Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • Online Resource  (36)
  • American Association for Cancer Research (AACR)  (36)
Type of Medium
  • Online Resource  (36)
Publisher
  • American Association for Cancer Research (AACR)  (36)
Language
Subjects(RVK)
  • 1
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 11, No. 11 ( 2021-11-01), p. 2764-2779
    Abstract: INFORM is a prospective, multinational registry gathering clinical and molecular data of relapsed, progressive, or high-risk pediatric patients with cancer. This report describes long-term follow-up of 519 patients in whom molecular alterations were evaluated according to a predefined seven-scale target prioritization algorithm. Mean turnaround time from sample receipt to report was 25.4 days. The highest target priority level was observed in 42 patients (8.1%). Of these, 20 patients received matched targeted treatment with a median progression-free survival of 204 days [95% confidence interval (CI), 99–not applicable], compared with 117 days (95% CI, 106–143; P = 0.011) in all other patients. The respective molecular targets were shown to be predictive for matched treatment response and not prognostic surrogates for improved outcome. Hereditary cancer predisposition syndromes were identified in 7.5% of patients, half of which were newly identified through the study. Integrated molecular analyses resulted in a change or refinement of diagnoses in 8.2% of cases. Significance: The pediatric precision oncology INFORM registry prospectively tested a target prioritization algorithm in a real-world, multinational setting and identified subgroups of patients benefiting from matched targeted treatment with improved progression-free survival, refinement of diagnosis, and identification of hereditary cancer predisposition syndromes. See related commentary by Eggermont et al., p. 2677 . This article is highlighted in the In This Issue feature, p. 2659
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2607892-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3094-3094
    Abstract: Since it has become evident that histopathological grading of ependymoma according to the WHO classification of CNS tumors is not capable of accurately classifying patients into meaningful strata, a broadly accepted molecular classification scheme with prognostic significance is desperately needed. In recent years, ependymomas were classified into molecular subgroups based on transcriptomic alterations. In tumors localized within the posterior fossa, two distinct biological entities of ependymoma were delineated by several studies (designated posterior fossa A and posterior fossa B), which show striking differences in genetic characteristics and clinical outcome. A similar consensus for supratentorial and spinal ependymoma is lacking. We studied genome-wide DNA methylation (Illumina HumanMethylation450 (450k) array) in 180 primary ependymal tumors (80 with corresponding gene expression profiling data generated by Affymetrix 133plus2.0 arrays), including ependymomas (posterior fossa, supratentorial, spinal), subependymomas (SE), myxopapillary ependymoma (MPE), pineal parenchymal tumors of intermediate differentiation (PPTID), and papillary tumors of the pineal region (PTPR). We performed hierarchical clustering to identify robust molecular subgroups. Independent gene expression profiling datasets from previously published ependymoma studies (Johnson et al.; Wani et al.; Witt et al.) were used as validation cohorts. DNA methylation data showed that ependymal brain tumors can be classified into eight molecular subgroups. Notably, MPE, SE, PPTID and PTPR tumors formed robust distinct clusters, as did posterior fossa Group A and Group B ependymomas. Supratentorial ependymomas can be classified into two principle molecular subgroups, one of which displays a dismal prognosis, and comprises predominantly children and infants, and is associated with highly recurrent gene fusion. Notably, a significant number of ependymomas previously classified by histology as WHO Grade II/III look like SE by methylation, and also have extremely good survival. In summary, using genome-wide DNA methylation and transcriptome analysis we could decipher robust molecular subgroups of ependymal brain tumors including supratentorial ependymoma. Diagnoses of tumors with challenging histopathological features can now be supported by this technology. Hence, this approach offers the possibility to replace the unambiguous histological grading system that is currently in use with a robust molecular classification that readily distinguishes biologically, genetically, and clinically meaningful subgroups of ependymal brain tumors. Citation Format: Hendrik Witt, Martin Sill, Khalida Wani, Steve Mack, David Capper, Stephanie Heim, Pascal Johann, Sally Lambert, Marina Rhyzova, Volker Hovestadt, Theophilos Tzaridis, Kristian Pajtler, Sebastian Bender, Till Milde, Paul A. Northcott, Andreas E. Kulozik, Olaf Witt, Peter Lichter, V Peter Collins, Andreas von Deimling, Marcel Kool, Michael D. Taylor, Martin Hasselblatt, David TW Jones, Andrey Korshunov, Ken Aldape, Stefan Pfister. Epigenetic classification of ependymal brain tumors across age groups. