Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Linda Schild is active.

Publication


Featured researches published by Linda Schild.


Nature | 2012

Sequencing of neuroblastoma identifies chromothripsis and defects in neuritogenesis genes

Jan J. Molenaar; Jan Koster; Danny A. Zwijnenburg; Peter van Sluis; Linda J. Valentijn; Ida van der Ploeg; Mohamed Hamdi; Johan van Nes; Bart A. Westerman; Jennemiek van Arkel; Marli E. Ebus; Franciska Haneveld; Arjan Lakeman; Linda Schild; Piet Molenaar; Peter Stroeken; Max M. van Noesel; Ingrid Øra; Evan E. Santo; Huib N. Caron; Ellen M. Westerhout; Rogier Versteeg

Neuroblastoma is a childhood tumour of the peripheral sympathetic nervous system. The pathogenesis has for a long time been quite enigmatic, as only very few gene defects were identified in this often lethal tumour. Frequently detected gene alterations are limited to MYCN amplification (20%) and ALK activations (7%). Here we present a whole-genome sequence analysis of 87 neuroblastoma of all stages. Few recurrent amino-acid-changing mutations were found. In contrast, analysis of structural defects identified a local shredding of chromosomes, known as chromothripsis, in 18% of high-stage neuroblastoma. These tumours are associated with a poor outcome. Structural alterations recurrently affected ODZ3, PTPRD and CSMD1, which are involved in neuronal growth cone stabilization. In addition, ATRX, TIAM1 and a series of regulators of the Rac/Rho pathway were mutated, further implicating defects in neuritogenesis in neuroblastoma. Most tumours with defects in these genes were aggressive high-stage neuroblastomas, but did not carry MYCN amplifications. The genomic landscape of neuroblastoma therefore reveals two novel molecular defects, chromothripsis and neuritogenesis gene alterations, which frequently occur in high-risk tumours.


Nature Genetics | 2015

Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations

Thomas F. Eleveld; Derek A. Oldridge; Virginie Bernard; Jan Koster; Leo Colmet Daage; Sharon J. Diskin; Linda Schild; Nadia Bessoltane Bentahar; Angela Bellini; Mathieu Chicard; Eve Lapouble; Valérie Combaret; Patricia Legoix-Né; Jean Michon; Trevor J. Pugh; Lori S. Hart; JulieAnn Rader; Edward F. Attiyeh; Jun S. Wei; Shile Zhang; Arlene Naranjo; Julie M. Gastier-Foster; Michael D. Hogarty; Shahab Asgharzadeh; Malcolm A. Smith; Jaime M. Guidry Auvil; Thomas B. K. Watkins; Danny A. Zwijnenburg; Marli E. Ebus; Peter van Sluis

The majority of patients with neuroblastoma have tumors that initially respond to chemotherapy, but a large proportion will experience therapy-resistant relapses. The molecular basis of this aggressive phenotype is unknown. Whole-genome sequencing of 23 paired diagnostic and relapse neuroblastomas showed clonal evolution from the diagnostic tumor, with a median of 29 somatic mutations unique to the relapse sample. Eighteen of the 23 relapse tumors (78%) showed mutations predicted to activate the RAS-MAPK pathway. Seven of these events were detected only in the relapse tumor, whereas the others showed clonal enrichment. In neuroblastoma cell lines, we also detected a high frequency of activating mutations in the RAS-MAPK pathway (11/18; 61%), and these lesions predicted sensitivity to MEK inhibition in vitro and in vivo. Our findings provide a rationale for genetic characterization of relapse neuroblastomas and show that RAS-MAPK pathway mutations may function as a biomarker for new therapeutic approaches to refractory disease.


Endocrine-related Cancer | 2011

Knockdown of survivin (BIRC5) causes apoptosis in neuroblastoma via mitotic catastrophe

Fieke Lamers; Ida van der Ploeg; Linda Schild; Marli E. Ebus; Jan Koster; Bo Hansen; Troels Koch; Rogier Versteeg; Huib N. Caron; Jan J. Molenaar

BIRC5 (survivin) is one of the genes located on chromosome arm 17q in the region that is often gained in neuroblastoma. BIRC5 is a protein in the intrinsic apoptotic pathway that interacts with XIAP and DIABLO leading to caspase-3 and caspase-9 inactivation. BIRC5 is also involved in stabilizing the microtubule-kinetochore dynamics. Based on the Affymetrix mRNA expression data, we here show that BIRC5 expression is strongly upregulated in neuroblastoma compared with normal tissues, adult malignancies, and non-malignant fetal adrenal neuroblasts. The over-expression of BIRC5 correlates with an unfavorable prognosis independent of the presence of 17q gain. Silencing of BIRC5 in neuroblastoma cell lines by various antisense molecules resulted in massive apoptosis as measured by PARP cleavage and FACS analysis. As both the intrinsic apoptotic pathway and the chromosomal passenger complex can be therapeutically targeted, we investigated in which of them BIRC5 exerted its essential anti-apoptotic role. Immunofluorescence analysis of neuroblastoma cells after BIRC5 silencing showed formation of multinucleated cells indicating mitotic catastrophe, which leads to apoptosis via P53 and CASP2. We show that BIRC5 silencing indeed resulted in activation of P53 and we could rescue apoptosis by CASP2 inhibition. We conclude that BIRC5 stabilizes the microtubules in the chromosomal passenger complex in neuroblastoma and that the apoptotic response results from mitotic catastrophe, which makes BIRC5 an interesting target for therapy.


European Journal of Cancer | 2012

Targeted BIRC5 silencing using YM155 causes cell death in neuroblastoma cells with low ABCB1 expression

Fieke Lamers; Linda Schild; Jan Koster; Rogier Versteeg; Huib N. Caron; Jan J. Molenaar

The BIRC5 (Survivin) gene is located at chromosome 17q in the region that is frequently gained in high risk neuroblastoma. BIRC5 is strongly over expressed in neuroblastoma tumour samples, which correlates to a poor prognosis. We recently validated BIRC5 as a potential therapeutic target by showing that targeted knock down with shRNAs triggers an apoptotic response through mitotic catastrophe. We now tested YM155, a novel small molecule selective BIRC5 suppressant that is currently in phase I/II clinical trials. Drug response curves showed IC50 values in the low nM range (median: 35 nM, range: 0.5-> 10,000 nM) in a panel of 23 neuroblastoma cell lines and four TIC-lines, which resulted from an apoptotic response. Nine out of 23 cell lines were relatively resistant to YM155 with IC50 values > 200 nM, although in the same cells shRNA mediated knock down of BIRC5 caused massive apoptosis. Analysis of differentially expressed genes between five most sensitive and five most resistant cell lines using Affymetrix mRNA expression data revealed ABCB1 (MDR1) as the most predictive gene for resistance to YM155. Inhibition of the multi-drug resistance pump ABCB1 with cyclosporine or knockdown with shRNA prior to treatment with YM155 demonstrated that cell lines with ABCB1 expression became 27-695 times more sensitive to YM155 treatment. We conclude that most neuroblastoma cell lines are sensitive to YM155 in the low nM range and that resistant cells can be sensitised by ABCB1 inhibitors. Therefore YM155 is a promising novel compound for treatment of neuroblastoma with low ABCB1 expression.


European Journal of Cancer | 2012

Targeted BCL2 inhibition effectively inhibits neuroblastoma tumour growth.

Fieke Lamers; Linda Schild; Ilona J.M. den Hartog; Marli E. Ebus; Ellen M. Westerhout; Ingrid Øra; Jan Koster; Rogier Versteeg; Huib N. Caron; Jan J. Molenaar

Genomic aberrations of key regulators of the apoptotic pathway have hardly been identified in neuroblastoma. We detected high BCL2 mRNA and protein levels in the majority of neuroblastoma tumours by Affymetrix expression profiling and Tissue Micro Array analysis. This BCL2 mRNA expression is strongly elevated compared to normal tissues and other malignancies. Most neuroblastoma cell lines lack this high BCL2 expression. Only two neuroblastoma cell lines (KCNR and SJNB12) show BCL2 expression levels representative for neuroblastoma tumours. To validate BCL2 as a therapeutic target in neuroblastoma we employed lentivirally mediated shRNA. Silencing of BCL2 in KCNR and SJNB12 resulted in massive apoptosis, while cell lines with low BCL2 expression were insensitive. Identical results were obtained by treatment of the neuroblastoma cell lines with the small molecule BCL2 inhibitor ABT263, which is currently being clinically evaluated. Combination assays of ABT263 with most classical cytostatics showed strong synergistic responses. Subcutaneous xenografts of a neuroblastoma cell line with high BCL2 expression in NMRI nu/nu mice showed a strong response to ABT263. These findings establish BCL2 as a promising drug target in neuroblastoma and warrant further evaluation of ABT263 and other BCL2 inhibiting drugs.


Oncotarget | 2016

High efficacy of the BCL-2 inhibitor ABT199 (venetoclax) in BCL-2 high-expressing neuroblastoma cell lines and xenografts and rational for combination with MCL-1 inhibition

Laurel T. Bate-Eya; Ilona J.M. den Hartog; Ida van der Ploeg; Linda Schild; Jan Koster; Evan E. Santo; Ellen M. Westerhout; Rogier Versteeg; Huib N. Caron; Jan J. Molenaar; M. Emmy M. Dolman

The anti-apoptotic protein B cell lymphoma/leukaemia 2 (BCL-2) is highly expressed in neuroblastoma and plays an important role in oncogenesis. In this study, the selective BCL-2 inhibitor ABT199 was tested in a panel of neuroblastoma cell lines with diverse expression levels of BCL-2 and other BCL-2 family proteins. ABT199 caused apoptosis more potently in neuroblastoma cell lines expressing high BCL-2 and BIM/BCL-2 complex levels than low expressing cell lines. Effects on cell viability correlated with effects on BIM displacement from BCL-2 and cytochrome c release from the mitochondria. ABT199 treatment of mice with neuroblastoma tumors expressing high BCL-2 levels only resulted in growth inhibition, despite maximum BIM displacement from BCL-2 and the induction of a strong apoptotic response. We showed that neuroblastoma cells might survive ABT199 treatment due to its acute upregulation of the anti-apoptotic BCL-2 family protein myeloid cell leukaemia sequence 1 (MCL-1) and BIM sequestration by MCL-1. In vitro inhibition of MCL-1 sensitized neuroblastoma cell lines to ABT199, confirming the pivotal role of MCL-1 in ABT199 resistance. Our findings suggest that neuroblastoma patients with high BCL-2 and BIM/BCL-2 complex levels might benefit from combination treatment with ABT199 and compounds that inhibit MCL-1 expression.


European Journal of Cancer | 2014

Newly-derived neuroblastoma cell lines propagated in serum-free media recapitulate the genotype and phenotype of primary neuroblastoma tumours

Laurel T. Bate-Eya; Marli E. Ebus; Jan Koster; Ilona J.M. den Hartog; Danny A. Zwijnenburg; Linda Schild; Ida van der Ploeg; M. Emmy M. Dolman; Huib N. Caron; Rogier Versteeg; Jan J. Molenaar

Recently protocols have been devised for the culturing of cell lines from fresh tumours under serum-free conditions in defined neural stem cell medium. These cells, frequently called tumour initiating cells (TICs) closely retained characteristics of the tumours of origin. We report the isolation of eight newly-derived neuroblastoma TICs from six primary neuroblastoma tumours and two bone marrow metastases. The primary tumours from which these TICs were generated have previously been fully typed by whole genome sequencing (WGS). Array comparative genomic hybridisation (aCGH) analysis showed that TIC lines retained essential characteristics of the primary tumours and exhibited typical neuroblastoma chromosomal aberrations such as MYCN amplification, gain of chromosome 17q and deletion of 1p36. Protein analysis showed expression for neuroblastoma markers MYCN, NCAM, CHGA, DBH and TH while haematopoietic markers CD19 and CD11b were absent. We analysed the growth characteristics and confirmed tumour-forming potential using sphere-forming assays, subcutaneous and orthotopic injection of these cells into immune-compromised mice. Affymetrix mRNA expression profiling of TIC line xenografts showed an expression pattern more closely mimicking primary tumours compared to xenografts from classical cell lines. This establishes that these neuroblastoma TICs cultured under serum-free conditions are relevant and useful neuroblastoma tumour models.


BMC Cancer | 2012

Identification of BIRC6 as a novel intervention target for neuroblastoma therapy

Fieke Lamers; Linda Schild; Jan Koster; Franki Speleman; Ingrid Øra; Ellen M. Westerhout; Peter van Sluis; Rogier Versteeg; Huib N. Caron; Jan J. Molenaar

BackgroundNeuroblastoma are pediatric tumors of the sympathetic nervous system with a poor prognosis. Apoptosis is often deregulated in cancer cells, but only a few defects in apoptotic routes have been identified in neuroblastoma.MethodsHere we investigated genomic aberrations affecting genes of the intrinsic apoptotic pathway in neuroblastoma. We analyzed DNA profiling data (CGH and SNP arrays) and mRNA expression data of 31 genes of the intrinsic apoptotic pathway in a dataset of 88 neuroblastoma tumors using the R2 bioinformatic platform (http://r2.amc.nl). BIRC6 was selected for further analysis as a tumor driving gene. Knockdown experiments were performed using BIRC6 lentiviral shRNA and phenotype responses were analyzed by Western blot and MTT-assays. In addition, DIABLO levels and interactions were investigated with immunofluorescence and co-immunoprecipitation.ResultsWe observed frequent gain of the BIRC6 gene on chromosome 2, which resulted in increased mRNA expression. BIRC6 is an inhibitor of apoptosis protein (IAP), that can bind and degrade the cytoplasmic fraction of the pro-apoptotic protein DIABLO. DIABLO mRNA expression was exceptionally high in neuroblastoma but the protein was only detected in the mitochondria. Upon silencing of BIRC6 by shRNA, DIABLO protein levels increased and cells went into apoptosis. Co-immunoprecipitation confirmed direct interaction between DIABLO and BIRC6 in neuroblastoma cell lines.ConclusionOur findings indicate that BIRC6 may have a potential oncogenic role in neuroblastoma by inactivating cytoplasmic DIABLO. BIRC6 inhibition may therefore provide a means for therapeutic intervention in neuroblastoma.


Clinical Cancer Research | 2017

p53 Nongenotoxic Activation and mTORC1 Inhibition Lead to Effective Combination for Neuroblastoma Therapy

Myrthala Moreno-Smith; Anna Lakoma; Zaowen Chen; Ling Tao; Kathleen A. Scorsone; Linda Schild; Kevin Aviles-Padilla; Rana Nikzad; Yankai Zhang; Rikhia Chakraborty; Jan J. Molenaar; Sanjeev A. Vasudevan; Vivien Sheehan; Eugene S. Kim; Silke Paust; Jason M. Shohet; Eveline Barbieri

Purpose: mTORC1 inhibitors are promising agents for neuroblastoma therapy; however, they have shown limited clinical activity as monotherapy, thus rational drug combinations need to be explored to improve efficacy. Importantly, neuroblastoma maintains both an active p53 and an aberrant mTOR signaling. Experimental Design: Using an orthotopic xenograft model and modulating p53 levels, we investigated the antitumor effects of the mTORC1 inhibitor temsirolimus in neuroblastoma expressing normal, decreased, or mutant p53, both as single agent and in combination with first- and second-generation MDM2 inhibitors to reactivate p53. Results: Nongenotoxic p53 activation suppresses mTOR activity. Moreover, p53 reactivation via RG7388, a second-generation MDM2 inhibitor, strongly enhances the in vivo antitumor activity of temsirolimus. Single-agent temsirolimus does not elicit apoptosis, and tumors rapidly regrow after treatment suspension. In contrast, our combination therapy triggers a potent apoptotic response in wild-type p53 xenografts and efficiently blocks tumor regrowth after treatment completion. We also found that this combination uniquely led to p53-dependent suppression of survivin whose ectopic expression is sufficient to rescue the apoptosis induced by our combination. Conclusions: Our study supports a novel highly effective strategy that combines RG7388 and temsirolimus in wild-type p53 neuroblastoma, which warrants testing in early-phase clinical trials. Clin Cancer Res; 23(21); 6629–39. ©2017 AACR.


Cancer Research | 2016

Abstract A31: Activation of the RAS-MAPK pathway in primary neuroblastoma tumors is associated with poor prognosis

Thomas F. Eleveld; Linda Schild; Marli E. Ebus; P.G. van Sluis; Ellen M. Westerhout; H.N. Caron; J.J.B. Koster; Rogier Versteeg; Jan J. Molenaar

We recently identified that mutations affecting the RAS-MAPK pathway occur frequently in relapsed neuroblastoma, which suggests that activation of this pathway may play an important role in the aggressive phenotype of these tumors. Furthermore, it was shown that the presence of these mutations may serve as a biomarker for treatment with MEK inhibitors both in vitro and in vivo. These results suggest that MEK inhibitors may be a viable treatment option for aggressive neuroblastoma tumors and warrant further investigation into the role of the RAS-MAPK pathway in neuroblastoma. To identify if, and to what extent, the RAS-MAPK pathway is also activated in primary neuroblastoma tumors we have developed a RAS-MAPK mRNA signature. Genes were included that were I) regulated by mutant ALK, which is a known activator of the RAS-MAPK pathway in neuroblastoma, and II) were conversely regulated by treatment with the MEK inhibitor U0126. We ended up with a core signature consisting of 8 genes, which was tested in a panel of 17 neuroblastoma cell lines. The expression of our gene signature in this panel correlates with mutations in the RAS-MAPK pathway, with the expression of phospho-MEK and phospho-ERK on Western Blot and with sensitivity to the MEK inhibitor Trametinib. Clustering analyses in our set of 122 primary neuroblastoma tumors shows that high expression of this signature is associated with significantly poorer survival and similar analyses in other publicly available datasets corroborate these results, even if applied to only stage 4 tumors . In our data set all tumors containing mutations in known RAS-MAPK pathway genes, such as ALK and NF1 , show high signature scores, which makes it likely that this signature can indeed detect the activation state of the pathway through mRNA profiling. However, there are also tumors with high signature scores that do not show mutations that are known to be associated with RAS-MAPK signaling (as detectable by Whole Genome Sequencing). Therefore we looked in these tumors if there were other mutations that could explain activation of this pathway. We detected mutations in known RAS-MAPK genes, such as SOS1 , but also recurrent aberrations in genes not previously known to be related to RAS-MAPK signaling, such as PHOX2B , DMD and ERF . We are currently characterizing the effect of these mutations on the RAS-MAPK signaling pathway in neuroblastoma cell lines. Our findings show that activation of the RAS-MAPK pathway, as determined by our mRNA gene signature, is associated with poor prognosis in neuroblastoma. In some tumors this activation is caused by known activating mutations in this pathway, but in most cases the causative events remain unknown. We present several other candidate genes, which when mutated may also lead to pathway activation. MEK inhibitors were already shown to be effective against neuroblastoma xenografts with RAS or NF1 mutations, and our results indicate that a broader selection of neuroblastoma tumors might respond to these compounds. Citation Format: T.F. Eleveld, L Schild, M.E. Ebus, P.G. van Sluis, E.M. Westerhout, H.N. Caron, J.J.B. Koster, R. Versteeg, J.J. Molenaar. Activation of the RAS-MAPK pathway in primary neuroblastoma tumors is associated with poor prognosis. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Pediatric Cancer Research: From Mechanisms and Models to Treatment and Survivorship; 2015 Nov 9-12; Fort Lauderdale, FL. Philadelphia (PA): AACR; Cancer Res 2016;76(5 Suppl):Abstract nr A31.

Collaboration


Dive into the Linda Schild's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Koster

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Huib N. Caron

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fieke Lamers

University of Amsterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge