Combined risk stratification models were designed based on clinical and cytogenetic biomarkers identified by multivariable Cox proportional hazards analyses. Identified biomarkers were tested using fluorescent in situ hybridization (FISH) on a nonoverlapping medulloblastoma tissue microarray (n = 453), with subsequent validation of the risk stratification models. RESULTS: Subgroup information improves the predictive accuracy of a multivariable survival model compared with clinical biomarkers alone. Most previously published cytogenetic biomarkers are only prognostic within a single medulloblastoma subgroup. Profiling six FISH biomarkers (GLI2, MYC, chromosome 11 [chr11], chr14, 17p, and 17q) on formalin-fixed paraffin-embedded tissues, we can reliably and reproducibly identify very low-risk and very high-risk patients within SHH, Group 3, and Group 4 medulloblastomas. CONCLUSION: Combining subgroup and cytogenetic biomarkers with established clinical biomarkers substantially improves patient prognostication, even in the context of heterogeneous clinical therapies. The prognostic significance of most molecular biomarkers is restricted to a specific subgroup. We have identified a small panel of cytogenetic biomarkers that reliably identifies very high-risk and very low-risk groups of patients, making it an excellent tool for selecting patients for therapy intensification and therapy de-escalation in future clinical trials.


Subject(s)
Biomarkers, Tumor/genetics , Hedgehog Proteins , Medulloblastoma/genetics , Wnt Proteins , Adolescent , Child , Child, Preschool , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Cytogenetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Infant , Kruppel-Like Transcription Factors/genetics , Male , Medulloblastoma/mortality , Medulloblastoma/pathology , Medulloblastoma/therapy , Nuclear Proteins/genetics , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins c-myc/genetics , Reproducibility of Results , Risk Assessment , Risk Factors , Tissue Array Analysis , Wnt Proteins/genetics , Young Adult , Zinc Finger Protein Gli2
12.
Oncol Rep ; 31(1): 480-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24270553

ABSTRACT

Epidermal growth factor receptor (EGFR) gene amplification and the overexpression of EGFR are described as common features of glioblastoma multiforme (GBM). Nevertheless, we previously reported the loss of EGFR gene copy in a GBM specimen from a patient with an unusually favorable course of the disease, and the HGG-02 cell line with this aberration was successfully derived from this tumor. Here, we present a detailed analysis of changes in gene expression and cell signaling in the HGG-02 cell line; the GM7 reference cell line with a standard EGFR gene copy number derived from a very aggressive GBM was used as a control. We confirmed the downregulation of EGFR expression and signaling in HGG-02 cells using different methods (RTK analysis, gene profiling and RT-PCR). Other changes that may have contributed to the non-aggressive phenotype of the primary tumor were identified, including the downregulated phosphorylation of the Axl and Trk receptors, as well as increased activity of JNK and p38 kinases. Notably, differences in PDGF signaling were detected in both of these cell lines; HGG-02 cells preferentially expressed and signaled through PDGFRα, and PDGFRß was strongly overexpressed and phosphorylated in the GM7 reference cell line. Using expression profiling of cancer-related genes, we revealed the specific profile of HGG-02 cells that included upregulated tumor-suppressors as well as downregulated genes associated with the extracellular matrix. This study represents the first comprehensive analysis of gene expression and cell signaling in glioblastoma cells with lower EGFR gene dosage. As indicated by our results, the TAM receptors, Trk receptors and PDGFRs need to be investigated further since their regulation appears to be important for glioblastoma biological features as well as the clinical course of the disease.


Subject(s)
Brain Neoplasms/genetics , ErbB Receptors/genetics , Gene Deletion , Gene Dosage , Glioblastoma/genetics , Cell Line, Tumor , Gene Expression Profiling , Humans , JNK Mitogen-Activated Protein Kinases/biosynthesis , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, trkA/metabolism , Signal Transduction/genetics , p38 Mitogen-Activated Protein Kinases/biosynthesis , Axl Receptor Tyrosine Kinase
13.
Acta Neuropathol ; 126(6): 917-29, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24174164

ABSTRACT

Telomerase reverse transcriptase (TERT) promoter mutations were recently shown to drive telomerase activity in various cancer types, including medulloblastoma. However, the clinical and biological implications of TERT mutations in medulloblastoma have not been described. Hence, we sought to describe these mutations and their impact in a subgroup-specific manner. We analyzed the TERT promoter by direct sequencing and genotyping in 466 medulloblastomas. The mutational distributions were determined according to subgroup affiliation, demographics, and clinical, prognostic, and molecular features. Integrated genomics approaches were used to identify specific somatic copy number alterations in TERT promoter-mutated and wild-type tumors. Overall, TERT promoter mutations were identified in 21 % of medulloblastomas. Strikingly, the highest frequencies of TERT mutations were observed in SHH (83 %; 55/66) and WNT (31 %; 4/13) medulloblastomas derived from adult patients. Group 3 and Group 4 harbored this alteration in <5 % of cases and showed no association with increased patient age. The prognostic implications of these mutations were highly subgroup-specific. TERT mutations identified a subset with good and poor prognosis in SHH and Group 4 tumors, respectively. Monosomy 6 was mostly restricted to WNT tumors without TERT mutations. Hallmark SHH focal copy number aberrations and chromosome 10q deletion were mutually exclusive with TERT mutations within SHH tumors. TERT promoter mutations are the most common recurrent somatic point mutation in medulloblastoma, and are very highly enriched in adult SHH and WNT tumors. TERT mutations define a subset of SHH medulloblastoma with distinct demographics, cytogenetics, and outcomes.


Subject(s)
Brain Neoplasms/genetics , Medulloblastoma/genetics , Mutation , Promoter Regions, Genetic , Telomerase/genetics , Adolescent , Adult , Brain Neoplasms/pathology , Child , Child, Preschool , DNA Mutational Analysis , Female , Gene Expression Profiling , Genotype , Humans , Infant , Male , Medulloblastoma/pathology , Middle Aged , Prognosis
14.
J Clin Oncol ; 31(23): 2927-35, 2013 Aug 10.
Article in English | MEDLINE | ID: mdl-23835706

ABSTRACT

PURPOSE: Reports detailing the prognostic impact of TP53 mutations in medulloblastoma offer conflicting conclusions. We resolve this issue through the inclusion of molecular subgroup profiles. PATIENTS AND METHODS: We determined subgroup affiliation, TP53 mutation status, and clinical outcome in a discovery cohort of 397 medulloblastomas. We subsequently validated our results on an independent cohort of 156 medulloblastomas. RESULTS: TP53 mutations are enriched in wingless (WNT; 16%) and sonic hedgehog (SHH; 21%) medulloblastomas and are virtually absent in subgroups 3 and 4 tumors (P < .001). Patients with SHH/TP53 mutant tumors are almost exclusively between ages 5 and 18 years, dramatically different from the general SHH distribution (P < .001). Children with SHH/TP53 mutant tumors harbor 56% germline TP53 mutations, which are not observed in children with WNT/TP53 mutant tumors. Five-year overall survival (OS; ± SE) was 41% ± 9% and 81% ± 5% for patients with SHH medulloblastomas with and without TP53 mutations, respectively (P < .001). Furthermore, TP53 mutations accounted for 72% of deaths in children older than 5 years with SHH medulloblastomas. In contrast, 5-year OS rates were 90% ± 9% and 97% ± 3% for patients with WNT tumors with and without TP53 mutations (P = .21). Multivariate analysis revealed that TP53 status was the most important risk factor for SHH medulloblastoma. Survival rates in the validation cohort mimicked the discovery results, revealing that poor survival of TP53 mutations is restricted to patients with SHH medulloblastomas (P = .012) and not WNT tumors. CONCLUSION: Subgroup-specific analysis reconciles prior conflicting publications and confirms that TP53 mutations are enriched among SHH medulloblastomas, in which they portend poor outcome and account for a large proportion of treatment failures in these patients.


Subject(s)
Cerebellar Neoplasms/genetics , Genes, p53 , Medulloblastoma/genetics , Mutation , Adolescent , Adult , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Female , Gene Expression Profiling , Humans , Infant , Male , Medulloblastoma/pathology , Middle Aged , Prognosis , Young Adult
15.
Histol Histopathol ; 28(7): 913-24, 2013 07.
Article in English | MEDLINE | ID: mdl-23338942

ABSTRACT

The p73 protein is a member of the p53 family of transcription factors that has two N-terminal isoforms: the TAp73 isoform is reported to have a tumor suppressor function, whereas the ΔNp73 isoform likely has oncogenic potential. The expression of these isoforms and the differences in their intracellular distribution have been described in many cancer types; however, little is known about the p73 isoforms in brain tumors. Our study is focused on the intracellular localization of ΔNp73 in medulloblastoma cell lines. Due to a lack of suitable anti-ΔNp73 antibodies, we developed two new rabbit polyclonal antibodies, ΔNp73-26 and ΔNp73-27, with sufficient specificity, as demonstrated by immunodetection methods using transiently transfected cell lines. Both of these new antibodies were subsequently used for analysis of the ΔNp73 distribution in medulloblastoma cells using immunofluorescence, immunoblotting and immunogold labeling for transmission electron microscopy. We found a nuclear localization of the ΔNp73 isoform in all of the medulloblastoma cell lines included in this study. Furthermore, a non-random accumulation of the ΔNp73 isoform near the cell nuclei was observable in all of these cell lines. By double-labeling with ΔNp73 and golgin-97, we showed the co-localization of the ΔNp73 isoform with the Golgi apparatus. Nevertheless, further detailed analyses of possible interactions of ΔNp73 with the proteins accumulated in the Golgi apparatus should be performed to explain the dynamics of ΔNp73 outside the cell nucleus.


Subject(s)
Antibodies/chemistry , DNA-Binding Proteins/metabolism , Medulloblastoma/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Child , Child, Preschool , Female , Golgi Apparatus/metabolism , Humans , Immunohistochemistry , Infant , MCF-7 Cells , Male , Microscopy, Electron, Transmission , Protein Isoforms/metabolism , Rabbits , Transfection , Tumor Protein p73
16.
Oncol Rep ; 27(5): 1592-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22344865

ABSTRACT

Nestin has been detected in various malignancies and its expression correlates with advanced grade in some neoplasms. The aim of this study was to examine nestin expression in high-grade osteosarcomas and to determine its prognostic value. Using immunohistochemistry and immunofluorescence, we evaluated nestin expression in tumor tissue samples from 45 patients with high-grade osteosarcomas. In both methods, the frequency of nestin-positive tumor cells was classified into three categories (1+, 2+ and 3+ for immunohistochemistry; 1F+, 2F+ and 3F+ for immunofluorescence) and clinicopathological correlations were statistically evaluated and analyzed. Nestin-positive tumor cells were detected in all of the examined osteosarcomas using both immunohistochemistry and immunofluorescence, although the proportion of undoubtedly positive neoplastic cells varied in individual samples from a few nestin-positive tumor cells to diffuse nestin positivity. High levels of nestin expression detected by immunofluorescence (2F+ and 3F+) were associated with worse clinical outcomes (OS, p=0.031; EFS, p<0.001). However, high levels of nestin expression as measured by immunohistochemistry trended towards shorter patient survival rates but did not reach statistical significance. Despite significantly shorter survival rates observed in patients with high levels of nestin expression assessed by immunofluorescence, nestin does not seem to represent a powerful prognostic marker that would be superior to conventional methods.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Bone Neoplasms/mortality , Child , Female , Humans , Intermediate Filament Proteins/genetics , Male , Middle Aged , Neoplasm Grading , Nerve Tissue Proteins/genetics , Nestin , Osteosarcoma/mortality , Prognosis , Young Adult
17.
Anal Cell Pathol (Amst) ; 34(6): 303-18, 2011.
Article in English | MEDLINE | ID: mdl-22156015

ABSTRACT

BACKGROUND: Co-expression of CD133, cell surface glycoprotein, and nestin, an intermediate filament protein, was determined to be a marker of neural stem cells and of cancer stem cells in neurogenic tumors. METHODS: We examined the expression of CD133 and nestin in ten tumor tissue samples taken from patients with rhabdomyosarcomas and in five rhabdomyosarcoma cell lines. Immunohistochemistry and immunofluorescence were used to examine FFPE tumor tissue samples. Cell lines were analyzed by immunofluorescence, immunoblotting, flow cytometry, and RT-PCR. Functional assays (clonogenic in vitro assay and tumorigenic in vivo assay) were also performed using these cell lines. RESULTS: CD133 and nestin were detected in all 10 tumor tissue samples and in all 5 cell lines; however, the frequency of CD133+, Nes+, and CD133+/Nes+ cells, as well as the intensity of fluorescence varied in individual samples or cell lines. The expression of CD133 and nestin was subsequently confirmed in all cell lines by immunoblotting. Furthermore, we observed an increasing expression of CD133 in relation to the cultivation. All cell lines were positive for Oct3/4 and nucleostemin; NSTS-11 cells were also able to form xenograft tumors in mice. CONCLUSION: Our results represent the first evidence of CD133 expression in rhabdomyosarcoma tissue and in rhabdomyosarcoma cell lines. In addition, the co-expression of CD133 and nestin as well as results of the functional assays suggest a possible presence of cancer cells with a stem-like phenotype in these tumors.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Peptides/metabolism , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , AC133 Antigen , Adolescent , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Child , Child, Preschool , Cohort Studies , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Male , Nestin , Rhabdomyosarcoma/genetics , Stem Cells/metabolism , Young Adult
18.
Tumour Biol ; 32(4): 631-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21340483

ABSTRACT

Nestin is a class VI intermediate filament protein expressed in the cytoplasm of stem and progenitor cells in the mammalian CNS during development. In adults, nestin is present only in a small subset of cells and tissues, including the subventricular zone of the adult mammalian brain, where neurogenesis occurs. Nestin expression has also been detected under such pathological conditions as ischemia, inflammation, and brain injury, as well as in various types of human solid tumors and their corresponding cell lines. Furthermore, nestin was recently found in the nuclei of glioblastoma, neuroblastoma, and angiosarcoma cells and it was proved to interact directly with the nuclear DNA in neuroblastoma cells. Here, we perform the first study of the intracellular distribution of nestin in cell lines derived from neurogenic tumors. Using immunodetection methods, we examined nestin expression in tumor-derived cell lines obtained from 11 patients with neuroblastoma, medulloblastoma, or glioblastoma multiforme. Besides its standard cytoplasmic localization, nestin was present in the nuclei of two neuroblastoma cell lines and one medulloblastoma cell line. Nestin was only present in the nuclei of cells with diffuse cytoplasmic staining for this protein, and the proportion of cells positive for nestin in nuclei, as well as the intensity of staining, varied. The presence of nestin in the nuclei was confirmed by both transmission electron microscopy and Western blotting. Our results indicate that the presence of nestin in the nuclei of tumor cells is not very rare, especially under in vitro conditions.


Subject(s)
Cell Nucleus/metabolism , Glioblastoma/metabolism , Intermediate Filament Proteins/metabolism , Medulloblastoma/metabolism , Nerve Tissue Proteins/metabolism , Neuroblastoma/metabolism , Aged , Blotting, Western , Cell Line, Tumor , Cell Nucleus/chemistry , Cell Nucleus/ultrastructure , Child , Child, Preschool , Female , Fluorescent Antibody Technique , Glioblastoma/ultrastructure , Humans , Immunohistochemistry , Infant , Intermediate Filament Proteins/analysis , Male , Medulloblastoma/ultrastructure , Microscopy, Electron, Transmission , Middle Aged , Nerve Tissue Proteins/analysis , Nestin , Neuroblastoma/ultrastructure
19.
J Neurooncol ; 102(1): 25-33, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20607354

ABSTRACT

High-level amplifications of MYC genes are associated with poor outcomes in childhood medulloblastoma (MB). However, the occurrence of MYCN and MYCC copy number increases below the intense amplification pattern is rarely reported, and its clinical impact has not yet been determined. Here, we describe this phenomenon and its prognostic significance in a cohort of 29 MB patients. Using interphase fluorescence in situ hybridization (I-FISH), low-level copy number alterations, i.e. gain of MYCN, were shown in 5/27 (19%) samples, whereas amplification was revealed in only 1/27 (4%) samples. MYCC gain was revealed in 6/29 (21%) MB, while amplification was disclosed in only 2/29 (7%). Hyperploidy and co-incidence of gains in both MYC loci were frequently observed in samples with copy number aberrations. Survival analysis has clearly shown that MYC copy number increases are associated with lowered event-free survival and overall survival in MB. In the case of MYCN, this negative correlation was statistically significant. We conclude that limited numerical alterations in loci 2p24 (MYCN) and 8q24 (MYCC), as assessed by I-FISH, are present in MB with a higher frequency than high-level amplifications. Poor prognoses were observed in patients with copy number increases in MYC genes. Our data illustrate the importance of further investigations in multicenter trials to better refine the emerging genomic-based prognostic stratification in MB.


Subject(s)
Cerebellar Neoplasms/genetics , Gene Amplification , Gene Dosage , Medulloblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Adolescent , Child , Child, Preschool , Cohort Studies , Female , Humans , In Situ Hybridization, Fluorescence , Interphase , Male , N-Myc Proto-Oncogene Protein , Prognosis , Prospective Studies , Retrospective Studies , Survival Rate
20.
J Mol Histol ; 41(4-5): 267-75, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20803057

ABSTRACT

The protein homologous to the tumor suppressor p53, p73, has essential roles in development and tumorigenesis. This protein exists in a wide range of isoforms with different, even antagonistic, functions. However, there are virtually no detailed morphological studies analyzing the endogenous expression of p73 isoforms at the cellular level in cancer cells. In this study, we investigated the expression and subcellular distribution of two N-terminal isoforms, TAp73 and ΔNp73, in medulloblastoma cells using immunofluorescence microscopy. Both proteins were observed in all cell lines examined, but differences were noted in their intracellular localization between the reference Daoy cell line and four newly established medulloblastoma cell lines (MBL-03, MBL-06, MBL-07 and MBL-10). In the new cell lines, TAp73 and ΔNp73 were located predominantly in cell nuclei. However, there was heterogeneity in TAp73 distribution in the cells of all MBL cell lines, with the protein located in the nucleus and also in a limited non-random area in the cytoplasm. In a small percentage of cells, we detected cytoplasmic localization of TAp73 only, i.e., nuclear exclusion was observed. Our results provide a basis for future studies on the causes and function of distinct intracellular localization of p73 protein isoforms with respect to different protein-protein interactions in medulloblastoma cells.


Subject(s)
DNA-Binding Proteins/metabolism , Intracellular Space/metabolism , Medulloblastoma/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Child , Child, Preschool , DNA-Binding Proteins/chemistry , Female , Humans , Male , Medulloblastoma/pathology , Nuclear Proteins/chemistry , Protein Isoforms/metabolism , Protein Transport , Tumor Protein p73 , Tumor Suppressor Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...