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3094. doi:10.1158/1538-7445.AM2014-3094
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3453-3453
    Abstract: Despite intensive radio- and chemotherapy, incompletely resectable ependymomas are associated with a poor prognosis, and novel treatments have been difficult to develop due to the lack of appropriate pre-clinical models. We here report on the generation of a first pre-clinical ependymoma model (termed DKFZ-EP1NS) with long term self-renewal capacity, and the use of histonedeacetylase-inhibitors (HDACi) in the treatment of highly aggressive ependymoma cells. We were able to isolate ependymoma cells and grow them in spheres using serum-free neurosphere media conditions. We then characterized them for genetic aberrations, marker expression and tumorigeneity in an orthotopic xenotransplant model. The DKFZ-EP1NS cells were kept under neurosphere culture conditions for up to 9 months, displaying long term self-renewal. Freezing for storage and thawing was routinely possible. Genetic aberrations (loss at 1p36, chromosome 9 incl. homozygous deletion at CDKN2A and 14q) found in the DKFZ-EP1NS corresponded to the aberrations discovered in the primary ependymoma and subsequent recurrent tumors of the patient. DKFZ-EP1NS cells display several markers associated with normal stem cells as well as cancer stem cells. Orthotopically transplanted mice displayed first tumors after 9 months in the striatum of the brain, and tumors phenotypically recapitulated the original tumor. Serial transplantation yielded secondary tumors in half the time. Subcutaneous or intraperitoneal transplantation did not recapitulate the original intracranial histopathological phenotype, indicating that the orthotopic niche is required for the induction of the chracteristic phenotype. Although the cells were resistant to commonly used chemotherapeutic agents, they responded to HDACi-treatment in vitro at doses comparable to peak plasma levels in patients. In vitro-treatment of DKFZ-EP1NS cells with the HDACi Vorinostat led to morphological changes resembling neuronal differentiation. Indeed, neuronal markers showed a marked upregulation as measured by quantitative RT-PCR. Finally, stem cell-specific properties such as the neurosphere initiation capacity were lost upon treatment with Vorinostat in vitro. In summary, we were able to establish a first ependymoma cell line with stem cell-like properties recapitulating human disease in an orthotopic xenograft model, allowing for pre-clinical evaluation of drugs targeting the cancer stem cell compartment in ependymoma. Our data derived from applying well known drugs to this in vitro-model suggest that Vorinostat may have differentiation potential in the treatment of ependymoma cells resistant to conventional chemotherapeutic drugs in vivo as well. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3453. doi:10.1158/1538-7445.AM2011-3453
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4699-4699
    Abstract: Pilocytic astrocytoma (PA) is the most common brain tumor in children. Underlying genetic driver aberrations can currently be determined for 75-80% of cases. In particular, we and others have recently shown that tandem duplication at 7q34, resulting in BRAF fusion genes and constitutive activation of the MAPK signaling pathway, is a hallmark genetic lesion in PA development. Alternative mechanisms of MAPK activation include BRAF and KRAS point mutations, RAF1 fusions, and Neurofibromatosis-associated NF1 mutations. In order to examine more precisely the spectrum of alterations in PA, we screened 79 tumor samples for RAF fusion genes and mutations in KRAS, NRAS, PTPN11, BRAF and RAF1. We used multiplex and long-distance inverse (LDI) PCR to identify BRAF and RAF1 fusion genes and direct sequencing for detailed breakpoint mapping. Strikingly, LDI-PCR revealed a novel BRAF fusion gene with an uncharacterized gene, FAM131B, as a partner. Array-based comparative genomic hybridization (aCGH), revealed an interstitial deletion of ∼2.5 Mb as a novel mechanism forming the FAM131B-BRAF fusion. As with the more common duplication, this deletion removes the N-terminal auto-inhibitory domain of BRAF kinase, resulting in constitutive kinase activity. Functional characterization of the novel fusion gene demonstrated constitutive MEK phosphorylation potential and transforming activity in NIH 3T3 cells. The same fusion gene was also identified in one PA in an additional series from Cambridge, UK (n=7, with no previously identified alteration). Furthermore, we have detected a larger deletion at 7q in one additional case from our series, in which the alternative fusion partner is currently being identified. Overall, gene fusions targeting RAF kinases were identified in 68% (54/79) of PA. Detailed analysis of genomic DNA mapped 96% (52/54) of the breakpoints to the same breakpoint cluster region in intron 8 of the BRAF gene. Moreover, we identified the first non-intronic breakpoint in exon 8 of BRAF and two novel SRGAP3-RAF1 fusion variants. BRAF, KRAS or NF1 mutations were observed as alternative mechanisms of MAPK activation in 9 tumors in which no RAF duplication was detected, as well as in two cases in our series which concomitantly harbored two or even three hits in the MAPK pathway. In summary, we have identified a novel, recurrent BRAF fusion gene resulting in MAPK pathway activation in PA caused by a genomic deletion rather than amplification at 7q34, suggesting the possibility of further undiscovered fusion variants targeting RAF genes in this and other tumor types. Being a hallmark of PA tumorigenesis, these RAF fusion genes are expected to have clinical utility as both a specific marker for PA and a tumor-specific therapeutic target, which offers promise for applying novel treatment strategies in the near future. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4699. doi:10.1158/1538-7445.AM2011-4699
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4872-4872
    Abstract: Introduction: The International Cancer Genome Consortium (ICGC) is a worldwide network aiming to provide comprehensive molecular genetic profiles of 50 clinically and societally important tumors. A major focus of the ICGC PedBrain Tumor project is medulloblastoma (MB) - the most common malignant brain tumor in childhood. Importantly, recent work has shown that MB is not a single disease, but is in fact comprised of 4 distinct molecular subgroups (WNT, SHH, Group 3, Group 4). Using an integrative next-generation sequencing-based approach, we have investigated tumorigenic events underlying medulloblastoma, with the aim of identifying novel diagnostic or prognostic markers, and therapeutic targets. Methods: The whole genome of 50 tumor-normal DNA pairs, plus the whole exome or 2,500 target genes in a further 100 sample pairs from different histological and transcriptomic subgroups, was sequenced on an Illumina HiSeq platform. Sequencing-based high-resolution copy-number data was also generated for all cases. Results: Overall, a smaller number of non-synonymous somatic mutations was seen than in most adult malignancies. The number of somatic changes also clearly showed a positive correlation with patient age, suggesting either longer-term selection for more weakly transforming alterations, or the acquisition of additional passenger mutations in tumors which grew over a longer period. Whilst some point mutations and small InDels affected known MB or cancer-related genes (e.g. CTNNB1, PTCH1, MLL2), the vast majority were in genes which have not previously been implicated in medulloblastoma. Furthermore, even the most recurrently altered genes were mutated in & lt;20% of samples overall, although several showed a clear enrichment in distinct subgroups. Pathway analysis revealed chromatin modification as one of the most frequently altered cellular processes, with hits in multiple pathway members. Novel copy-number alterations were also identified. For example, a pattern of catastrophic chromosome shattering (‘chromothripsis’) was observed almost exclusively in the SHH tumor subgroup. Conclusion: Next-generation sequencing of this large tumor cohort has provided a number of new insights into this deadly pediatric tumor. The overriding pattern is one of enormous heterogeneity, but integration with molecular subgrouping shows clear enrichment of certain alterations. This first stage of the ICGC PedBrain project has demonstrated the power of next-gen sequencing approaches to reveal significant new insights, providing a number of novel targets for potential therapeutic intervention. It also, however, indicates the importance of personalised approaches to treatment optimisation, and shows the scale of the coming challenge in terms of functionally validating the huge number of novel mutations. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4872. doi:1538-7445.AM2012-4872
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 17, No. 14 ( 2011-07-15), p. 4650-4660
    Abstract: Purpose: Oncogenic BRAF/Ras or NF1 loss can potentially trigger oncogene-induced senescence (OIS) through activation of the mitogen-activated protein kinase (MAPK) pathway. Somatic genetic abnormalities affecting this pathway occur in the majority of pilocytic astrocytomas (PA), the most prevalent brain neoplasm in children. We investigated whether OIS is induced in PA. Experimental Design: We tested expression of established senescence markers in three independent cohorts of sporadic PA. We also assessed for OIS in vitro, using forced expression of wild-type and V600E-mutant BRAF in two astrocytic cell lines: human telomerase reverse transcriptase (hTERT)-immortalized astrocytes and fetal astrocytes. Results: Our results indicate that PAs are senescent as evidenced by marked senescence-associated acidic β-galactosidase activity, low KI-67 index, and induction of p16INK4a but not p53 in the majority of 52 PA samples (46 of 52; 88.5%). Overexpression of a number of senescence-associated genes [CDKN2A (p16), CDKN1A (p21), CEBPB, GADD45A, and IGFBP7] was shown at the mRNA level in two independent PA tumor series. In vitro, sustained activation of wild-type or mutant BRAF induced OIS in both astrocytic cell lines. Loss of p16INK4a in immortalized astrocytes abrogated OIS, indicative of the role of this pathway in mediating this phenomenon in astrocytes. OIS is a mechanism of tumor suppression that restricts the progression of benign tumors. We show that it is triggered in PAs through p16INK4a pathway induction following aberrant MAPK activation. Conclusions: OIS may account for the slow growth pattern in PA, the lack of progression to higher-grade astrocytomas, and the high overall survival of affected patients. Clin Cancer Res; 17(14); 4650–60. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1432-1432
    Abstract: Brain tumors are the most common cause of cancer-related death in childhood. Ependymomas, are the third most common pediatric brain tumor. The disease remains incurable for about 45% of patients even after gross total resection and radiotherapy. Despite showing a very homogeneous histological picture, ependymomas display distinct molecular behavior, which supports the existence of several independent entities of the disease. We examined two non-overlapping cohorts of 102 and 75 ependymomas by mRNA expression profiling, on two different array platforms (Affymetrix, Agilent). When performing multiple statistical clustering methods (unsupervised consensus NMF and consensus HCL), we could consistently identify three major clusters, including two subgroups of posterior fossa (PF) ependymoma, a variant common in children and associated with heterogeneous clinical outcome. Subgroup-specific chromosome aberrations of PF tumors were detected by aCGH, and biological signaling pathways distinguishing PF subgroups were identified by gene set enrichment analysis and visualized in Cytoscape. We validated the most significantly classifying markers of each subgroup by immunohistochemistry on a tissue microarray containing an independent set of 265 PF ependymomas. Our findings delineate two subgroups of PF ependymoma (groups A and B) which are demographically, transcriptionally, genetically, and clinically distinct. Group A patients are younger, have laterally located tumors with a balanced genome, more frequently develop secondary metastases and are much more likely to have an extremely poor outcome as compared with group B patients. Based on a multi-variate Cox proportional-hazards model, our identified markers have the strongest independent prognostic value among demographic and molecular variables with Hazard ratios of 8.45 (PFS) and 10.55 (OS). Prognostic significance and predictive impact is being validated in the GPOH HIT2000 Ependymoma study. The identification of two distinct subgroups of PF ependymoma, and markers applicable for their clinical distinction, will allow for better prognostication of individual cases, independent of age, level of resection and WHO grade, and also for stratification in future ependymoma clinical trials. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1432. doi:1538-7445.AM2012-1432
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 14_Supplement ( 2020-07-15), p. A25-A25
    Abstract: Introduction: The majority of pediatric supratentorial (ST) ependymomas (EPN) is driven by distinct gene fusions between C11orf95 and RELA. The resultant molecular group of ST-EPN-RELA tumors is characterized by constitutive activation of NF-κB signaling and deregulation of the p53 pathway. In contrast to surgery and radiotherapy, chemotherapy has failed to demonstrate significant benefit in the management of affected children. Alternative strategies including enhanced drug delivery, combination treatments, or application of new selective compounds are needed to tackle this disease. Material and Methods: RNAi and drug screening methods were applied to identify potential therapeutic approaches using ST-EPN-RELA cell lines. In order to identify optimal dosing strategies of selected drugs and to assess effects of combinatorial treatment approaches on blood-brain barrier (BBB) penetration, cerebral microdialysis combined with ultraperformance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS) was applied. This approach allowed for exact, continuous, and time-dependent drug quantification in tumors or healthy tissue in freely moving experimental mice. Patient-derived xenograft models of ST-EPN-RELA were treated to investigate toxicity and outcome parameters. Results: Regulation of p53 signaling and nuclear protein shuttling were identified as promising therapeutic approaches. While low-dose dactinomycin could successfully reestablish p53 function in ST-EPN-RELA cells in vitro, penetration of the drug across the BBB was found to be very poor and did not result in a survival benefit of tumor-bearing mice. Preliminary results of alternative strategies such as combination with efflux pump inhibitors, liposomal packaging, and inhibition of XPO1 being the sole nuclear exporter of p53 hold promise to overcome these constraints. Conclusion: Oncogenic dependencies of ST-EPN-RELA are currently difficult to target. Preclinical evaluation of effective drug disposition combined with long-term treatment studies may help to better select promising compounds and thereby increase success rates of early clinical trials in patients with ST-EPN-RELA in the future. Citation Format: Julia Benzel, Max Sauter, Norman Mack, Abigail Davis, Johanna Weiss, Philipp Uhl, Jürgen Burhenne, Kendra K. Maass, Jens-Martin Hübner, Hendrik Witt, Anang Shelat, Amar Gajjar, Santhosh A. Upadhyaya, Aylin Camgoz, Frank Buchholz, Sina Oppermann, Marcel Kool, Daisuke Kawauchi, Olaf Witt, Walter E. Haefeli, Stefan M. Pfister, Clinton Stewart, Kristian W. Pajtler. Evaluation of Drug Disposition in Supratentorial Ependymoma [abstract]. In: Proceedings of the AACR Special Conference on the Advances in Pediatric Cancer Research; 2019 Sep 17-20; Montreal, QC, Canada. Philadelphia (PA): AACR; Cancer Res 2020;80(14 Suppl):Abstract nr A25.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 31-31
    Abstract: Intracranial ependymoma comprises the second most common malignant brain tumor in childhood. The prognosis of these tumors remains generally poor and its biological behavior is unpredictable based on current stratification approaches. Neither clinical variables nor histopathological grading or molecular markers have so far been successful in defining a well circumscribed group of high-risk patients. Thus, an innovative staging model for ependymoma is desperately needed. We studied 122 samples from patients with intracranial ependymoma with a median follow-up of circa 8 years by genome-wide assessment of DNA copy-number aberrations using array-CGH (10K BAC array). Aberrations with a potential prognostic value were validated in an independent cohort of 170 patients by FISH analysis. Consecutively, we investigated genome-wide mRNA expression profiling (Agilent 44k) in 65 primary ependymomas and performed unsupervised clustering to identify potential transcriptome-based subgroups. We compared these findings with the previously identified DNA copy-number profiles. For validation of single molecular markers, selected candidate genes were investigated by QRT-PCR on transcriptional level, and protein expression was measured by immunohistochemistry on tissue microarrays (n=170). We were able to define a novel molecular staging system comprised of three genetically distinct subgroups of ependymoma based on DNA copy-number aberrations: i) a low risk group (34% of patients) including tumors with gain of chromosomes 9, 15q, 18, or loss of chromosome 6, or a combination thereof with patients showing a 5-year OS of 100%; ii) an intermediate risk group (41% of patients) characterized by a balanced cytogenetic profile especially for aberrations of chromosomes 1q, 9, 15q, 18, 6 and without a homozygous deletion of CDKN2A which was associated with a 5-year OS of 77%; iii) a high risk group (25% of patients) defined by tumors harbouring a gain of 1q and/or a homozygous deletion of CDKN2A, which was concurrent with a 5-year OS of only 33%. Interestingly, these cytogenetic risk-groups showed a significant overlap with transcriptome-based subgroups identified by unsupervised clustering. Thus, we aimed at the identification of interesting candidate genes which show subgroup-specific expression and have the potential to be used as surrogate marker for certain biological subgroups. The most robust subgroup-specific molecular markers for poor and good outcome were SHC1 and WDR16, respectively. In summary, we could decipher a novel stratification model for intracranial ependymoma consisting of three subgroups based on cytogenetic aberrations. By integrative genomics looking at DNA aberrations and mRNA levels in a large subset of samples, we were able to identify novel biomarkers in ependymoma, which have high potential to be useful for stratifying patients in future clinical trials. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 31.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3458-3458
    Abstract: Pilocytic astrocytoma (PA) comprises the most common primary brain tumor in children. Although this WHO grade I neoplasm shows a relatively benign biologic behavior many patients suffer from therapy-related long term neurologic impairment and up to 20% of patients experience tumor recurrence. Due to the slow growing nature of PA, adjuvant radiation and chemotherapy are only moderately effective. In order to pre-clinically evaluate novel therapy approaches, we aimed to establish a murine model of these tumors. Based on our previous work which showed that the MAPK intermediate BRAF is altered by duplication, activating mutation (V600E) or translocation of its kinase domain in a vast majority of cases, we applied somatic gene transfer using the Replication-Competent ASLV long terminal repeat (LTR) with a Splice acceptor (RCAS)/Tv-a technique to ectopically express different BRAF variants in Nestin-positive neural progenitors in the brains of newborn mice. While expression of either the BRAF i) full length wildtype, ii) full length V600E or iii) isolated kinase domain wildtype did not induce neoplastic lesions in mice, expression of the iv) isolated V600E mutated kinase domain induced tumors either in the cerebral hemispheres or in the brainstem depending on the site of injection. Although showing no clinical symptoms after an observation period of 4 months, post mortem analysis revealed presence of tumors in & gt;90% of cases. By histopathologic analysis, presence of strongly GFAP-positive lesions with a proliferation index (Ki67) below 1% could be identified. The slow tumor growth compared to other tumors induced with this technique together with presence of piloid-like tumor cells and eosinophilic structures (Rosenthal fibers) closely recapitulate clinical and biological features of human PA. Further analysis using primary murine Ntv-a astrocytes transduced with either of the four constructs in vitro revealed that expression of the isolated BRAF V600E kinase domain led to a twofold increase in proliferation compared to full length BRAF V600E or wild type BRAF kinase domain, while cells transduced with either full length wild type BRAF or GFP (control) showed almost no proliferation. Furthermore, this increase in proliferation could be markedly decreased by in vitro treatment with the kinase inhibitor Sorafenib at a concentration of 5 µM. This could be correlated with the amount of phosphorylation of the MAP kinases MEK and ERK, the downstream targets of BRAF. Treatment of mice with Sorafenib upon tumor induction is currently being conducted using ultrahigh field magnetic resonance imaging to monitor tumor growth and therapy response. Taken together, our data provide first in vivo evidence for the oncogenic role of BRAF in pediatric pilocytic astrocytomas and it will be of paramount clinical importance to use this first animal model for pre-clinical testing of novel treatment approaches. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3458. doi:10.1158/1538-7445.AM2011-3458
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages