scispace - formally typeset
Search or ask a question

Showing papers by "Jo Vandesompele published in 2012"


Journal ArticleDOI
Martin Peifer1, Lynnette Fernandez-Cuesta1, Martin L. Sos1, Julie George1, Danila Seidel1, Lawryn H. Kasper, Dennis Plenker1, Frauke Leenders1, Ruping Sun2, Thomas Zander1, Roopika Menon3, Mirjam Koker1, Ilona Dahmen1, Christian Müller1, Vincenzo Di Cerbo2, Hans Ulrich Schildhaus1, Janine Altmüller1, Ingelore Baessmann1, Christian Becker1, Bram De Wilde4, Jo Vandesompele4, Diana Böhm3, Sascha Ansén1, Franziska Gabler1, Ines Wilkening1, Stefanie Heynck1, Johannes M. Heuckmann1, Xin Lu1, Scott L. Carter5, Kristian Cibulskis5, Shantanu Banerji5, Gad Getz5, Kwon-Sik Park6, Daniel Rauh7, Christian Grütter7, Matthias Fischer1, Laura Pasqualucci8, Gavin M. Wright9, Zoe Wainer9, Prudence A. Russell10, Iver Petersen11, Yuan Chen11, Erich Stoelben, Corinna Ludwig, Philipp A. Schnabel, Hans Hoffmann, Thomas Muley, Michael Brockmann, Walburga Engel-Riedel, Lucia Anna Muscarella12, Vito Michele Fazio12, Harry J.M. Groen13, Wim Timens13, Hannie Sietsma13, Erik Thunnissen14, Egber Smit14, Daniëlle A M Heideman14, Peter J.F. Snijders14, Federico Cappuzzo, C. Ligorio15, Stefania Damiani15, John K. Field16, Steinar Solberg17, Odd Terje Brustugun17, Marius Lund-Iversen17, Jörg Sänger, Joachim H. Clement11, Alex Soltermann18, Holger Moch18, Walter Weder18, Benjamin Solomon19, Jean-Charles Soria20, Pierre Validire, Benjamin Besse20, Elisabeth Brambilla21, Christian Brambilla21, Sylvie Lantuejoul21, Philippe Lorimier21, Peter M. Schneider1, Michael Hallek1, William Pao22, Matthew Meyerson23, Matthew Meyerson5, Julien Sage6, Jay Shendure24, Robert Schneider2, Robert Schneider25, Reinhard Büttner1, Jürgen Wolf1, Peter Nürnberg1, Sven Perner3, Lukas C. Heukamp1, Paul K. Brindle, Stefan A. Haas2, Roman K. Thomas1 
TL;DR: This study implicates histone modification as a major feature of SCLC, reveals potentially therapeutically tractable genomic alterations and provides a generalizable framework for the identification of biologically relevant genes in the context of high mutational background.
Abstract: Small-cell lung cancer (SCLC) is an aggressive lung tumor subtype with poor prognosis(1-3). We sequenced 29 SCLC exomes, 2 genomes and 15 transcriptomes and found an extremely high mutation rate of 7.4 +/- 1 protein-changing mutations per million base pairs. Therefore, we conducted integrated analyses of the various data sets to identify pathogenetically relevant mutated genes. In all cases, we found evidence for inactivation of TP53 and RB1 and identified recurrent mutations in the CREBBP, EP300 and MLL genes that encode histone modifiers. Furthermore, we observed mutations in PTEN, SLIT2 and EPHA7, as well as focal amplifications of the FGFR1 tyrosine kinase gene. Finally, we detected many of the alterations found in humans in SCLC tumors from Tp53 and Rb1 double knockout mice(4). Our study implicates histone modification as a major feature of SCLC, reveals potentially therapeutically tractable genomic alterations and provides a generalizable framework for the identification of biologically relevant genes in the context of high mutational background.

1,177 citations


Journal ArticleDOI
TL;DR: It is reported that LIN28B showed genomic aberrations and extensive overexpression in high-risk neuroblastoma compared to several other tumor entities and normal tissues and was an independent risk factor for adverse outcome in neuroblastomas.
Abstract: LIN28B regulates developmental processes by modulating microRNAs (miRNAs) of the let-7 family. A role for LIN28B in cancer has been proposed but has not been established in vivo. Here, we report that LIN28B showed genomic aberrations and extensive overexpression in high-risk neuroblastoma compared to several other tumor entities and normal tissues. High LIN28B expression was an independent risk factor for adverse outcome in neuroblastoma. LIN28B signaled through repression of the let-7 miRNAs and consequently resulted in elevated MYCN protein expression in neuroblastoma cells. LIN28B-let-7-MYCN signaling blocked differentiation of normal neuroblasts and neuroblastoma cells. These findings were fully recapitulated in a mouse model in which LIN28B expression in the sympathetic adrenergic lineage induced development of neuroblastomas marked by low let-7 miRNA levels and high MYCN protein expression. Interference with this pathway might offer therapeutic perspectives.

333 citations


Journal ArticleDOI
13 Jun 2012-Leukemia
TL;DR: These findings distinguish LSD1 as a critical regulator of hematopoiesis and point to severe, but reversible, side effects of a LSD1-targeted therapy.
Abstract: Lysine (K)-specific demethylase 1A (LSD1/KDM1A) has been identified as a potential therapeutic target in solid cancers and more recently in acute myeloid leukemia. However, the potential side effects of a LSD1-inhibitory therapy remain elusive. Here, we show, with a newly established conditional in vivo knockdown model, that LSD1 represents a central regulator of hematopoietic stem and progenitor cells. LSD1 knockdown (LSD1-kd) expanded progenitor numbers by enhancing their proliferative behavior. LSD1-kd led to an extensive expansion of granulomonocytic, erythroid and megakaryocytic progenitors. In contrast, terminal granulopoiesis, erythropoiesis and platelet production were severely inhibited. The only exception was monopoiesis, which was promoted by LSD1 deficiency. Importantly, we showed that peripheral blood granulocytopenia, monocytosis, anemia and thrombocytopenia were reversible after LSD1-kd termination. Extramedullary splenic hematopoiesis contributed to the phenotypic reversion, and progenitor populations remained expanded. LSD1-kd was associated with the upregulation of key hematopoietic genes, including Gfi1b, Hoxa9 and Meis1, which are known regulators of the HSC/progenitor compartment. We also demonstrated that LSD1-kd abrogated Gfi1b-negative autoregulation by crossing LSD1-kd with Gfi1b:GFP mice. Taken together, our findings distinguish LSD1 as a critical regulator of hematopoiesis and point to severe, but reversible, side effects of a LSD1-targeted therapy.

178 citations


Journal ArticleDOI
TL;DR: It is concluded that an activating mutation within the ALK kinase domain is sufficient to induce neuroblastoma development, and ALK inhibitors show promise for treating human neuroblastomas harboring ALK mutations.
Abstract: Activating anaplastic lymphoma kinase (ALK) mutations were recently detected in most familial and 10% of sporadic neuroblastomas. However, the role of mutated ALK in tumorigenesis remains elusive. We demonstrate that targeted expression of the most frequent and aggressive variant, ALK(F1174L), is tumorigenic in mice. Tumors resembled human neuroblastomas in morphology, metastasis pattern, gene expression, and the presence of neurosecretory vesicles as well as synaptic structures. This ALK-driven neuroblastoma mouse model precisely recapitulated the genetic spectrum of the disease. Chromosomal aberrations were syntenic to those in human neuroblastoma, including 17q gain and MYCN oncogene amplification. Targeted ALK(F1174L) and MYCN coexpression revealed a strong synergism in inducing neuroblastoma with minimal chromosomal aberrations, suggesting that fewer secondary hits are required for tumor induction if both oncoproteins are targeted. Treatment of ALK(F1174L) transgenic mice with the ALK inhibitor TAE-684 induced complete tumor regression, indicating that tumor cells were addicted to ALK(F1174L) activity. We conclude that an activating mutation within the ALK kinase domain is sufficient to induce neuroblastoma development, and ALK inhibitors show promise for treating human neuroblastomas harboring ALK mutations.

159 citations


Book ChapterDOI
TL;DR: The power of the previously described global meannormalization method is summarized in comparison to the multiple reference gene normalization method using the most stably expressed small RNA controls and a modified global mean normalization strategy based on the attribution of equal weight to each individual miRNA during normalization is compared.
Abstract: MicroRNAs (miRNAs) are an important class of gene regulators, acting on several aspects of cellular function such as differentiation, cell cycle control, and stemness. These master regulators constitute an invaluable source of biomarkers, and several miRNA signatures correlating with patient diagnosis, prognosis, and response to treatment have been identified. Within this exciting field of research, whole-genome RT-qPCR-based miRNA profiling in combination with a global mean normalization strategy has proven to be the most sensitive and accurate approach for high-throughput miRNA profiling (Mestdagh et al., Genome Biol 10:R64, 2009). In this chapter, we summarize the power of the previously described global mean normalization method in comparison to the multiple reference gene normalization method using the most stably expressed small RNA controls. In addition, we compare the original global mean method to a modified global mean normalization strategy based on the attribution of equal weight to each individual miRNA during normalization. This modified algorithm is implemented in Biogazelle's qbasePLUS software and is presented here for the first time.

153 citations


Journal ArticleDOI
TL;DR: It is shown that Dicer1 targeting prevents retinoblastoma formation in mice by synthetic lethality with combined inactivation of p53 and Rb, and a proof-of-concept that targeting miRNAs may potentially represent a general approach for synthetic lethal targeting of cancer cells that harbour specific cancer-inducing genotypes.
Abstract: Synthetic lethality is a promising strategy for specific targeting of cancer cells that carry mutations that are absent in normal cells. This approach may help overcome the challenge associated with targeting dysfunctional tumour suppressors, such as p53 and Rb (refs 1, 2). Here we show that Dicer1 targeting prevents retinoblastoma formation in mice by synthetic lethality with combined inactivation of p53 and Rb. Although Dicer1 functions as a haploinsufficient tumour suppressor, its complete loss of function is selected against during tumorigenesis(3-5). We show that Dicer1 deficiency is tolerated in Rb-deficient retinal progenitor cells harbouring an intact p53 pathway, but not in the absence of p53. This synthetic lethality is mediated by the oncogenic miR-17-92 cluster because its deletion phenocopies Dicer1 loss in this context. miR-17-92 inactivation suppresses retinoblastoma formation in mice and co-silencing of miR-17/20a and p53 cooperatively decreases the viability of human retinoblastoma cells. These data provide an explanation for the selective pressure against loss of Dicer1 during tumorigenesis and a proof-of-concept that targeting miRNAs may potentially represent a general approach for synthetic lethal targeting of cancer cells that harbour specific cancer-inducing genotypes.

76 citations


Journal ArticleDOI
TL;DR: In this article, the authors applied two independent genome-wide methylation screening methodologies to eight neuroblastoma cell lines to identify prognostic tumor DNA methylation biomarkers, which yielded 43 candidate biomarkers that were subsequently tested by high-throughput methylation-specific PCR.
Abstract: Background: Accurate outcome prediction in neuroblastoma, which is necessary to enable the optimal choice of risk-related therapy, remains a challenge. To improve neuroblastoma patient stratification, this study aimed to identify prognostic tumor DNA methylation biomarkers. Results: To identify genes silenced by promoter methylation, we first applied two independent genome-wide methylation screening methodologies to eight neuroblastoma cell lines. Specifically, we used re-expression profiling upon 5-aza-2’-deoxycytidine (DAC) treatment and massively parallel sequencing after capturing with a methyl-CpG-binding domain (MBD-seq). Putative methylation markers were selected from DAC-upregulated genes through a literature search and an upfront methylation-specific PCR on 20 primary neuroblastoma tumors, as well as through MBD- seq in combination with publicly available neuroblastoma tumor gene expression data. This yielded 43 candidate biomarkers that were subsequently tested by high-throughput methylation-specific PCR on an independent cohort of 89 primary neuroblastoma tumors that had been selected for risk classification and survival. Based on this analysis, methylation of KRT19, FAS, PRPH, CNR1, QPCT, HIST1H3C, ACSS3 and GRB10 was found to be associated with at least one of the classical risk factors, namely age, stage or MYCN status. Importantly, HIST1H3C and GNAS methylation was associated with overall and/or event-free survival. Conclusions: This study combines two genome-wide methylation discovery methodologies and is the most extensive validation study in neuroblastoma performed thus far. We identified several novel prognostic DNA methylation markers and provide a basis for the development of a DNA methylation-based prognostic classifier in neuroblastoma.

66 citations


Journal ArticleDOI
22 Mar 2012-Oncogene
TL;DR: It is concluded that FOXR1 is recurrently activated in neuroblastoma by intrachromosomal deletion/fusion events, resulting in overexpression of fusion transcripts.
Abstract: Neuroblastoma tumors frequently show loss of heterozygosity of chromosome 11q with a shortest region of overlap in the 11q23 region. These deletions are thought to cause inactivation of tumor suppressor genes leading to haploinsufficiency. Alternatively, micro-deletions could lead to gene fusion products that are tumor driving. To identify such events we analyzed a series of neuroblastomas by comparative genomic hybridization and single-nucleotide polymorphism arrays and integrated these data with Affymetrix mRNA profiling data with the bioinformatic tool R2 (http://r2.amc.nl). We identified three neuroblastoma samples with small interstitial deletions at 11q23, upstream of the forkhead-box R1 transcription factor (FOXR1). Genes at the proximal side of the deletion were fused to FOXR1, resulting in fusion transcripts of MLL-FOXR1 and PAFAH1B2-FOXR1. FOXR1 expression has only been detected in early embryogenesis. Affymetrix microarray analysis showed high FOXR1 mRNA expression exclusively in the neuroblastomas with micro-deletions and rare cases of other tumor types, including osteosarcoma cell line HOS. RNAi silencing of FOXR1 strongly inhibited proliferation of HOS cells and triggered apoptosis. Expression profiling of these cells and reporter assays suggested that FOXR1 is a negative regulator of fork-head box factor-mediated transcription. The neural crest stem cell line JoMa1 proliferates in culture conditional to activity of a MYC-ER transgene. Over-expression of the wild-type FOXR1 could functionally replace MYC and drive proliferation of JoMa1. We conclude that FOXR1 is recurrently activated in neuroblastoma by intrachromosomal deletion/fusion events, resulting in overexpression of fusion transcripts. Forkhead-box transcription factors have not been previously implicated in neuroblastoma pathogenesis. Furthermore, this is the first identification of intrachromosomal fusion genes in neuroblastoma. Oncogene (2012) 31, 1571-1581; doi:10.1038/onc.2011.344; published online 22 August 2011

61 citations


Journal ArticleDOI
TL;DR: An assay that holds great promise as a tool for screening patients with familial autosomal recessive nonsyndromal hearing loss (ARNSHL), and for the first time, an efficient, reliable and cost effective genetic test, based on PCR enrichment, for newborns with undiagnosed deafness is available.
Abstract: Background: Hereditary hearing loss (HL) can originate from mutations in one of many genes involved in the complex process of hearing. Identification of the genetic defects in patients is currently labor intensive and expensive. While screening with Sanger sequencing for GJB2 mutations is common, this is not the case for the other known deafness genes (> 60). Next generation sequencing technology (NGS) has the potential to be much more cost efficient. Published methods mainly use hybridization based target enrichment procedures that are time saving and efficient, but lead to loss in sensitivity. In this study we used a semi-automated PCR amplification and NGS in order to combine high sensitivity, speed and cost efficiency. Results: In this proof of concept study, we screened 15 autosomal recessive deafness genes in 5 patients with congenital genetic deafness. 646 specific primer pairs for all exons and most of the UTR of the 15 selected genes were designed using primerXL. Using patient specific identifiers, all amplicons were pooled and analyzed using the Roche 454 NGS technology. Three of these patients are members of families in which a region of interest has previously been characterized by linkage studies. In these, we were able to identify two new mutations in CDH23 and OTOF. For another patient, the etiology of deafness was unclear, and no causal mutation was found. In a fifth patient, included as a positive control, we could confirm a known mutation in TMC1. Conclusions: We have developed an assay that holds great promise as a tool for screening patients with familial autosomal recessive nonsyndromal hearing loss (ARNSHL). For the first time, an efficient, reliable and cost effective genetic test, based on PCR enrichment, for newborns with undiagnosed deafness is available.

57 citations


Journal ArticleDOI
TL;DR: An approach in which cDNA synthesis is carried out on crude cell lysates instead of on purified RNA samples can offer a fast and straightforward alternative to the classic workflow of RNA purification and c DNA synthesis.
Abstract: Gene expression quantification on cultured cells using the reverse transcription quantitative polymerase chain reaction (RT-qPCR) typically involves an RNA purification step that limits sample processing throughput and precludes parallel analysis of large numbers of samples. An approach in which cDNA synthesis is carried out on crude cell lysates instead of on purified RNA samples can offer a fast and straightforward alternative. Here, we evaluate such an approach, benchmarking Ambion’s Cells-to-CT kit with the classic workflow of RNA purification and cDNA synthesis, and demonstrate its good accuracy and superior sensitivity.

54 citations


Journal ArticleDOI
TL;DR: A novel targeted MPS-based approach is proposed that is suitable for accurate, fast, and cost-effective early molecular testing in LCA, and easily applicable in other genetic disorders.

Journal ArticleDOI
TL;DR: It is demonstrated that the MYCN‐induced downregulation of DKK3 results from direct upregulation of miR‐17‐92 components effecting both DKK 3 mRNA stability and translation which further contributes to the pleiotropic oncogenic effect of elevated MYCN levels.
Abstract: Neuroblastoma (NB) is a paediatric tumour with a remarkable diverse clinical behaviour. Approximately half of the high stage aggressive tumours are characterized by MYCN gene amplification but our understanding of the role of MYCN in NB oncogenesis is incomplete. Previous studies have shown that MYCN expression is inversely correlated with expression of Dickkopf-3 (DKK3), a gene encoding an extracellular protein with presumed tumour suppressor activity, but direct MYCN regulation of DKK3 was excluded leaving the mechanism of regulation unexplained. Given the recently established role of MYCN-regulated miRNAs in downregulation of protein-coding genes and predicted seeds for miR-17-92 cluster members within the DKK3 3 0 UTR, we hypothesized that this mechanism would act in MYCN regulation of DKK3. To investigate this, we used a validated miR-17-92-inducible cellular system and could demonstrate robust downregulation of DKK3 mRNA and protein levels upon miR-17-92 overexpression. Next, two of the three predicted miRNAs, miR-19b and miR-92a, were shown to lower DKK3 protein levels, in addition to measurable DKK3 mRNA knock-down by miR-92a. Direct interaction between miR-19b or miR-92a and the 3 0 UTR of DKK3 was validated using luciferase reporter assays. In conclusion, this study demonstrates that the MYCN-induced downregulation of DKK3 results from direct upregulation of miR-17-92 components effecting both DKK3 mRNA stability and translation which further contributes to the pleiotropic oncogenic effect of elevated MYCN levels. The strict MYCN-mediated regulation of DKK3 is suggestive for an important downstream function of the MYCN protein and thus warrants further investigations to unravel the role of DKK3 in NB. Neuroblastoma (NB) is a paediatric tumour with a worldwide incidence of 10 cases per million in children younger than 15 years old. 1 NB originates from sympathetic neuronal progenitors and is characterized by a remarkable diverse clinical behaviour ranging from spontaneously regressing to highly aggressive metastasized tumours. Amplification of MYCN occurs with a high incidence in aggressive tumour phenotype and, furthermore, MYCN status can be used as an independent predictor of poor prognosis. MYCN is member of the

Journal ArticleDOI
TL;DR: This finding further contributes to the delineation of the genomic profile of aggressive neuroblastoma, offers perspectives for the identification of genes contributing to the disease phenotype and may be relevant in the light of assessment of response to new molecular treatments.
Abstract: Neuroblastoma is an aggressive embryonal tumor that accounts for ∼15% of childhood cancer deaths. Hitherto, despite the availability of comprehensive genomic data on DNA copy number changes in neuroblastoma, relatively little is known about the genes driving neuroblastoma tumorigenesis. In this study, high resolution array comparative genome hybridization (CGH) was performed on 188 primary neuroblastoma tumors and 33 neuroblastoma cell lines to search for previously undetected recurrent DNA copy number gains and losses. A new recurrent distal chromosome 1q deletion (del(1)(q42.2qter)) was detected in seven cases. Further analysis of available array CGH datasets revealed 13 additional similar distal 1q deletions. The majority of all detected 1q deletions was found in high risk 11q deleted tumors without MYCN amplification (Fisher exact test p = 5.61 × 10(-5) ). Using ultra-high resolution (∼115 bp resolution) custom arrays covering the breakpoints on 1q for 11 samples, clustering of nine breakpoints was observed within a 12.5-kb region, of which eight were found in a 7-kb copy number variable region, whereas the remaining two breakpoints were colocated 1.4-Mb proximal. The commonly deleted region contains one miRNA (hsa-mir-1537), four transcribed ultra conserved region elements (uc.43-uc.46) and 130 protein coding genes including at least two bona fide tumor suppressor genes, EGLN1 (or PHD2) and FH. This finding further contributes to the delineation of the genomic profile of aggressive neuroblastoma, offers perspectives for the identification of genes contributing to the disease phenotype and may be relevant in the light of assessment of response to new molecular treatments.

Journal ArticleDOI
15 Feb 2012-PLOS ONE
TL;DR: The results suggest that N-cadherin signaling may play a role in neuroblastoma disease, marking involvement of metastasis and determining Neuroblastoma cell viability.
Abstract: One of the first and most important steps in the metastatic cascade is the loss of cell-cell and cell-matrix interactions. N-cadherin, a crucial mediator of homotypic and heterotypic cell-cell interactions, might play a central role in the metastasis of neuroblastoma (NB), a solid tumor of neuroectodermal origin. Using Reverse Transcription Quantitative PCR (RT-qPCR), Western blot, immunocytochemistry and Tissue MicroArrays (TMA) we demonstrate the expression of N-cadherin in neuroblastoma tumors and cell lines. All neuroblastic tumors (n = 356) and cell lines (n = 10) expressed various levels of the adhesion protein. The N-cadherin mRNA expression was significantly lower in tumor samples from patients suffering metastatic disease. Treatment of NB cell lines with the N-cadherin blocking peptide ADH-1 (Exherin, Adherex Technologies Inc.), strongly inhibited tumor cell proliferation in vitro by inducing apoptosis. Our results suggest that N-cadherin signaling may play a role in neuroblastoma disease, marking involvement of metastasis and determining neuroblastoma cell viability.

Journal ArticleDOI
TL;DR: This review intends to provide a road map in the jungle of publications of miRNA in colorectal cancer, focusing on data availability and new ways to generate biologically relevant information out of these huge amounts of data.

Journal ArticleDOI
TL;DR: As exon level-based prediction yields comparable, but not significantly better, prediction accuracy than gene expression-based predictors, gene-based assays seem to be sufficiently precise for predicting outcome of neuroblastoma patients.
Abstract: Using mRNA expression-derived signatures as predictors of individual patient outcome has been a goal ever since the introduction of microarrays. Here, we addressed whether analyses of tumour mRNA at the exon level can improve on the predictive power and classification accuracy of gene-based expression profiles using neuroblastoma as a model. In a patient cohort comprising 113 primary neuroblastoma specimens expression profiling using exon-level analyses was performed to define predictive signatures using various machine-learning techniques. Alternative transcript use was calculated from relative exon expression. Validation of alternative transcripts was achieved using qPCR- and cell-based approaches. Both predictors derived from the gene or the exon levels resulted in prediction accuracies >80% for both event-free and overall survival and proved as independent prognostic markers in multivariate analyses. Alternative transcript use was most prominently linked to the amplification status of the MYCN oncogene, expression of the TrkA/NTRK1 neurotrophin receptor and survival. As exon level-based prediction yields comparable, but not significantly better, prediction accuracy than gene expression-based predictors, gene-based assays seem to be sufficiently precise for predicting outcome of neuroblastoma patients. However, exon-level analyses provide added knowledge by identifying alternative transcript use, which should deepen the understanding of neuroblastoma biology.

Book ChapterDOI
TL;DR: This chapter presents different RT-qPCR technologies that enable whole genome miRNA expression quantification and describes the different approaches used to achieve this goal.
Abstract: MicroRNAs (miRNAs) are small noncoding RNA molecules that function as negative regulators of gene expression. They are essential components of virtually every biological process and deregulated miRNA expression has been reported in a multitude of human diseases including cancer. Owing to their small size (20–22 nucleotides), accurate quantification of miRNA expression is particularly challenging. In this chapter, we present different RT-qPCR technologies that enable whole genome miRNA expression quantification.

Journal ArticleDOI
01 Mar 2012-PLOS ONE
TL;DR: A gene prioritization strategy based on the fitSNP scores appears to be most successful in identifying mutated cancer genes across different tumor entities, with variance of gene expression levels as a good second best.
Abstract: Background Although the throughput of next generation sequencing is increasing and at the same time the cost is substantially reduced, for the majority of laboratories whole genome sequencing of large cohorts of cancer samples is still not feasible. In addition, the low number of genomes that are being sequenced is often problematic for the downstream interpretation of the significance of the variants. Targeted resequencing can partially circumvent this problem; by focusing on a limited number of candidate cancer genes to sequence, more samples can be included in the screening, hence resulting in substantial improvement of the statistical power. In this study, a successful strategy for prioritizing candidate genes for targeted resequencing of cancer genomes is presented. Results Four prioritization strategies were evaluated on six different cancer types: genes were ranked using these strategies, and the positive predictive value (PPV) or mutation rate within the top-ranked genes was compared to the baseline mutation rate in each tumor type. Successful strategies generate gene lists in which the top is enriched for known mutated genes, as evidenced by an increase in PPV. A clear example of such an improvement is seen in colon cancer, where the PPV is increased by 2.3 fold compared to the baseline level when 100 top fitSNP genes are sequenced. Conclusions A gene prioritization strategy based on the fitSNP scores appears to be most successful in identifying mutated cancer genes across different tumor entities, with variance of gene expression levels as a good second best.

Proceedings ArticleDOI
TL;DR: The results of an unprecedented integrated analysis aimed at identifying networks of highly co-regulated miRNA-lncRNA-mRNA clusters centered around key cancer genes are presented and the complex interplay is described.
Abstract: With the discovery of different non-coding RNA species, the complexity of the transcriptome and its regulation has increased dramatically. Next to the well-studied small non-coding miRNAs, several thousands of long non-coding RNAs (lncRNAs) have recently been described. Like miRNAs, lncRNAs appear to predominantly function as regulators of gene expression and are implicated in various regulatory networks involving both miRNAs and protein-coding genes. In order to facilitate the search for miRNA-lncRNA-mRNA regulatory networks in cancer we have profiled the expression of each of these RNA information layers using the high-throughput SmartChip RT-qPCR technology on the entire NCI60 cancer cell line panel. In total, three SmartChip Panels with 1050 miRNAs, 1250 cancer-focused mRNAs, and 1718 lncRNAs, respectively, were quantified with a minimum of 3 technical replicates against each cell line. Here, we present the results of an unprecedented integrated analysis aimed at identifying networks of highly co-regulated miRNA-lncRNA-mRNA clusters. In brief, individual clusters are annotated using a pathway enrichment approach whereby network edges are evaluated using miRNA target, lncRNA target, and transcription factor target predictions. MiRNA-lncRNA-mRNA clusters centered around key cancer genes are annotated and the complex interplay is described. This unique and extensive high quality dataset, comprised of three major information layers of the NCI60 cell line transcriptome offers numerous opportunities towards a better understanding of complex regulatory networks in cancer. 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 2987. doi:1538-7445.AM2012-2987


01 Jan 2012
TL;DR: This study, being the most extensive methylation study in NB performed thus far, has led to the discovery of several novel prognostic DNA- methylation markers, and is of great value for the development of a DNA-methylation-based prognostic classifier.
Abstract: Background: Due to the heterogenic biology of neuroblastoma (NB), current risk assessment schemes sometimes lead to misclassification of the patient with respect to therapy protocols. To reduce unnecessary under- or overtreatment and to improve the power of predictive outcome, additional tumor-specific prognostic markers are required. As several genes have already been shown to be silenced by hypermethylation of their promoter region in NB, this study aimed at identifying tumor DNA-methylation markers with prognostic power in order to improve NB patient stratification. Methods: To identify genes silenced by promoter methylation we applied two genome-wide methylation screening methodologies on 8 NB cell lines: a re-expression analysis using 5-aza-2'-deoxycytidine (DAC) and massively parallel sequencing after capturing with a methyl-CpG-binding domain (MBD). To select for prognostic markers, 212 genes upregulated after DAC-treatment were subsequently screened using methylation-specific PCR (MSP) on primary tumors. After integrated data analysis, 43 unique candidate biomarkers were further analyzed with MSP on 89 primary tumors, representing low- and high-risk surviving and deceased patients. Results: We identified 12 novel DNA-methylation markers (COL6A3, miR-1225, miR-3177, PCDHA6, PLXNC1, ANKRD43, ADRB2, APOE, miR-671, QPCT, KCND2 and PRPH), methylated in >50% of the primary tumors. Promoter methylation of DPP4, HIST1H3C and GNAS was found to be associated with overall and event-free survival, and the methylation status of TGFBI, TNFRSF10D, KRT19, TNFRSF10A, RARRES1, FAS, PRPH, CNR1, QPCT, HIST1H3C, ACSS3, GNAS and GRB10 with at least one of the classical risk factors, such as age, stage and MYCN status. Conclusions: This study, being the most extensive methylation study in NB performed thus far, has led to the discovery of several novel prognostic DNA-methylation markers, and is of great value for the development of a DNA-methylation-based prognostic classifier. In addition, methylation in microRNA (miRNA) promoter regions could be demonstrated in primary NB tumor samples for the first time.