Koschmann, C.
Al-Holou, W.N.
Alonso, M.M.
Anastas, J.
Bandopadhayay, P.
Barron, T.
Becher, O.
Cartaxo, R.
Castro, M.G.
Chung, C.
Clausen, M.
Dang, D.
Doherty, R.
Duchatel, R.
Dun, M.
Filbin, M.
Franson, A.
Galban, S.
Garcia Moure, M.
Garton, H.
Gowda, P.
Marques, J.G.
Hawkins, C.
Heath, A.
Hulleman, E.
Ji, S.
Jones, C.
Kilburn, L.
Kline, C.
Koldobskiy, M.A.
Lim, D.
Lowenstein, P.R.
Lu, Q.R.
Lum, J.
Mack, S.
Magge, S.
Marini, B.
Martin, D.
Marupudi, N.
Messinger, D.
Mody, R.
Morgan, M.
Mota, M.
Muraszko, K.
Mueller, S.
Natarajan, S.K.
Nazarian, J.
Niculcea, M.
Nuechterlein, N.
Okada, H.
Opipari, V.
Pai, M.P.
Pal, S.
Peterson, E.
Phoenix, T.
Prensner, J.R.
Pun, M.
Raju, G.P.
Reitman, Z.J.
Resnick, A.
Rogawski, D.
Saratsis, A.
Sbergio, S.G.
Souweidane, M.
Stafford, J.M.
Tzaridis, T.
Venkataraman, S.
Vittorio, O.
Wadden, J.
Wahl, D.
Wechsler-Reya, R.J.
Yadav, V.N.
Zhang, X.
Zhang, Q.
Venneti, S.
(2024). A road map for the treatment of pediatric diffuse midline glioma. Cancer cell,
Vol.42
(1),
pp. 1-5.
show abstract
Recent clinical trials for H3K27-altered diffuse midline gliomas (DMGs) have shown much promise. We present a consensus roadmap and identify three major barriers: (1) refinement of experimental models to include immune and brain-specific components; (2) collaboration among researchers, clinicians, and industry to integrate patient-derived data through sharing, transparency, and regulatory considerations; and (3) streamlining clinical efforts including biopsy, CNS-drug delivery, endpoint determination, and response monitoring. We highlight the importance of comprehensive collaboration to advance the understanding, diagnostics, and therapeutics for DMGs..
Liu, I.
Alencastro Veiga Cruzeiro, G.
Bjerke, L.
Rogers, R.F.
Grabovska, Y.
Beck, A.
Mackay, A.
Barron, T.
Hack, O.A.
Quezada, M.A.
Molinari, V.
Shaw, M.L.
Perez-Somarriba, M.
Temelso, S.
Raynaud, F.
Ruddle, R.
Panditharatna, E.
Englinger, B.
Mire, H.M.
Jiang, L.
Nascimento, A.
LaBelle, J.
Haase, R.
Rozowsky, J.
Neyazi, S.
Baumgartner, A.-.
Castellani, S.
Hoffman, S.E.
Cameron, A.
Morrow, M.
Nguyen, Q.-.
Pericoli, G.
Madlener, S.
Mayr, L.
Dorfer, C.
Geyeregger, R.
Rota, C.
Ricken, G.
Ligon, K.L.
Alexandrescu, S.
Cartaxo, R.T.
Lau, B.
Uphadhyaya, S.
Koschmann, C.
Braun, E.
Danan-Gotthold, M.
Hu, L.
Siletti, K.
Sundström, E.
Hodge, R.
Lein, E.
Agnihotri, S.
Eisenstat, D.D.
Stapleton, S.
King, A.
Bleil, C.
Mastronuzzi, A.
Cole, K.A.
Waanders, A.J.
Montero Carcaboso, A.
Schüller, U.
Hargrave, D.
Vinci, M.
Carceller, F.
Haberler, C.
Slavc, I.
Linnarsson, S.
Gojo, J.
Monje, M.
Jones, C.
Filbin, M.G.
(2024). GABAergic neuronal lineage development determines clinically actionable targets in diffuse hemispheric glioma, H3G34-mutant. Cancer cell,
.
show abstract
Diffuse hemispheric gliomas, H3G34R/V-mutant (DHG-H3G34), are lethal brain tumors lacking targeted therapies. They originate from interneuronal precursors; however, leveraging this origin for therapeutic insights remains unexplored. Here, we delineate a cellular hierarchy along the interneuron lineage development continuum, revealing that DHG-H3G34 mirror spatial patterns of progenitor streams surrounding interneuron nests, as seen during human brain development. Integrating these findings with genome-wide CRISPR-Cas9 screens identifies genes upregulated in interneuron lineage progenitors as major dependencies. Among these, CDK6 emerges as a targetable vulnerability: DHG-H3G34 tumor cells show enhanced sensitivity to CDK4/6 inhibitors and a CDK6-specific degrader, promoting a shift toward more mature interneuron-like states, reducing tumor growth, and prolonging xenograft survival. Notably, a patient with progressive DHG-H3G34 treated with a CDK4/6 inhibitor achieved 17 months of stable disease. This study underscores interneuronal progenitor-like states, organized in characteristic niches, as a distinct vulnerability in DHG-H3G34, highlighting CDK6 as a promising clinically actionable target..
Sun, C.X.
Daniel, P.
Bradshaw, G.
Shi, H.
Loi, M.
Chew, N.
Parackal, S.
Tsui, V.
Liang, Y.
Koptyra, M.
Adjumain, S.
Sun, C.
Chong, W.C.
Fernando, D.
Drinkwater, C.
Tourchi, M.
Habarakada, D.
Sooraj, D.
Carvalho, D.
Storm, P.B.
Baubet, V.
Sayles, L.C.
Fernandez, E.
Nguyen, T.
Pörksen, M.
Doan, A.
Crombie, D.E.
Panday, M.
Zhukova, N.
Dun, M.D.
Ludlow, L.E.
Day, B.
Stringer, B.W.
Neeman, N.
Rubens, J.A.
Raabe, E.H.
Vinci, M.
Tyrrell, V.
Fletcher, J.I.
Ekert, P.G.
Dumevska, B.
Ziegler, D.S.
Tsoli, M.
Syed Sulaiman, N.F.
Loh, A.H.
Low, S.Y.
Sweet-Cordero, E.A.
Monje, M.
Resnick, A.
Jones, C.
Downie, P.
Williams, B.
Rosenbluh, J.
Gough, D.
Cain, J.E.
Firestein, R.
(2023). Generation and multi-dimensional profiling of a childhood cancer cell line atlas defines new therapeutic opportunities. Cancer cell,
Vol.41
(4),
pp. 660-677.e7.
show abstract
Pediatric solid and central nervous system tumors are the leading cause of cancer-related death among children. Identifying new targeted therapies necessitates the use of pediatric cancer models that faithfully recapitulate the patient's disease. However, the generation and characterization of pediatric cancer models has significantly lagged behind adult cancers, underscoring the urgent need to develop pediatric-focused cell line resources. Herein, we establish a single-site collection of 261 cell lines, including 224 pediatric cell lines representing 18 distinct extracranial and brain childhood tumor types. We subjected 182 cell lines to multi-omics analyses (DNA sequencing, RNA sequencing, DNA methylation), and in parallel performed pharmacological and genetic CRISPR-Cas9 loss-of-function screens to identify pediatric-specific treatment opportunities and biomarkers. Our work provides insight into specific pathway vulnerabilities in molecularly defined pediatric tumor classes and uncovers biomarker-linked therapeutic opportunities of clinical relevance. Cell line data and resources are provided in an open access portal..
Pericoli, G.
Galardi, A.
Paolini, A.
Petrilli, L.L.
Pepe, G.
Palma, A.
Colletti, M.
Ferretti, R.
Giorda, E.
Levi Mortera, S.
Burford, A.
Carai, A.
Mastronuzzi, A.
Mackay, A.
Putignani, L.
Jones, C.
Pascucci, L.
Peinado, H.
Helmer-Citterich, M.
de Billy, E.
Masotti, A.
Locatelli, F.
Di Giannatale, A.
Vinci, M.
(2023). Inhibition of exosome biogenesis affects cell motility in heterogeneous sub-populations of paediatric-type diffuse high-grade gliomas. Cell biosci,
Vol.13
(1),
p. 207.
show abstract
full text
BACKGROUND: Paediatric-type diffuse High-Grade Gliomas (PDHGG) are highly heterogeneous tumours which include distinct cell sub-populations co-existing within the same tumour mass. We have previously shown that primary patient-derived and optical barcoded single-cell-derived clones function as interconnected networks. Here, we investigated the role of exosomes as a route for inter-clonal communication mediating PDHGG migration and invasion. RESULTS: A comprehensive characterisation of seven optical barcoded single-cell-derived clones obtained from two patient-derived cell lines was performed. These analyses highlighted extensive intra-tumour heterogeneity in terms of genetic and transcriptional profiles between clones as well as marked phenotypic differences including distinctive motility patterns. Live single-cell tracking analysis of 3D migration and invasion assays showed that the single-cell-derived clones display a higher speed and longer travelled distance when in co-culture compared to mono-culture conditions. To determine the role of exosomes in PDHGG inter-clonal cross-talks, we isolated exosomes released by different clones and characterised them in terms of marker expression, size and concentration. We demonstrated that exosomes are actively internalized by the cells and that the inhibition of their biogenesis, using the phospholipase inhibitor GW4689, significantly reduced the cell motility in mono-culture and more prominently when the cells from the clones were in co-culture. Analysis of the exosomal miRNAs, performed with a miRNome PCR panel, identified clone-specific miRNAs and a set of miRNA target genes involved in the regulation of cell motility/invasion/migration. These genes were found differentially expressed in co-culture versus mono-culture conditions and their expression levels were significantly modulated upon inhibition of exosome biogenesis. CONCLUSIONS: In conclusion, our study highlights for the first time a key role for exosomes in the inter-clonal communication in PDHGG and suggests that interfering with the exosome biogenesis pathway may be a valuable strategy to inhibit cell motility and dissemination for these specific diseases..
de Billy, E.
Pellegrino, M.
Orlando, D.
Pericoli, G.
Ferretti, R.
Businaro, P.
Ajmone-Cat, M.A.
Rossi, S.
Petrilli, L.L.
Maestro, N.
Diomedi-Camassei, F.
Pezzullo, M.
De Stefanis, C.
Bencivenga, P.
Palma, A.
Rota, R.
Del Bufalo, F.
Massimi, L.
Weber, G.
Jones, C.
Carai, A.
Caruso, S.
De Angelis, B.
Caruana, I.
Quintarelli, C.
Mastronuzzi, A.
Locatelli, F.
Vinci, M.
(2022). Dual IGF1R/IR inhibitors in combination with GD2-CAR T-cells display a potent anti-tumor activity in diffuse midline glioma H3K27M-mutant. Neuro oncol,
Vol.24
(7),
pp. 1150-1163.
show abstract
full text
BACKGROUND: Diffuse midline gliomas (DMG) H3K27M-mutant, including diffuse intrinsic pontine glioma (DIPG), are pediatric brain tumors associated with grim prognosis. Although GD2-CAR T-cells demonstrated significant anti-tumor activity against DMG H3K27M-mutant in vivo, a multimodal approach may be needed to more effectively treat patients. We investigated GD2 expression in DMG/DIPG and other pediatric high-grade gliomas (pHGG) and sought to identify chemical compounds that would enhance GD2-CAR T-cell anti-tumor efficacy. METHODS: Immunohistochemistry in tumor tissue samples and immunofluorescence in primary patient-derived cell lines were performed to study GD2 expression. We developed a high-throughput cell-based assay to screen 42 kinase inhibitors in combination with GD2-CAR T-cells. Cell viability, western blots, flow-cytometry, real time PCR experiments, DIPG 3D culture models, and orthotopic xenograft model were applied to investigate the effect of selected compounds on DIPG cell death and CAR T-cell function. RESULTS: GD2 was heterogeneously, but widely, expressed in the tissue tested, while its expression was homogeneous and restricted to DMG/DIPG H3K27M-mutant cell lines. We identified dual IGF1R/IR antagonists, BMS-754807 and linsitinib, able to inhibit tumor cell viability at concentrations that do not affect CAR T-cells. Linsitinib, but not BMS-754807, decreases activation/exhaustion of GD2-CAR T-cells and increases their central memory profile. The enhanced anti-tumor activity of linsitinib/GD2-CAR T-cell combination was confirmed in DIPG models in vitro, ex vivo, and in vivo. CONCLUSION: Our study supports the development of IGF1R/IR inhibitors to be used in combination with GD2-CAR T-cells for treating patients affected by DMG/DIPG and, potentially, by pHGG..
Khadka, P.
Reitman, Z.J.
Lu, S.
Buchan, G.
Gionet, G.
Dubois, F.
Carvalho, D.M.
Shih, J.
Zhang, S.
Greenwald, N.F.
Zack, T.
Shapira, O.
Pelton, K.
Hartley, R.
Bear, H.
Georgis, Y.
Jarmale, S.
Melanson, R.
Bonanno, K.
Schoolcraft, K.
Miller, P.G.
Condurat, A.L.
Gonzalez, E.M.
Qian, K.
Morin, E.
Langhnoja, J.
Lupien, L.E.
Rendo, V.
Digiacomo, J.
Wang, D.
Zhou, K.
Kumbhani, R.
Guerra Garcia, M.E.
Sinai, C.E.
Becker, S.
Schneider, R.
Vogelzang, J.
Krug, K.
Goodale, A.
Abid, T.
Kalani, Z.
Piccioni, F.
Beroukhim, R.
Persky, N.S.
Root, D.E.
Carcaboso, A.M.
Ebert, B.L.
Fuller, C.
Babur, O.
Kieran, M.W.
Jones, C.
Keshishian, H.
Ligon, K.L.
Carr, S.A.
Phoenix, T.N.
Bandopadhayay, P.
(2022). PPM1D mutations are oncogenic drivers of de novo diffuse midline glioma formation. Nat commun,
Vol.13
(1),
p. 604.
show abstract
full text
The role of PPM1D mutations in de novo gliomagenesis has not been systematically explored. Here we analyze whole genome sequences of 170 pediatric high-grade gliomas and find that truncating mutations in PPM1D that increase the stability of its phosphatase are clonal driver events in 11% of Diffuse Midline Gliomas (DMGs) and are enriched in primary pontine tumors. Through the development of DMG mouse models, we show that PPM1D mutations potentiate gliomagenesis and that PPM1D phosphatase activity is required for in vivo oncogenesis. Finally, we apply integrative phosphoproteomic and functional genomics assays and find that oncogenic effects of PPM1D truncation converge on regulators of cell cycle, DNA damage response, and p53 pathways, revealing therapeutic vulnerabilities including MDM2 inhibition..
Izquierdo, E.
Carvalho, D.M.
Mackay, A.
Temelso, S.
Boult, J.K.
Pericoli, G.
Fernandez, E.
Das, M.
Molinari, V.
Grabovska, Y.
Rogers, R.F.
Ajmone-Cat, M.A.
Proszek, P.Z.
Stubbs, M.
Depani, S.
O'Hare, P.
Yu, L.
Roumelioti, G.
Choudhary, J.S.
Clarke, M.
Fairchild, A.R.
Jacques, T.S.
Grundy, R.G.
Howell, L.
Picton, S.
Adamski, J.
Wilson, S.
Gray, J.C.
Zebian, B.
Marshall, L.V.
Carceller, F.
Grill, J.
Vinci, M.
Robinson, S.P.
Hubank, M.
Hargrave, D.
Jones, C.
(2022). DIPG Harbors Alterations Targetable by MEK Inhibitors, with Acquired Resistance Mechanisms Overcome by Combinatorial Inhibition. Cancer discov,
Vol.12
(3),
pp. 712-729.
show abstract
full text
UNLABELLED: The survival of children with diffuse intrinsic pontine glioma (DIPG) remains dismal, with new treatments desperately needed. In a prospective biopsy-stratified clinical trial, we combined detailed molecular profiling and drug screening in newly established patient-derived models in vitro and in vivo. We identified in vitro sensitivity to MEK inhibitors in DIPGs harboring MAPK pathway alterations, but treatment of patient-derived xenograft models and a patient at relapse failed to elicit a significant response. We generated trametinib-resistant clones in a BRAFG469V model through continuous drug exposure and identified acquired mutations in MEK1/2 with sustained pathway upregulation. These cells showed hallmarks of mesenchymal transition and expression signatures overlapping with inherently trametinib-insensitive patient-derived cells, predicting sensitivity to dasatinib. Combined trametinib and dasatinib showed highly synergistic effects in vitro and on ex vivo brain slices. We highlight the MAPK pathway as a therapeutic target in DIPG and show the importance of parallel resistance modeling and combinatorial treatments for meaningful clinical translation. SIGNIFICANCE: We report alterations in the MAPK pathway in DIPGs to confer initial sensitivity to targeted MEK inhibition. We further identify for the first time the mechanism of resistance to single-agent targeted therapy in these tumors and suggest a novel combinatorial treatment strategy to overcome it in the clinic. This article is highlighted in the In This Issue feature, p. 587..
Carvalho, D.M.
Richardson, P.J.
Olaciregui, N.
Stankunaite, R.
Lavarino, C.
Molinari, V.
Corley, E.A.
Smith, D.P.
Ruddle, R.
Donovan, A.
Pal, A.
Raynaud, F.I.
Temelso, S.
Mackay, A.
Overington, J.P.
Phelan, A.
Sheppard, D.
Mackinnon, A.
Zebian, B.
Al-Sarraj, S.
Merve, A.
Pryce, J.
Grill, J.
Hubank, M.
Cruz, O.
Morales La Madrid, A.
Mueller, S.
Carcaboso, A.M.
Carceller, F.
Jones, C.
(2022). Repurposing Vandetanib plus Everolimus for the Treatment of ACVR1-Mutant Diffuse Intrinsic Pontine Glioma. Cancer discov,
Vol.12
(2),
pp. 416-431.
show abstract
full text
Somatic mutations in ACVR1 are found in a quarter of children with diffuse intrinsic pontine glioma (DIPG), but there are no ACVR1 inhibitors licensed for the disease. Using an artificial intelligence-based platform to search for approved compounds for ACVR1-mutant DIPG, the combination of vandetanib and everolimus was identified as a possible therapeutic approach. Vandetanib, an inhibitor of VEGFR/RET/EGFR, was found to target ACVR1 (K d = 150 nmol/L) and reduce DIPG cell viability in vitro but has limited ability to cross the blood-brain barrier. In addition to mTOR, everolimus inhibited ABCG2 (BCRP) and ABCB1 (P-gp) transporters and was synergistic in DIPG cells when combined with vandetanib in vitro. This combination was well tolerated in vivo and significantly extended survival and reduced tumor burden in an orthotopic ACVR1-mutant patient-derived DIPG xenograft model. Four patients with ACVR1-mutant DIPG were treated with vandetanib plus an mTOR inhibitor, informing the dosing and toxicity profile of this combination for future clinical studies. SIGNIFICANCE: Twenty-five percent of patients with the incurable brainstem tumor DIPG harbor somatic activating mutations in ACVR1, but there are no approved drugs targeting the receptor. Using artificial intelligence, we identify and validate, both experimentally and clinically, the novel combination of vandetanib and everolimus in these children based on both signaling and pharmacokinetic synergies.This article is highlighted in the In This Issue feature, p. 275..
Rodriguez, D.
Calmon, R.
Aliaga, E.S.
Warren, D.
Warmuth-Metz, M.
Jones, C.
Mackay, A.
Varlet, P.
Le Deley, M.-.
Hargrave, D.
Cañete, A.
Massimino, M.
Azizi, A.A.
Saran, F.
Zahlmann, G.
Garcia, J.
Vassal, G.
Grill, J.
Peet, A.
Dineen, R.A.
Morgan, P.S.
Jaspan, T.
(2022). MRI and Molecular Characterization of Pediatric High-Grade Midline Thalamic Gliomas: The HERBY Phase II Trial. Radiology,
Vol.304
(1),
pp. 174-182.
show abstract
Background Diffuse midline gliomas (DMG) are characterized by a high incidence of H3 K27 mutations and poorer outcome. The HERBY trial has provided one of the largest cohorts of pediatric DMGs with available radiologic, histologic-genotypic, and survival data. Purpose To define MRI and molecular characteristics of DMG. Materials and Methods This study is a secondary analysis of a prospective trial (HERBY; ClinicalTrials.gov identifier, NCT01390948) undertaken between October 2011 and February 2016. Among 121 HERBY participants, 50 had midline nonpontine-based tumors. Midline high-grade gliomas were reclassified into DMG H3 K27 mutant, H3 wild type with enhancer of zest homologs inhibitory protein overexpression, epidermal growth factor receptormutant, or not otherwise stated. The epicenter of each tumor and other radiologic characteristics were ascertained from MRI and correlated with the new subtype classification, histopathologic characteristics, surgical extent, and outcome parameters. Kaplan-Meier curves and log-rank tests were applied to determine and describe survival differences between groups. Results There were 42 participants (mean age, 12 years ± 4 [SD]; 23 girls) with radiologically evaluable thalamic-based DMG. Eighteen had partial thalamic involvement (12 thalamopulvinar, six anteromedial), 10 involved a whole thalamus, nine had unithalamic tumors with diffuse contiguous extension, and five had bithalamic tumors (two symmetric, three partial). Twenty-eight participants had DMG H3 K27 mutant tumors; there were no differences in outcome compared with other DMGs (n = 4). Participants who underwent major debulking or total or near-total resection had longer overall survival (OS): 18.5 months vs 11.4 months (P = .02). Enrolled participants who developed leptomeningeal metastatic dissemination before starting treatment had worse outcomes (event-free survival, 2.9 months vs 8.0 months [P = .02]; OS, 11.4 months vs 18.5 months [P = .004]). Conclusion Thalamic involvement of diffuse midline gliomas ranged from localized partial thalamic to holo- or bithalamic with diffuse contiguous spread and had poor outcomes, irrespective of H3 K27 subtype alterations. Leptomeningeal dissemination and less than 50% surgical resection were adverse risk factors for survival. Clinical trial registration no. NCT01390948 © RSNA, 2022 Online supplemental material is available for this article. See also the editorial by Widjaja in this issue..
Dubois, F.P.
Shapira, O.
Greenwald, N.F.
Zack, T.
Wala, J.
Tsai, J.W.
Crane, A.
Baguette, A.
Hadjadj, D.
Harutyunyan, A.S.
Kumar, K.H.
Blattner-Johnson, M.
Vogelzang, J.
Sousa, C.
Kang, K.S.
Sinai, C.
Wang, D.K.
Khadka, P.
Lewis, K.
Nguyen, L.
Malkin, H.
Ho, P.
O'Rourke, R.
Zhang, S.
Gold, R.
Deng, D.
Serrano, J.
Snuderl, M.
Jones, C.
Wright, K.D.
Chi, S.N.
Grill, J.
Kleinman, C.L.
Goumnerova, L.C.
Jabado, N.
Jones, D.T.
Kieran, M.W.
Ligon, K.L.
Beroukhim, R.
Bandopadhayay, P.
(2022). Structural variants shape driver combinations and outcomes in pediatric high-grade glioma. Nat cancer,
Vol.3
(8),
pp. 994-1011.
show abstract
full text
We analyzed the contributions of structural variants (SVs) to gliomagenesis across 179 pediatric high-grade gliomas (pHGGs). The most recurrent SVs targeted MYC isoforms and receptor tyrosine kinases (RTKs), including an SV amplifying a MYC enhancer in 12% of diffuse midline gliomas (DMG), indicating an underappreciated role for MYC in pHGG. SV signature analysis revealed that tumors with simple signatures were TP53 wild type (TP53WT) but showed alterations in TP53 pathway members PPM1D and MDM4. Complex signatures were associated with direct aberrations in TP53, CDKN2A and RB1 early in tumor evolution and with later-occurring extrachromosomal amplicons. All pHGGs exhibited at least one simple-SV signature, but complex-SV signatures were primarily restricted to subsets of H3.3K27M DMGs and hemispheric pHGGs. Importantly, DMGs with complex-SV signatures were associated with shorter overall survival independent of histone mutation and TP53 status. These data provide insight into the impact of SVs on gliomagenesis and the mechanisms that shape them..
Tari, H.
Kessler, K.
Trahearn, N.
Werner, B.
Vinci, M.
Jones, C.
Sottoriva, A.
(2022). Quantification of spatial subclonal interactions enhancing the invasive phenotype of pediatric glioma. Cell rep,
Vol.40
(9),
p. 111283.
show abstract
full text
Diffuse midline gliomas (DMGs) are highly aggressive, incurable childhood brain tumors. They present a clinical challenge due to many factors, including heterogeneity and diffuse infiltration, complicating disease management. Recent studies have described the existence of subclonal populations that may co-operate to drive pro-tumorigenic processes such as cellular invasion. However, a precise quantification of subclonal interactions is lacking, a problem that extends to other cancers. In this study, we combine spatial computational modeling of cellular interactions during invasion with co-evolution experiments of clonally disassembled patient-derived DMG cells. We design a Bayesian inference framework to quantify spatial subclonal interactions between molecular and phenotypically distinct lineages with different patterns of invasion. We show how this approach could discriminate genuine interactions, where one clone enhanced the invasive phenotype of another, from those apparently only due to the complex dynamics of spatially restricted growth. This study provides a framework for the quantification of subclonal interactions in DMG..
Petrilli, L.L.
Fuoco, C.
Palma, A.
Pasquini, L.
Pericoli, G.
Grabovska, Y.
Mackay, A.
Rossi, S.
Carcaboso, A.M.
Carai, A.
Mastronuzzi, A.
Jones, C.
Cesareni, G.
Locatelli, F.
Vinci, M.
(2022). Inter and intra-tumor heterogeneity of paediatric type diffuse high-grade gliomas revealed by single-cell mass cytometry. Front oncol,
Vol.12,
p. 1016343.
show abstract
full text
Paediatric-type diffuse high-grade gliomas (PDHGG) are aggressive tumors affecting children and young adults, with no effective treatment. These highly heterogeneous malignancies arise in different sites of the Central Nervous System (CNS), carrying distinctive molecular alterations and clinical outcomes (inter-tumor heterogeneity). Moreover, deep cellular and molecular profiling studies highlighted the coexistence of genetically and phenotypically different subpopulations within the same tumor mass (intra-tumor heterogeneity). Despite the recent advances made in the field, the marked heterogeneity of PDHGGs still impedes the development of effective targeted therapies and the identification of suitable biomarkers. In order to fill the existing gap, we used mass cytometry to dissect PDHGG inter- and intra-heterogeneity. This is one of the most advanced technologies of the "-omics" era that, using antibodies conjugated to heavy metals, allows the simultaneous measurement of more than 40 markers at single-cell level. To this end, we analyzed eight PDHGG patient-derived cell lines from different locational and molecular subgroups. By using a panel of 15 antibodies, directly conjugated to metals or specifically customized to detect important histone variants, significant differences were highlighted in the expression of the considered antigens. The single-cell multiparametric approach realized has deepened our understanding of PDHGG, confirming a high degree of intra- and inter-tumoral heterogeneity and identifying some antigens that could represent useful biomarkers for the specific PDHGG locational or molecular subgroups..
Pearce, J.
Khabra, K.
Nanji, H.
Stone, J.
Powell, K.
Martin, D.
Zebian, B.
Hettige, S.
Reisz, Z.
Bodi, I.
Al-Sarraj, S.
Bridges, L.R.
Clarke, M.
Jones, C.
Mandeville, H.C.
Vaidya, S.
Marshall, L.V.
Carceller, F.
(2021). High grade gliomas in young children: The South Thames Neuro-Oncology unit experience and recent advances in molecular biology and targeted therapies. Pediatr hematol oncol,
Vol.38
(8),
pp. 707-721.
show abstract
High grade gliomas (HGG) have a dismal prognosis with survival rates of 15-35%. Approximately 10-12% of pediatric HGG occur in young children and their molecular biology and clinical outcomes differ from those arising at older ages. We report on four children aged <5 years newly diagnosed with non-brainstem HGG between 2011 and 2018 who were treated with surgery and BBSFOP chemotherapy. Two died of tumor progression. The other two are still alive without radiotherapy at 3.8 and 3.9 years from diagnosis: one of whom remains disease-free off treatment; and the other one, whose tumor harbored a KCTD16:NTRK2 fusion, went on to receive larotrectinib. Additionally we review the general management, outcomes and latest updates in molecular biology and targeted therapies for young children with HGG. Infant gliomas can be stratified in molecular subgroups with clinically actionable oncogenic drivers. Chemotherapy-based strategies can avoid or delay the need for radiotherapy in young children with HGG. Harnessing the potential of NTRK, ALK, ROS1 and MET inhibitors offers the opportunity to optimize the therapeutic armamentarium to improve current outcomes for these children..
Sweha, S.R.
Chung, C.
Natarajan, S.K.
Panwalkar, P.
Pun, M.
Ghali, A.
Bayliss, J.
Pratt, D.
Shankar, A.
Ravikumar, V.
Rao, A.
Cieslik, M.
Wilder-Romans, K.
Scott, A.J.
Wahl, D.R.
Jessa, S.
Kleinman, C.L.
Jabado, N.
Mackay, A.
Jones, C.
Martinez, D.
Santi, M.
Judkins, A.R.
Yadav, V.N.
Qin, T.
Phoenix, T.N.
Koschmann, C.J.
Baker, S.J.
Chinnaiyan, A.M.
Venneti, S.
(2021). Epigenetically defined therapeutic targeting in H3 3G34R/V high-grade gliomas. Sci transl med,
Vol.13
(615),
p. eabf7860.
show abstract
full text
High-grade gliomas with arginine or valine substitutions of the histone H3.3 glycine-34 residue (H3.3G34R/V) carry a dismal prognosis, and current treatments, including radiotherapy and chemotherapy, are not curative. Because H3.3G34R/V mutations reprogram epigenetic modifications, we undertook a comprehensive epigenetic approach using ChIP sequencing and ChromHMM computational analysis to define therapeutic dependencies in H3.3G34R/V gliomas. Our analyses revealed a convergence of epigenetic alterations, including (i) activating epigenetic modifications on histone H3 lysine (K) residues such as H3K36 trimethylation (H3K36me3), H3K27 acetylation (H3K27ac), and H3K4 trimethylation (H3K4me3); (ii) DNA promoter hypomethylation; and (iii) redistribution of repressive histone H3K27 trimethylation (H3K27me3) to intergenic regions at the leukemia inhibitory factor (LIF) locus to drive increased LIF abundance and secretion by H3.3G34R/V cells. LIF activated signal transducer and activator of transcription 3 (STAT3) signaling in an autocrine/paracrine manner to promote survival of H3.3G34R/V glioma cells. Moreover, immunohistochemistry and single-cell RNA sequencing from H3.3G34R/V patient tumors revealed high STAT3 protein and RNA expression, respectively, in tumor cells with both inter- and intratumor heterogeneity. We targeted STAT3 using a blood-brain barrier–penetrable small-molecule inhibitor, WP1066, currently in clinical trials for adult gliomas. WP1066 treatment resulted in H3.3G34R/V tumor cell toxicity in vitro and tumor suppression in preclinical mouse models established with KNS42 cells, SJ-HGGx42-c cells, or in utero electroporation techniques. Our studies identify the LIF/STAT3 pathway as a key epigenetically driven and druggable vulnerability in H3.3G34R/V gliomas. This finding could inform development of targeted, combination therapies for these lethal brain tumors..
Ramachandran, A.
Mehić, M.
Wasim, L.
Malinova, D.
Gori, I.
Blaszczyk, B.K.
Carvalho, D.M.
Shore, E.M.
Jones, C.
Hyvönen, M.
Tolar, P.
Hill, C.S.
(2021). Pathogenic ACVR1R206H activation by Activin A-induced receptor clustering and autophosphorylation. Embo j,
Vol.40
(14),
p. e106317.
show abstract
full text
Fibrodysplasia ossificans progressiva (FOP) and diffuse intrinsic pontine glioma (DIPG) are debilitating diseases that share causal mutations in ACVR1, a TGF-β family type I receptor. ACVR1R206H is a frequent mutation in both diseases. Pathogenic signaling via the SMAD1/5 pathway is mediated by Activin A, but how the mutation triggers aberrant signaling is not known. We show that ACVR1 is essential for Activin A-mediated SMAD1/5 phosphorylation and is activated by two distinct mechanisms. Wild-type ACVR1 is activated by the Activin type I receptors, ACVR1B/C. In contrast, ACVR1R206H activation does not require upstream kinases, but is predominantly activated via Activin A-dependent receptor clustering, which induces its auto-activation. We use optogenetics and live-imaging approaches to demonstrate Activin A-induced receptor clustering and show it requires the type II receptors ACVR2A/B. Our data provide molecular mechanistic insight into the pathogenesis of FOP and DIPG by linking the causal activating genetic mutation to disrupted signaling..
Rooney, L.
Jones, C.
(2021). Recent Advances in ALK2 Inhibitors. Acs omega,
Vol.6
(32),
pp. 20729-20734.
show abstract
full text
Activin receptor-like kinase-2 (ALK2) is a type I bone morphogenetic protein (BMP) receptor which has a role in biological processes that control the development of bone, heart, brain, and other tissue. Gain of function mutations in ALK2 have been identified in fibrodysplasia ossificans progressiva (FOP) and the childhood brain tumor, diffuse intrinsic pontine glioma (DIPG), which has given focus to the development of ALK2 inhibitors as targeted treatments. This review covers the structural features of ALK2 inhibitors which contribute to their ALK2 potency and selectivity, and the pharmacokinetic or in vivo efficacy data available to demonstrate their suitability for treating a peripheral or CNS disease..
Roig-Carles, D.
Jackson, H.
Loveson, K.F.
Mackay, A.
Mather, R.L.
Waters, E.
Manzo, M.
Alborelli, I.
Golding, J.
Jones, C.
Fillmore, H.L.
Crea, F.
(2021). The Long Non-Coding RNA H19 Drives the Proliferation of Diffuse Intrinsic Pontine Glioma with H3K27 Mutation. Int j mol sci,
Vol.22
(17).
show abstract
full text
Diffuse intrinsic pontine glioma (DIPG) is an incurable paediatric malignancy. Identifying the molecular drivers of DIPG progression is of the utmost importance. Long non-coding RNAs (lncRNAs) represent a large family of disease- and tissue-specific transcripts, whose functions have not yet been elucidated in DIPG. Herein, we studied the oncogenic role of the development-associated H19 lncRNA in DIPG. Bioinformatic analyses of clinical datasets were used to measure the expression of H19 lncRNA in paediatric high-grade gliomas (pedHGGs). The expression and sub-cellular location of H19 lncRNA were validated in DIPG cell lines. Locked nucleic acid antisense oligonucleotides were designed to test the function of H19 in DIPG cells. We found that H19 expression was higher in DIPG vs. normal brain tissue and other pedHGGs. H19 knockdown resulted in decreased cell proliferation and survival in DIPG cells. Mechanistically, H19 buffers let-7 microRNAs, resulting in the up-regulation of oncogenic let-7 target (e.g., SULF2 and OSMR). H19 is the first functionally characterized lncRNA in DIPG and a promising therapeutic candidate for treating this incurable cancer..
Izquierdo, E.
Proszek, P.
Pericoli, G.
Temelso, S.
Clarke, M.
Carvalho, D.M.
Mackay, A.
Marshall, L.V.
Carceller, F.
Hargrave, D.
Lannering, B.
Pavelka, Z.
Bailey, S.
Entz-Werle, N.
Grill, J.
Vassal, G.
Rodriguez, D.
Morgan, P.S.
Jaspan, T.
Mastronuzzi, A.
Vinci, M.
Hubank, M.
Jones, C.
(2021). Droplet digital PCR-based detection of circulating tumor DNA from pediatric high grade and diffuse midline glioma patients. Neurooncol adv,
Vol.3
(1),
p. vdab013.
show abstract
full text
BACKGROUND: The use of liquid biopsy is of potential high importance for children with high grade (HGG) and diffuse midline gliomas (DMG), particularly where surgical procedures are limited, and invasive biopsy sampling not without risk. To date, however, the evidence that detection of cell-free DNA (cfDNA) or circulating tumor DNA (ctDNA) could provide useful information for these patients has been limited, or contradictory. METHODS: We optimized droplet digital PCR (ddPCR) assays for the detection of common somatic mutations observed in pediatric HGG/DMG, and applied them to liquid biopsies from plasma, serum, cerebrospinal fluid (CSF), and cystic fluid collected from 32 patients. RESULTS: Although detectable in all biomaterial types, ctDNA presented at significantly higher levels in CSF compared to plasma and/or serum. When applied to a cohort of 127 plasma specimens from 41 patients collected from 2011 to 2018 as part of a randomized clinical trial in pediatric non-brainstem HGG/DMG, ctDNA profiling by ddPCR was of limited use due to the small volumes (mean = 0.49 mL) available. In anecdotal cases where sufficient material was available, cfDNA concentration correlated with disease progression in two examples each of poor response in H3F3A_K27M-mutant DMG, and longer survival times in hemispheric BRAF_V600E-mutant cases. CONCLUSION: Tumor-specific DNA alterations are more readily detected in CSF than plasma. Although we demonstrate the potential of the approach to assessing tumor burden, our results highlight the necessity for adequate sample collection and approach to improve detection if plasma samples are to be used..
Guerra-García, P.
Marshall, L.V.
Cockle, J.V.
Ramachandran, P.V.
Saran, F.H.
Jones, C.
Carceller, F.
(2020). Challenging the indiscriminate use of temozolomide in pediatric high-grade gliomas: A review of past, current, and emerging therapies. Pediatr blood cancer,
Vol.67
(1),
p. e28011.
show abstract
Pediatric high-grade gliomas (pHGG) constitute 8% to 12% of primary brain tumors in childhood. The most widely utilized treatment encompasses surgical resection followed by focal radiotherapy and temozolomide. However, experiences over past decades have not demonstrated improved outcomes. pHGG have been classified into different molecular subgroups defined by mutations in histone 3, IDH gene, MAPK pathway, and others, thereby providing a rationale for various targeted therapies. Additionally, immunotherapy and drug repurposing have also become attractive adjunctive treatments. This review focuses on past, present, and emerging treatments for pHGG integrating molecular research with the mainstream pediatric drug development in Europe and the United States to sketch a way forward in the development of novel therapeutic approaches. The implementation of randomized clinical trials with adaptive designs, underpinned by a robust biological rationale, and harnessing collaboration between the pharmaceutical industry, academia, regulators and patients/parents organizations will be essential to improve the outcomes for these children..
Pickles, J.C.
Fairchild, A.R.
Stone, T.J.
Brownlee, L.
Merve, A.
Yasin, S.A.
Avery, A.
Ahmed, S.W.
Ogunbiyi, O.
Gonzalez Zapata, J.
Peary, A.F.
Edwards, M.
Wilkhu, L.
Dryden, C.
Ladon, D.
Kristiansen, M.
Rowe, C.
Kurian, K.M.
Nicoll, J.A.
Mitchell, C.
Bloom, T.
Hilton, D.A.
Al-Sarraj, S.
Doey, L.
Johns, P.N.
Bridges, L.R.
Chakrabarty, A.
Ismail, A.
Rathi, N.
Syed, K.
Lammie, G.A.
Limback-Stanic, C.
Smith, C.
Torgersen, A.
Rae, F.
Hill, R.M.
Clifford, S.C.
Grabovska, Y.
Williamson, D.
Clarke, M.
Jones, C.
Capper, D.
Sill, M.
von Deimling, A.
Pfister, S.M.
Jones, D.T.
Hargrave, D.
Chalker, J.
Jacques, T.S.
(2020). DNA methylation-based profiling for paediatric CNS tumour diagnosis and treatment: a population-based study. Lancet child adolesc health,
Vol.4
(2),
pp. 121-130.
show abstract
BACKGROUND: Marked variation exists in the use of genomic data in tumour diagnosis, and optimal integration with conventional diagnostic technology remains uncertain despite several studies reporting improved diagnostic accuracy, selection for targeted treatments, and stratification for trials. Our aim was to assess the added value of molecular profiling in routine clinical practice and the impact on conventional and experimental treatments. METHODS: This population-based study assessed the diagnostic and clinical use of DNA methylation-based profiling in childhood CNS tumours using two large national cohorts in the UK. In the diagnostic cohort-which included routinely diagnosed CNS tumours between Sept 1, 2016, and Sept 1, 2018-we assessed how the methylation profile altered or refined diagnosis in routine clinical practice and estimated how this would affect standard patient management. For the archival cohort of diagnostically difficult cases, we established how many cases could be solved using modern standard pathology, how many could only be solved using the methylation profile, and how many remained unsolvable. FINDINGS: Of 484 patients younger than 20 years with CNS tumours, 306 had DNA methylation arrays requested by the neuropathologist and were included in the diagnostic cohort. Molecular profiling added a unique contribution to clinical diagnosis in 107 (35%; 95% CI 30-40) of 306 cases in routine diagnostic practice-providing additional molecular subtyping data in 99 cases, amended the final diagnosis in five cases, and making potentially significant predictions in three cases. We estimated that it could change conventional management in 11 (4%; 95% CI 2-6) of 306 patients. Among 195 historically difficult-to-diagnose tumours in the archival cohort, 99 (51%) could be diagnosed using standard methods, with the addition of methylation profiling solving a further 34 (17%) cases. The remaining 62 (32%) cases were unresolved despite specialist pathology and methylation profiling. INTERPRETATION: Together, these data provide estimates of the impact that could be expected from routine implementation of genomic profiling into clinical practice, and indicate limitations where additional techniques will be required. We conclude that DNA methylation arrays are a useful diagnostic adjunct for childhood CNS tumours. FUNDING: The Brain Tumour Charity, Children with Cancer UK, Great Ormond Street Hospital Children's Charity, Olivia Hodson Cancer Fund, Cancer Research UK, and the National Institute of Health Research..
Li, B.K.
Vasiljevic, A.
Dufour, C.
Yao, F.
Ho, B.L.
Lu, M.
Hwang, E.I.
Gururangan, S.
Hansford, J.R.
Fouladi, M.
Nobusawa, S.
Laquerriere, A.
Delisle, M.-.
Fangusaro, J.
Forest, F.
Toledano, H.
Solano-Paez, P.
Leary, S.
Birks, D.
Hoffman, L.M.
Szathmari, A.
Faure-Conter, C.
Fan, X.
Catchpoole, D.
Zhou, L.
Schultz, K.A.
Ichimura, K.
Gauchotte, G.
Jabado, N.
Jones, C.
Loussouarn, D.
Mokhtari, K.
Rousseau, A.
Ziegler, D.S.
Tanaka, S.
Pomeroy, S.L.
Gajjar, A.
Ramaswamy, V.
Hawkins, C.
Grundy, R.G.
Hill, D.A.
Bouffet, E.
Huang, A.
Jouvet, A.
(2020). Pineoblastoma segregates into molecular sub-groups with distinct clinico-pathologic features: a Rare Brain Tumor Consortium registry study. Acta neuropathol,
Vol.139
(2),
pp. 223-241.
show abstract
full text
Pineoblastomas (PBs) are rare, aggressive pediatric brain tumors of the pineal gland with modest overall survival despite intensive therapy. We sought to define the clinical and molecular spectra of PB to inform new treatment approaches for this orphan cancer. Tumor, blood, and clinical data from 91 patients with PB or supratentorial primitive neuroectodermal tumor (sPNETs/CNS-PNETs), and 2 pineal parenchymal tumors of intermediate differentiation (PPTIDs) were collected from 29 centres in the Rare Brain Tumor Consortium. We used global DNA methylation profiling to define a core group of PB from 72/93 cases, which were delineated into five molecular sub-groups. Copy number, whole exome and targeted sequencing, and miRNA expression analyses were used to evaluate the clinico-pathologic significance of each sub-group. Tumors designated as group 1 and 2 almost exclusively exhibited deleterious homozygous loss-of-function alterations in miRNA biogenesis genes (DICER1, DROSHA, and DGCR8) in 62 and 100% of group 1 and 2 tumors, respectively. Recurrent alterations of the oncogenic MYC-miR-17/92-RB1 pathway were observed in the RB and MYC sub-group, respectively, characterized by RB1 loss with gain of miR-17/92, and recurrent gain or amplification of MYC. PB sub-groups exhibited distinct clinical features: group 1-3 arose in older children (median ages 5.2-14.0 years) and had intermediate to excellent survival (5-year OS of 68.0-100%), while Group RB and MYC PB patients were much younger (median age 1.3-1.4 years) with dismal survival (5-year OS 37.5% and 28.6%, respectively). We identified age < 3 years at diagnosis, metastatic disease, omission of upfront radiation, and chr 16q loss as significant negative prognostic factors across all PBs. Our findings demonstrate that PB exhibits substantial molecular heterogeneity with sub-group-associated clinical phenotypes and survival. In addition to revealing novel biology and therapeutics, molecular sub-grouping of PB can be exploited to reduce treatment intensity for patients with favorable biology tumors..
Simpson, A.D.
Soo, Y.W.
Rieunier, G.
Aleksic, T.
Ansorge, O.
Jones, C.
Macaulay, V.M.
(2020). Type 1 IGF receptor associates with adverse outcome and cellular radioresistance in paediatric high-grade glioma. Br j cancer,
Vol.122
(5),
pp. 624-629.
show abstract
full text
High-grade glioma (HGG) is highly resistant to therapy, prompting us to investigate the contribution of insulin-like growth factor receptor (IGF-1R), linked with radioresistance in other cancers. IGF-1R immunohistochemistry in 305 adult HGG (aHGG) and 103 paediatric/young adult HGG (pHGG) cases revealed significant association with adverse survival in pHGG, with median survival of 13.5 vs 29 months for pHGGs with moderate/strong vs negative/weak IGF-1R (p = 0.011). Secondly, we tested IGF-1R inhibitor BMS-754807 in HGG cells, finding minimal radiosensitisation of 2/3 aHGG cell lines (dose enhancement ratios DERs < 1.60 at 2-8 Gy), and greater radiosensitisation of 2/2 pHGG cell lines (DERs ≤ 4.16). BMS-754807 did not influence radiation-induced apoptosis but perturbed the DNA damage response with altered induction/resolution of γH2AX, 53BP1 and RAD51 foci. These data indicate that IGF-1R promotes radioresistance in pHGG, potentially contributing to the association of IGF-1R with adverse outcome and suggesting IGF-1R as a candidate treatment target in pHGG..
Pinto, F.
Costa, Â.M.
Santos, G.C.
Matsushita, M.M.
Costa, S.
Silva, V.A.
Miranda-Gonçalves, V.
Lopes, C.M.
Clara, C.A.
Becker, A.P.
Neder, L.
Hajj, G.N.
da Cunha, I.W.
Jones, C.
Andrade, R.P.
Reis, R.M.
(2020). The T-box transcription factor brachyury behaves as a tumor suppressor in gliomas. J pathol,
Vol.251
(1),
pp. 87-99.
show abstract
The oncogene brachyury (TBXT) is a T-box transcription factor that is overexpressed in multiple solid tumors and is associated with tumor aggressiveness and poor patient prognosis. Gliomas comprise the most common and aggressive group of brain tumors, and at the present time the functional and clinical impact of brachyury expression has not been investigated previously in these neoplasms. Brachyury expression (mRNA and protein) was assessed in normal brain (n = 67), glioma tissues (n = 716) and cell lines (n = 42), and further in silico studies were undertaken using genomic databases totaling 3115 samples. Our glioma samples were analyzed for copy number (n = 372), promoter methylation status (n = 170), and mutation status (n = 1569 tissues and n = 52 cell lines) of the brachyury gene. The prognostic impact of brachyury expression was studied in 1524 glioma patient tumors. The functional impact of brachyury on glioma proliferation, viability, and cell death was evaluated both in vitro and in vivo. Brachyury was expressed in the normal brain, and significantly downregulated in glioma tissues. Loss of brachyury was associated with tumor aggressiveness and poor survival in glioma patients. Downregulation of brachyury was not associated with gene deletion, promoter methylation, or inactivating point mutations. Brachyury re-expression in glioma cells was found to decrease glioma tumorigenesis by induction of autophagy. These data strongly suggest that brachyury behaves as a tumor suppressor gene in gliomas by modulating autophagy. It is important to note that brachyury constitutes an independent positive biomarker of patient prognosis. Our findings indicate that the role of brachyury in tumorigenesis may be tissue-dependent and demands additional investigation to guide rational interventions. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd..
Clarke, M.
Mackay, A.
Ismer, B.
Pickles, J.C.
Tatevossian, R.G.
Newman, S.
Bale, T.A.
Stoler, I.
Izquierdo, E.
Temelso, S.
Carvalho, D.M.
Molinari, V.
Burford, A.
Howell, L.
Virasami, A.
Fairchild, A.R.
Avery, A.
Chalker, J.
Kristiansen, M.
Haupfear, K.
Dalton, J.D.
Orisme, W.
Wen, J.
Hubank, M.
Kurian, K.M.
Rowe, C.
Maybury, M.
Crosier, S.
Knipstein, J.
Schüller, U.
Kordes, U.
Kram, D.E.
Snuderl, M.
Bridges, L.
Martin, A.J.
Doey, L.J.
Al-Sarraj, S.
Chandler, C.
Zebian, B.
Cairns, C.
Natrajan, R.
Boult, J.K.
Robinson, S.P.
Sill, M.
Dunkel, I.J.
Gilheeney, S.W.
Rosenblum, M.K.
Hughes, D.
Proszek, P.Z.
Macdonald, T.J.
Preusser, M.
Haberler, C.
Slavc, I.
Packer, R.
Ng, H.-.
Caspi, S.
Popović, M.
Faganel Kotnik, B.
Wood, M.D.
Baird, L.
Davare, M.A.
Solomon, D.A.
Olsen, T.K.
Brandal, P.
Farrell, M.
Cryan, J.B.
Capra, M.
Karremann, M.
Schittenhelm, J.
Schuhmann, M.U.
Ebinger, M.
Dinjens, W.N.
Kerl, K.
Hettmer, S.
Pietsch, T.
Andreiuolo, F.
Driever, P.H.
Korshunov, A.
Hiddingh, L.
Worst, B.C.
Sturm, D.
Zuckermann, M.
Witt, O.
Bloom, T.
Mitchell, C.
Miele, E.
Colafati, G.S.
Diomedi-Camassei, F.
Bailey, S.
Moore, A.S.
Hassall, T.E.
Lowis, S.P.
Tsoli, M.
Cowley, M.J.
Ziegler, D.S.
Karajannis, M.A.
Aquilina, K.
Hargrave, D.R.
Carceller, F.
Marshall, L.V.
von Deimling, A.
Kramm, C.M.
Pfister, S.M.
Sahm, F.
Baker, S.J.
Mastronuzzi, A.
Carai, A.
Vinci, M.
Capper, D.
Popov, S.
Ellison, D.W.
Jacques, T.S.
Jones, D.T.
Jones, C.
(2020). Infant High-Grade Gliomas Comprise Multiple Subgroups Characterized by Novel Targetable Gene Fusions and Favorable Outcomes. Cancer discov,
Vol.10
(7),
pp. 942-963.
show abstract
full text
Infant high-grade gliomas appear clinically distinct from their counterparts in older children, indicating that histopathologic grading may not accurately reflect the biology of these tumors. We have collected 241 cases under 4 years of age, and carried out histologic review, methylation profiling, and custom panel, genome, or exome sequencing. After excluding tumors representing other established entities or subgroups, we identified 130 cases to be part of an "intrinsic" spectrum of disease specific to the infant population. These included those with targetable MAPK alterations, and a large proportion of remaining cases harboring gene fusions targeting ALK (n = 31), NTRK1/2/3 (n = 21), ROS1 (n = 9), and MET (n = 4) as their driving alterations, with evidence of efficacy of targeted agents in the clinic. These data strongly support the concept that infant gliomas require a change in diagnostic practice and management. SIGNIFICANCE: Infant high-grade gliomas in the cerebral hemispheres comprise novel subgroups, with a prevalence of ALK, NTRK1/2/3, ROS1, or MET gene fusions. Kinase fusion-positive tumors have better outcome and respond to targeted therapy clinically. Other subgroups have poor outcome, with fusion-negative cases possibly representing an epigenetically driven pluripotent stem cell phenotype.See related commentary by Szulzewsky and Cimino, p. 904.This article is highlighted in the In This Issue feature, p. 890..
Jiao, F.
Li, Z.
He, C.
Xu, W.
Yang, G.
Liu, T.
Shen, H.
Cai, J.
Anastas, J.N.
Mao, Y.
Yu, Y.
Lan, F.
Shi, Y.G.
Jones, C.
Xu, Y.
Baker, S.J.
Shi, Y.
Guo, R.
(2020). RACK7 recognizes H3 3G34R mutation to suppress expression of MHC class II complex components and their delivery pathway in pediatric glioblastoma. Sci adv,
Vol.6
(29),
p. eaba2113.
show abstract
full text
Histone H3 point mutations have been identified in incurable pediatric brain cancers, but the mechanisms through which these mutations drive tumorigenesis are incompletely understood. Here, we provide evidence that RACK7 (ZMYND8) recognizes the histone H3.3 patient mutation (H3.3G34R) in vitro and in vivo. We show that RACK7 binding to H3.3G34R suppresses transcription of CIITA, which is the master regulator of MHC (major histocompatibility complex) class II molecules and genes involved in vesicular transport of MHC class II molecules to the cell surface, resulting in suppression of MHC class II molecule expression and transport. CRISPR-based knock-in correction of the H3.3G34R mutation in human pediatric glioblastoma (pGBM) cells significantly reduces overall RACK7 chromatin binding and derepresses the same set of genes as does knocking out RACK7 in the H3.3G34R pGBM cells. By demonstrating that H3.3G34R and RACK7 work together, our findings suggest a potential molecular mechanism by which H3.3G34R promotes cancer..
Dodgshun, A.J.
Fukuoka, K.
Edwards, M.
Bianchi, V.J.
Das, A.
Sexton-Oates, A.
Larouche, V.
Vanan, M.I.
Lindhorst, S.
Yalon, M.
Mason, G.
Crooks, B.
Constantini, S.
Massimino, M.
Chiaravalli, S.
Ramdas, J.
Mason, W.
Ashraf, S.
Farah, R.
Van Damme, A.
Opocher, E.
Hamid, S.A.
Ziegler, D.S.
Samuel, D.
Cole, K.A.
Tomboc, P.
Stearns, D.
Thomas, G.A.
Lossos, A.
Sullivan, M.
Hansford, J.R.
Mackay, A.
Jones, C.
Jones, D.T.
Ramaswamy, V.
Hawkins, C.
Bouffet, E.
Tabori, U.
(2020). Germline-driven replication repair-deficient high-grade gliomas exhibit unique hypomethylation patterns. Acta neuropathol,
Vol.140
(5),
pp. 765-776.
show abstract
Replication repair deficiency (RRD) leading to hypermutation is an important driving mechanism of high-grade glioma (HGG) occurring predominantly in the context of germline mutations in RRD-associated genes. Although HGG presents specific patterns of DNA methylation corresponding to oncogenic mutations, this has not been well studied in replication repair-deficient tumors. We analyzed 51 HGG arising in the background of gene mutations in RRD utilizing either 450 k or 850 k methylation arrays. These were compared with HGG not known to be from patients with RRD. RRD HGG harboring secondary mutations in glioma genes such as IDH1 and H3F3A displayed a methylation pattern corresponding to these methylation subgroups. Strikingly, RRD HGG lacking these known secondary mutations clustered together with an incompletely described group of HGG previously labeled "Wild type-C" or "Paediatric RTK 1". Independent analysis of two comparator HGG cohorts showed that other RRD/hypermutant tumors clustered within these subgroups, suggesting that undiagnosed RRD may be driving some HGG clustering in this location. RRD HGG displayed a unique CpG Island Demethylator Phenotype in contrast to the CpG Island Methylator Phenotype described in other cancers. Hypomethylation was enriched at gene promoters with prominent demethylation in genes and pathways critical to cellular survival including cell cycle, gene expression, cellular metabolism, and organization. These data suggest that methylation arrays may provide diagnostic information for the detection of RRD HGG. Furthermore, our findings highlight the unique natural selection pressures in these highly dysregulated, hypermutant cancers and provide the novel impact of hypermutation and RRD on the cancer epigenome..
Rodriguez Gutierrez, D.
Jones, C.
Varlet, P.
Mackay, A.
Warren, D.
Warmuth-Metz, M.
Aliaga, E.S.
Calmon, R.
Hargrave, D.R.
Cañete, A.
Massimino, M.
Azizi, A.A.
Le Deley, M.-.
Saran, F.
Rousseau, R.F.
Zahlmann, G.
Garcia, J.
Vassal, G.
Grill, J.
Morgan, P.S.
Jaspan, T.
(2020). Radiological Evaluation of Newly Diagnosed Non-Brainstem Pediatric High-Grade Glioma in the HERBY Phase II Trial. Clin cancer res,
Vol.26
(8),
pp. 1856-1865.
show abstract
full text
PURPOSE: The HERBY trial evaluated the benefit of the addition of the antiangiogenic agent Bevacizumab (BEV) to radiotherapy/temozolomide (RT/TMZ) in pediatric patients with newly diagnosed non-brainstem high-grade glioma (HGG). The work presented here aims to correlate imaging characteristics and outcome measures with pathologic and molecular data. EXPERIMENTAL DESIGN: Radiological, pathologic, and molecular data were correlated with trial clinical information to retrospectively re-evaluate event-free survival (EFS) and overall survival (OS). RESULTS: One-hundred thirteen patients were randomized to the RT/TMZ arm (n = 54) or the RT/TMZ+BEV (BEV arm; n = 59). The tumor arose in the cerebral hemispheres in 68 patients (Cerebral group) and a midline location in 45 cases (Midline group). Pathologic diagnosis was available in all cases and molecular data in 86 of 113. H3 K27M histone mutations were present in 23 of 32 Midline cases and H3 G34R/V mutations in 7 of 54 Cerebral cases. Total/near-total resection occurred in 44 of 68 (65%) Cerebral cases but in only 5 of 45 (11%) Midline cases (P < 0.05). Leptomeningeal metastases (27 cases, 13 with subependymal spread) at relapse were more frequent in Midline (17/45) than in Cerebral tumors (10/68, P < 0.05). Mean OS (14.1 months) and EFS (9.0 months) in Midline tumors were significantly lower than mean OS (20.7 months) and EFS (14.9 months) in Cerebral tumors (P < 0.05). Pseudoprogression occurred in 8 of 111 (6.2%) cases. CONCLUSIONS: This study has shown that the poor outcome of midline tumors (compared with cerebral) may be related to (1) lesser surgical resection, (2) H3 K27M histone mutations, and (3) higher leptomeningeal dissemination..
Pericoli, G.
Petrini, S.
Giorda, E.
Ferretti, R.
Ajmone-Cat, M.A.
Court, W.
Conti, L.A.
De Simone, R.
Bencivenga, P.
Palma, A.
Di Giannatale, A.
Jones, C.
Carai, A.
Mastronuzzi, A.
de Billy, E.
Locatelli, F.
Vinci, M.
(2020). Integration of Multiple Platforms for the Analysis of Multifluorescent Marking Technology Applied to Pediatric GBM and DIPG. Int j mol sci,
Vol.21
(18).
show abstract
full text
The intratumor heterogeneity represents one of the most difficult challenges for the development of effective therapies to treat pediatric glioblastoma (pGBM) and diffuse intrinsic pontine glioma (DIPG). These brain tumors are composed of heterogeneous cell subpopulations that coexist and cooperate to build a functional network responsible for their aggressive phenotype. Understanding the cellular and molecular mechanisms sustaining such network will be crucial for the identification of new therapeutic strategies. To study more in-depth these mechanisms, we sought to apply the Multifluorescent Marking Technology. We generated multifluorescent pGBM and DIPG bulk cell lines randomly expressing six different fluorescent proteins and from which we derived stable optical barcoded single cell-derived clones. In this study, we focused on the application of the Multifluorescent Marking Technology in 2D and 3D in vitro/ex vivo culture systems. We discuss how we integrated different multimodal fluorescence analysis platforms, identifying their strengths and limitations, to establish the tools that will enable further studies on the intratumor heterogeneity and interclonal interactions in pGBM and DIPG..
Menezes, W.P.
Silva, V.A.
Gomes, I.N.
Rosa, M.N.
Spina, M.L.
Carloni, A.C.
Alves, A.L.
Melendez, M.
Almeida, G.C.
Silva, L.S.
Clara, C.
da Cunha, I.W.
Hajj, G.N.
Jones, C.
Bidinotto, L.T.
Reis, R.M.
(2020). Loss of 5'-Methylthioadenosine Phosphorylase (MTAP) is Frequent in High-Grade Gliomas; Nevertheless, it is Not Associated with Higher Tumor Aggressiveness. Cells,
Vol.9
(2).
show abstract
full text
The 5'-methylthioadenosine phosphorylase (MTAP) gene is located in the chromosomal region 9p21. MTAP deletion is a frequent event in a wide variety of human cancers; however, its biological role in tumorigenesis remains unclear. The purpose of this study was to characterize the MTAP expression profile in a series of gliomas and to associate it with patients' clinicopathological features. Moreover, we sought to evaluate, through glioma gene-edited cell lines, the biological impact of MTAP in gliomas. MTAP expression was evaluated in 507 glioma patients by immunohistochemistry (IHC), and the expression levels were associated with patients' clinicopathological features. Furthermore, an in silico study was undertaken using genomic databases totalizing 350 samples. In glioma cell lines, MTAP was edited, and following MTAP overexpression and knockout (KO), a transcriptome analysis was performed by NanoString Pan-Cancer Pathways panel. Moreover, MTAP's role in glioma cell proliferation, migration, and invasion was evaluated. Homozygous deletion of 9p21 locus was associated with a reduction of MTAP mRNA expression in the TCGA (The Cancer Genome Atlas) - glioblastoma dataset (p < 0.01). In addition, the loss of MTAP expression was markedly high in high-grade gliomas (46.6% of cases) determined by IHC and Western blotting (40% of evaluated cell lines). Reduced MTAP expression was associated with a better prognostic in the adult glioblastoma dataset (p < 0.001). Nine genes associated with five pathways were differentially expressed in MTAP-knockout (KO) cells, with six upregulated and three downregulated in MTAP. Analysis of cell proliferation, migration, and invasion did not show any significant differences between MTAP gene-edited and control cells. Our results integrating data from patients as well as in silico and in vitro models provide evidence towards the lack of strong biological importance of MTAP in gliomas. Despite the frequent loss of MTAP, it seems not to have a clinical impact in survival and does not act as a canonic tumor suppressor gene in gliomas..
Tsoli, M.
Shen, H.
Mayoh, C.
Franshaw, L.
Ehteda, A.
Upton, D.
Carvalho, D.
Vinci, M.
Meel, M.H.
van Vuurden, D.
Plessier, A.
Castel, D.
Drissi, R.
Farrell, M.
Cryan, J.
Crimmins, D.
Caird, J.
Pears, J.
Francis, S.
Ludlow, L.E.
Carai, A.
Mastronuzzi, A.
Liu, B.
Hansford, J.
Gottardo, N.
Hassall, T.
Kirby, M.
Fouladi, M.
Hawkins, C.
Monje, M.
Grill, J.
Jones, C.
Hulleman, E.
Ziegler, D.S.
(2019). International experience in the development of patient-derived xenograft models of diffuse intrinsic pontine glioma. J neurooncol,
Vol.141
(2),
pp. 253-263.
show abstract
PURPOSE: Diffuse intrinsic pontine glioma is the most aggressive form of high grade glioma in children with no effective therapies. There have been no improvements in survival in part due poor understanding of underlying biology, and lack of representative in vitro and in vivo models. Recently, it has been found feasible to use both biopsy and autopsy tumors to generate cultures and xenograft models. METHODS: To further model development, we evaluated the collective international experience from 8 collaborating centers to develop DIPG pre-clinical models from patient-derived autopsies and biopsies. Univariate and multivariate analysis was performed to determine key factors associated with the success of in vitro and in vivo PDX development. RESULTS: In vitro cultures were successfully established from 57% of samples (84.2% of biopsies and 38.2% of autopsies). Samples transferred in DMEM media were more likely to establish successful culture than those transported in Hibernate A. In vitro cultures were more successful from biopsies (84.2%) compared with autopsies (38.2%) and as monolayer on laminin-coated plates than as neurospheres. Primary cultures successfully established from autopsy samples were more likely to engraft in animal models than cultures established from biopsies (86.7% vs. 47.4%). Collectively, tumor engraftment was more successful when DIPG samples were directly implanted in mice (68%), rather than after culturing (40.7%). CONCLUSION: This multi-center study provides valuable information on the success rate of establishing patient-derived pre-clinical models of DIPG. The results can lead to further optimization of DIPG model development and ultimately assist in the investigation of new therapies for this aggressive pediatric brain tumor..
Carvalho, D.
Taylor, K.R.
Olaciregui, N.G.
Molinari, V.
Clarke, M.
Mackay, A.
Ruddle, R.
Henley, A.
Valenti, M.
Hayes, A.
Brandon, A.D.
Eccles, S.A.
Raynaud, F.
Boudhar, A.
Monje, M.
Popov, S.
Moore, A.S.
Mora, J.
Cruz, O.
Vinci, M.
Brennan, P.E.
Bullock, A.N.
Carcaboso, A.M.
Jones, C.
(2019). ALK2 inhibitors display beneficial effects in preclinical models of ACVR1 mutant diffuse intrinsic pontine glioma. Commun biol,
Vol.2
(1),
p. 156.
show abstract
full text
Diffuse intrinsic pontine glioma (DIPG) is a lethal childhood brainstem tumour, with a quarter of patients harbouring somatic mutations in ACVR1, encoding the serine/threonine kinase ALK2. Despite being an amenable drug target, little has been done to-date to systematically evaluate the role of ACVR1 in DIPG, nor to screen currently available inhibitors in patient-derived tumour models. Here we show the dependence of DIPG cells on the mutant receptor, and the preclinical efficacy of two distinct chemotypes of ALK2 inhibitor in vitro and in vivo. We demonstrate the pyrazolo[1,5-a]pyrimidine LDN-193189 and the pyridine LDN-214117 to be orally bioavailable and well-tolerated, with good brain penetration. Treatment of immunodeprived mice bearing orthotopic xenografts of H3.3K27M, ACVR1R206H mutant HSJD-DIPG-007 cells with 25 mg/kg LDN-193189 or LDN-214117 for 28 days extended survival compared with vehicle controls. Development of ALK2 inhibitors with improved potency, selectivity and advantageous pharmacokinetic properties may play an important role in therapy for DIPG patients..
George, S.L.
Izquierdo, E.
Campbell, J.
Koutroumanidou, E.
Proszek, P.
Jamal, S.
Hughes, D.
Yuan, L.
Marshall, L.V.
Carceller, F.
Chisholm, J.C.
Vaidya, S.
Mandeville, H.
Angelini, P.
Wasti, A.
Bexelius, T.
Thway, K.
Gatz, S.A.
Clarke, M.
Al-Lazikani, B.
Barone, G.
Anderson, J.
Tweddle, D.A.
Gonzalez, D.
Walker, B.A.
Barton, J.
Depani, S.
Eze, J.
Ahmed, S.W.
Moreno, L.
Pearson, A.
Shipley, J.
Jones, C.
Hargrave, D.
Jacques, T.S.
Hubank, M.
Chesler, L.
(2019). A tailored molecular profiling programme for children with cancer to identify clinically actionable genetic alterations. Eur j cancer,
Vol.121,
pp. 224-235.
show abstract
full text
BACKGROUND: For children with cancer, the clinical integration of precision medicine to enable predictive biomarker-based therapeutic stratification is urgently needed. METHODS: We have developed a hybrid-capture next-generation sequencing (NGS) panel, specifically designed to detect genetic alterations in paediatric solid tumours, which gives reliable results from as little as 50 ng of DNA extracted from formalin-fixed paraffin-embedded (FFPE) tissue. In this study, we offered an NGS panel, with clinical reporting via a molecular tumour board for children with solid tumours. Furthermore, for a cohort of 12 patients, we used a circulating tumour DNA (ctDNA)-specific panel to sequence ctDNA from matched plasma samples and compared plasma and tumour findings. RESULTS: A total of 255 samples were submitted from 223 patients for the NGS panel. Using FFPE tissue, 82% of all submitted samples passed quality control for clinical reporting. At least one genetic alteration was detected in 70% of sequenced samples. The overall detection rate of clinically actionable alterations, defined by modified OncoKB criteria, for all sequenced samples was 51%. A total of 8 patients were sequenced at different stages of treatment. In 6 of these, there were differences in the genetic alterations detected between time points. Sequencing of matched ctDNA in a cohort of extracranial paediatric solid tumours also identified a high detection rate of somatic alterations in plasma. CONCLUSION: We demonstrate that tailored clinical molecular profiling of both tumour DNA and plasma-derived ctDNA is feasible for children with solid tumours. Furthermore, we show that a targeted NGS panel-based approach can identify actionable genetic alterations in a high proportion of patients..
Li, J.
Zormpas-Petridis, K.
Boult, J.K.
Reeves, E.L.
Heindl, A.
Vinci, M.
Lopes, F.
Cummings, C.
Springer, C.J.
Chesler, L.
Jones, C.
Bamber, J.C.
Yuan, Y.
Sinkus, R.
Jamin, Y.
Robinson, S.P.
(2019). Investigating the Contribution of Collagen to the Tumor Biomechanical Phenotype with Noninvasive Magnetic Resonance Elastography. Cancer res,
Vol.79
(22),
pp. 5874-5883.
show abstract
full text
Increased stiffness in the extracellular matrix (ECM) contributes to tumor progression and metastasis. Therefore, stromal modulating therapies and accompanying biomarkers are being developed to target ECM stiffness. Magnetic resonance (MR) elastography can noninvasively and quantitatively map the viscoelastic properties of tumors in vivo and thus has clear clinical applications. Herein, we used MR elastography, coupled with computational histopathology, to interrogate the contribution of collagen to the tumor biomechanical phenotype and to evaluate its sensitivity to collagenase-induced stromal modulation. Elasticity (G d) and viscosity (G l) were significantly greater for orthotopic BT-474 (G d = 5.9 ± 0.2 kPa, G l = 4.7 ± 0.2 kPa, n = 7) and luc-MDA-MB-231-LM2-4 (G d = 7.9 ± 0.4 kPa, G l = 6.0 ± 0.2 kPa, n = 6) breast cancer xenografts, and luc-PANC1 (G d = 6.9 ± 0.3 kPa, G l = 6.2 ± 0.2 kPa, n = 7) pancreatic cancer xenografts, compared with tumors associated with the nervous system, including GTML/Trp53KI/KI medulloblastoma (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 7), orthotopic luc-D-212-MG (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 7), luc-RG2 (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 5), and luc-U-87-MG (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 8) glioblastoma xenografts, intracranially propagated luc-MDA-MB-231-LM2-4 (G d = 3.7 ± 0.2 kPa, G l = 2.2 ± 0.1 kPa, n = 7) breast cancer xenografts, and Th-MYCN neuroblastomas (G d = 3.5 ± 0.2 kPa, G l = 2.3 ± 0.2 kPa, n = 5). Positive correlations between both elasticity (r = 0.72, P < 0.0001) and viscosity (r = 0.78, P < 0.0001) were determined with collagen fraction, but not with cellular or vascular density. Treatment with collagenase significantly reduced G d (P = 0.002) and G l (P = 0.0006) in orthotopic breast tumors. Texture analysis of extracted images of picrosirius red staining revealed significant negative correlations of entropy with G d (r = -0.69, P < 0.0001) and G l (r = -0.76, P < 0.0001), and positive correlations of fractal dimension with G d (r = 0.75, P < 0.0001) and G l (r = 0.78, P < 0.0001). MR elastography can thus provide sensitive imaging biomarkers of tumor collagen deposition and its therapeutic modulation. SIGNIFICANCE: MR elastography enables noninvasive detection of tumor stiffness and will aid in the development of ECM-targeting therapies..
Fons, N.R.
Sundaram, R.K.
Breuer, G.A.
Peng, S.
McLean, R.L.
Kalathil, A.N.
Schmidt, M.S.
Carvalho, D.M.
Mackay, A.
Jones, C.
Carcaboso, Á.M.
Nazarian, J.
Berens, M.E.
Brenner, C.
Bindra, R.S.
(2019). PPM1D mutations silence NAPRT gene expression and confer NAMPT inhibitor sensitivity in glioma. Nat commun,
Vol.10
(1),
p. 3790.
show abstract
full text
Pediatric high-grade gliomas are among the deadliest of childhood cancers due to limited knowledge of early driving events in their gliomagenesis and the lack of effective therapies available. In this study, we investigate the oncogenic role of PPM1D, a protein phosphatase often found truncated in pediatric gliomas such as DIPG, and uncover a synthetic lethal interaction between PPM1D mutations and nicotinamide phosphoribosyltransferase (NAMPT) inhibition. Specifically, we show that mutant PPM1D drives hypermethylation of CpG islands throughout the genome and promotes epigenetic silencing of nicotinic acid phosphoribosyltransferase (NAPRT), a key gene involved in NAD biosynthesis. Notably, PPM1D mutant cells are shown to be sensitive to NAMPT inhibitors in vitro and in vivo, within both engineered isogenic astrocytes and primary patient-derived model systems, suggesting the possible application of NAMPT inhibitors for the treatment of pediatric gliomas. Overall, our results reveal a promising approach for the targeting of PPM1D mutant tumors, and define a critical link between oncogenic driver mutations and NAD metabolism, which can be exploited for tumor-specific cell killing..
Blandin, A.-.
Durand, A.
Litzler, M.
Tripp, A.
Guérin, É.
Ruhland, E.
Obrecht, A.
Keime, C.
Fuchs, Q.
Reita, D.
Lhermitte, B.
Coca, A.
Jones, C.
Rebel, I.L.
Villa, P.
Namer, I.J.
Dontenwill, M.
Guenot, D.
Entz-Werle, N.
(2019). Hypoxic Environment and Paired Hierarchical 3D and 2D Models of Pediatric H3 3-Mutated Gliomas Recreate the Patient Tumor Complexity. Cancers (basel),
Vol.11
(12).
show abstract
full text
BACKGROUND: Pediatric high-grade gliomas (pHGGs) are facing a very dismal prognosis and representative pre-clinical models are needed for new treatment strategies. Here, we examined the relevance of collecting functional, genomic, and metabolomics data to validate patient-derived models in a hypoxic microenvironment. METHODS: From our biobank of pediatric brain tumor-derived models, we selected 11 pHGGs driven by the histone H3.3K28M mutation. We compared the features of four patient tumors to their paired cell lines and mouse xenografts using NGS (next generation sequencing), aCGH (array comparative genomic hybridization), RNA sequencing, WES (whole exome sequencing), immunocytochemistry, and HRMAS (high resolution magic angle spinning) spectroscopy. We developed a multicellular in vitro model of cell migration to mimic the brain hypoxic microenvironment. The live cell technology Incucyte© was used to assess drug responsiveness in variable oxygen conditions. RESULTS: The concurrent 2D and 3D cultures generated from the same tumor sample exhibited divergent but complementary features, recreating the patient intra-tumor complexity. Genomic and metabolomic data described the metabolic changes during pHGG progression and supported hypoxia as an important key to preserve the tumor metabolism in vitro and cell dissemination present in patients. The neurosphere features preserved tumor development and sensitivity to treatment. CONCLUSION: We proposed a novel multistep work for the development and validation of patient-derived models, considering the immature and differentiated content and the tumor microenvironment of pHGGs..
Martínez-Vélez, N.
Garcia-Moure, M.
Marigil, M.
González-Huarriz, M.
Puigdelloses, M.
Gallego Pérez-Larraya, J.
Zalacaín, M.
Marrodán, L.
Varela-Guruceaga, M.
Laspidea, V.
Aristu, J.J.
Ramos, L.I.
Tejada-Solís, S.
Díez-Valle, R.
Jones, C.
Mackay, A.
Martínez-Climent, J.A.
García-Barchino, M.J.
Raabe, E.
Monje, M.
Becher, O.J.
Junier, M.P.
El-Habr, E.A.
Chneiweiss, H.
Aldave, G.
Jiang, H.
Fueyo, J.
Patiño-García, A.
Gomez-Manzano, C.
Alonso, M.M.
(2019). The oncolytic virus Delta-24-RGD elicits an antitumor effect in pediatric glioma and DIPG mouse models. Nat commun,
Vol.10
(1),
p. 2235.
show abstract
full text
Pediatric high-grade glioma (pHGG) and diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors in desperate need of a curative treatment. Oncolytic virotherapy is emerging as a solid therapeutic approach. Delta-24-RGD is a replication competent adenovirus engineered to replicate in tumor cells with an aberrant RB pathway. This virus has proven to be safe and effective in adult gliomas. Here we report that the administration of Delta-24-RGD is safe in mice and results in a significant increase in survival in immunodeficient and immunocompetent models of pHGG and DIPGs. Our results show that the Delta-24-RGD antiglioma effect is mediated by the oncolytic effect and the immune response elicited against the tumor. Altogether, our data highlight the potential of this virus as treatment for patients with these tumors. Of clinical significance, these data have led to the start of a phase I/II clinical trial at our institution for newly diagnosed DIPG (NCT03178032)..
Grill, J.
Massimino, M.
Bouffet, E.
Azizi, A.A.
McCowage, G.
Cañete, A.
Saran, F.
Le Deley, M.-.
Varlet, P.
Morgan, P.S.
Jaspan, T.
Jones, C.
Giangaspero, F.
Smith, H.
Garcia, J.
Elze, M.C.
Rousseau, R.F.
Abrey, L.
Hargrave, D.
Vassal, G.
(2018). Phase II, Open-Label, Randomized, Multicenter Trial (HERBY) of Bevacizumab in Pediatric Patients With Newly Diagnosed High-Grade Glioma. J clin oncol,
Vol.36
(10),
pp. 951-958.
show abstract
full text
Purpose Bevacizumab (BEV) is approved in more than 60 countries for use in adults with recurrent glioblastoma. We evaluated the addition of BEV to radiotherapy plus temozolomide (RT+TMZ) in pediatric patients with newly diagnosed high-grade glioma (HGG). Methods The randomized, parallel group, multicenter, open-label HERBY trial ( ClinicalTrials.gov identifier: NCT01390948) enrolled patients age ≥ 3 years to ≤ 18 years with localized, centrally neuropathology-confirmed, nonbrainstem HGG. Eligible patients were randomly assigned to receive RT + TMZ (RT: 1.8 Gy, 5 days per week, and TMZ: 75 mg/m2 per day for 6 weeks; 4-week treatment break; then up to 12 × 28-day cycles of TMZ [cycle 1: 150 mg/m2 per day, days 1 to 5; cycles 2 to 12: 200 mg/m2 per day, days 1 to 5]) with or without BEV (10 mg/kg every 2 weeks). The primary end point was event-free survival (EFS) as assessed by a central radiology review committee that was blinded to treatment. We report findings of EFS at 12 months after the enrollment of the last patient. Results One hundred twenty-one patients were enrolled (RT+TMZ [n = 59]; BEV plus RT+TMZ [n = 62]). Central radiology review committee-assessed median EFS did not differ significantly between treatment groups (RT+TMZ, 11.8 months; 95% CI, 7.9 to 16.4 months; BEV plus RT+TMZ, 8.2 months; 95% CI, 7.8 to 12.7 months; hazard ratio, 1.44; P = .13 [stratified log-rank test]). In the overall survival analysis, the addition of BEV did not reduce the risk of death (hazard ratio, 1.23; 95% CI, 0.72 to 2.09). More patients in the BEV plus RT+TMZ group versus the RT+TMZ group experienced one or more serious adverse events (n = 35 [58%] v n = 27 [48%]), and more patients who received BEV discontinued study treatment as a result of adverse events (n = 13 [22%] v n = 3 [5%]). Conclusion Adding BEV to RT+TMZ did not improve EFS in pediatric patients with newly diagnosed HGG. Our findings were not comparable to those of previous adult trials, which highlights the importance of performing pediatric-specific studies..
Oyen, W.J.
Jones, C.
(2018). Immuno-PET in Pontine Glioma: More Than Meets the Eye?. J nucl med,
Vol.59
(4),
pp. 610-611.
full text
Deng, H.
Zeng, J.
Zhang, T.
Gong, L.
Zhang, H.
Cheung, E.
Jones, C.
Li, G.
(2018). Histone H3 3K27M Mobilizes Multiple Cancer/Testis (CT) Antigens in Pediatric Glioma. Mol cancer res,
Vol.16
(4),
pp. 623-633.
show abstract
full text
Lysine to methionine mutations at position 27 (K27M) in the histone H3 (H3.3 and H3.1) are highly prevalent in pediatric high-grade gliomas (HGG) that arise in the midline of the central nervous system. H3K27M perturbs the activity of polycomb repressor complex 2 and correlates with DNA hypomethylation; however, the pathways whereby H3K27M drives the development of pediatric HGG remain poorly understood. To understand the mechanism of pediatric HGG development driven by H3.3K27M and discover potential therapeutic targets or biomarkers, we established pediatric glioma cell model systems harboring H3.3K27M and performed microarray analysis. H3.3K27M caused the upregulation of multiple cancer/testis (CT) antigens, such as ADAMTS1, ADAM23, SPANXA1, SPANXB1/2, IL13RA2, VCY, and VCX3A, in pediatric glioma cells. Chromatin immunoprecipitation analysis from H3.3K27M cells revealed decreased H3K27me3 levels and increased H3K4me3 levels on the VCX3A promoter. Knockdown of VCX3A by siRNA significantly inhibited the growth of pediatric glioma cells harboring H3.3K27M. Overexpression of VCX3A/B genes stimulated the expression of several HLA genes, including HLA-A, HLA-B, HLA-E, HLA-F, and HLA-G The expression of VCX3A in pediatric HGG was confirmed using a tissue microarray. Gene set enrichment analysis revealed that CT antigens are enriched in pediatric HGG clinical specimens with H3.3K27M, with the upregulation of IL13RA2 contributing to the enrichment significantly. These results indicate that the upregulation of CT antigens, such as VCX3A and IL13RA2, correlates with pediatric gliomagenesis. Mol Cancer Res; 16(4); 623-33. ©2018 AACR..
Castel, D.
Philippe, C.
Kergrohen, T.
Sill, M.
Merlevede, J.
Barret, E.
Puget, S.
Sainte-Rose, C.
Kramm, C.M.
Jones, C.
Varlet, P.
Pfister, S.M.
Grill, J.
Jones, D.T.
Debily, M.-.
(2018). Transcriptomic and epigenetic profiling of 'diffuse midline gliomas, H3 K27M-mutant' discriminate two subgroups based on the type of histone H3 mutated and not supratentorial or infratentorial location. Acta neuropathol commun,
Vol.6
(1),
p. 117.
show abstract
full text
Diffuse midline glioma (DMG), H3 K27M-mutant, is a new entity in the updated WHO classification grouping together diffuse intrinsic pontine gliomas and infiltrating glial neoplasms of the midline harboring the same canonical mutation at the Lysine 27 of the histones H3 tail.Two hundred and fifteen patients younger than 18 years old with centrally-reviewed pediatric high-grade gliomas (pHGG) were included in this study. Comprehensive transcriptomic (n = 140) and methylation (n = 80) profiling was performed depending on the material available, in order to assess the biological uniqueness of this new entity compared to other midline and hemispheric pHGG.Tumor classification based on gene expression (GE) data highlighted the similarity of K27M DMG independently of their location along the midline. T-distributed Stochastic Neighbor Embedding (tSNE) analysis of methylation profiling confirms the discrimination of DMG from other well defined supratentorial tumor subgroups. Patients with diffuse intrinsic pontine gliomas (DIPG) and thalamic DMG exhibited a similarly poor prognosis (11.1 and 10.8 months median overall survival, respectively). Interestingly, H3.1-K27M and H3.3-K27M primary tumor samples could be distinguished based both on their GE and DNA methylation profiles, suggesting that they might arise from a different precursor or from a different epigenetic reorganization.These differences in DNA methylation profiles were conserved in glioma stem-like cell culture models of DIPG which mimicked their corresponding primary tumor. ChIP-seq profiling of H3K27me3 in these models indicate that H3.3-K27M mutated DIPG stem cells exhibit higher levels of H3K27 trimethylation which are correlated with fewer genes expressed by RNAseq. When considering the global distribution of the H3K27me3 mark, we observed that intergenic regions were more trimethylated in the H3.3-K27M mutated cells compared to the H3.1-K27M mutated ones.H3 K27M-mutant DMG represent a homogenous group of neoplasms compared to other pediatric gliomas that could be further separated based on the type of histone H3 variant mutated and their respective epigenetic landscapes. As these characteristics drive different phenotypes, these findings may have important implication for the design of future trials in these specific types of neoplasms..
Burford, A.
Mackay, A.
Popov, S.
Vinci, M.
Carvalho, D.
Clarke, M.
Izquierdo, E.
Avery, A.
Jacques, T.S.
Ingram, W.J.
Moore, A.S.
Frawley, K.
Hassall, T.E.
Robertson, T.
(2018). THE TEN-YEAR EVOLUTIONARY TRAJECTORY OF A HIGHLY RECURRENT PAEDIATRIC HIGH GRADE NEUROEPITHELIAL TUMOUR WITH MN1:BEND2 FUSION. Neuro-oncology,
Vol.20,
pp. 40-40.
full text
Rutkowski, S.
Modena, P.
Williamson, D.
Kerl, K.
Nysom, K.
Pizer, B.
Bartels, U.
Puget, S.
Doz, F.
Michalski, A.
von Hoff, K.
Chevignard, M.
Avula, S.
Murray, M.J.
Schönberger, S.
Czech, T.
Schouten-van Meeteren, A.Y.
Kordes, U.
Kramm, C.M.
van Vuurden, D.G.
Hulleman, E.
Janssens, G.O.
Solanki, G.A.
van Veelen, M.-.
Thomale, U.
Schuhmann, M.U.
Jones, C.
Giangaspero, F.
Figarella-Branger, D.
Pietsch, T.
Clifford, S.C.
Pfister, S.M.
Van Gool, S.W.
(2018). Biological material collection to advance translational research and treatment of children with CNS tumours: position paper from the SIOPE Brain Tumour Group. The lancet oncology,
Vol.19
(8),
pp. e419-e428.
full text
Vinci, M.
Burford, A.
Molinari, V.
Kessler, K.
Popov, S.
Clarke, M.
Taylor, K.R.
Pemberton, H.N.
Lord, C.J.
Gutteridge, A.
Forshew, T.
Carvalho, D.
Marshall, L.V.
Qin, E.Y.
Ingram, W.J.
Moore, A.S.
Ng, H.-.
Trabelsi, S.
H'mida-Ben Brahim, D.
Entz-Werle, N.
Zacharoulis, S.
Vaidya, S.
Mandeville, H.C.
Bridges, L.R.
Martin, A.J.
Al-Sarraj, S.
Chandler, C.
Sunol, M.
Mora, J.
de Torres, C.
Cruz, O.
Carcaboso, A.M.
Monje, M.
Mackay, A.
Jones, C.
(2018). Functional diversity and cooperativity between subclonal populations of pediatric glioblastoma and diffuse intrinsic pontine glioma cells. Nat med,
Vol.24
(8),
pp. 1204-1215.
show abstract
full text
The failure to develop effective therapies for pediatric glioblastoma (pGBM) and diffuse intrinsic pontine glioma (DIPG) is in part due to their intrinsic heterogeneity. We aimed to quantitatively assess the extent to which this was present in these tumors through subclonal genomic analyses and to determine whether distinct tumor subpopulations may interact to promote tumorigenesis by generating subclonal patient-derived models in vitro and in vivo. Analysis of 142 sequenced tumors revealed multiple tumor subclones, spatially and temporally coexisting in a stable manner as observed by multiple sampling strategies. We isolated genotypically and phenotypically distinct subpopulations that we propose cooperate to enhance tumorigenicity and resistance to therapy. Inactivating mutations in the H4K20 histone methyltransferase KMT5B (SUV420H1), present in <1% of cells, abrogate DNA repair and confer increased invasion and migration on neighboring cells, in vitro and in vivo, through chemokine signaling and modulation of integrins. These data indicate that even rare tumor subpopulations may exert profound effects on tumorigenesis as a whole and may represent a new avenue for therapeutic development. Unraveling the mechanisms of subclonal diversity and communication in pGBM and DIPG will be an important step toward overcoming barriers to effective treatments..
Hoffman, L.M.
Veldhuijzen van Zanten, S.E.
Colditz, N.
Baugh, J.
Chaney, B.
Hoffmann, M.
Lane, A.
Fuller, C.
Miles, L.
Hawkins, C.
Bartels, U.
Bouffet, E.
Goldman, S.
Leary, S.
Foreman, N.K.
Packer, R.
Warren, K.E.
Broniscer, A.
Kieran, M.W.
Minturn, J.
Comito, M.
Broxson, E.
Shih, C.-.
Khatua, S.
Chintagumpala, M.
Carret, A.S.
Escorza, N.Y.
Hassall, T.
Ziegler, D.S.
Gottardo, N.
Dholaria, H.
Doughman, R.
Benesch, M.
Drissi, R.
Nazarian, J.
Jabado, N.
Boddaert, N.
Varlet, P.
Giraud, G.
Castel, D.
Puget, S.
Jones, C.
Hulleman, E.
Modena, P.
Giagnacovo, M.
Antonelli, M.
Pietsch, T.
Gielen, G.H.
Jones, D.T.
Sturm, D.
Pfister, S.M.
Gerber, N.U.
Grotzer, M.A.
Pfaff, E.
von Bueren, A.O.
Hargrave, D.
Solanki, G.A.
Jadrijevic Cvrlje, F.
Kaspers, G.J.
Vandertop, W.P.
Grill, J.
Bailey, S.
Biassoni, V.
Massimino, M.
Calmon, R.
Sanchez, E.
Bison, B.
Warmuth-Metz, M.
Leach, J.
Jones, B.
van Vuurden, D.G.
Kramm, C.M.
Fouladi, M.
(2018). Clinical, Radiologic, Pathologic, and Molecular Characteristics of Long-Term Survivors of Diffuse Intrinsic Pontine Glioma (DIPG): A Collaborative Report From the International and European Society for Pediatric Oncology DIPG Registries. J clin oncol,
,
pp. JCO2017759308-JCO2017759308.
show abstract
full text
Purpose Diffuse intrinsic pontine glioma (DIPG) is a brainstem malignancy with a median survival of < 1 year. The International and European Society for Pediatric Oncology DIPG Registries collaborated to compare clinical, radiologic, and histomolecular characteristics between short-term survivors (STSs) and long-term survivors (LTSs). Materials and Methods Data abstracted from registry databases included patients from North America, Australia, Germany, Austria, Switzerland, the Netherlands, Italy, France, the United Kingdom, and Croatia. Results Among 1,130 pediatric and young adults with radiographically confirmed DIPG, 122 (11%) were excluded. Of the 1,008 remaining patients, 101 (10%) were LTSs (survival ≥ 2 years). Median survival time was 11 months (interquartile range, 7.5 to 16 months), and 1-, 2-, 3-, 4-, and 5-year survival rates were 42.3% (95% CI, 38.1% to 44.1%), 9.6% (95% CI, 7.8% to 11.3%), 4.3% (95% CI, 3.2% to 5.8%), 3.2% (95% CI, 2.4% to 4.6%), and 2.2% (95% CI, 1.4% to 3.4%), respectively. LTSs, compared with STSs, more commonly presented at age < 3 or > 10 years (11% v 3% and 33% v 23%, respectively; P < .001) and with longer symptom duration ( P < .001). STSs, compared with LTSs, more commonly presented with cranial nerve palsy (83% v 73%, respectively; P = .008), ring enhancement (38% v 23%, respectively; P = .007), necrosis (42% v 26%, respectively; P = .009), and extrapontine extension (92% v 86%, respectively; P = .04). LTSs more commonly received systemic therapy at diagnosis (88% v 75% for STSs; P = .005). Biopsies and autopsies were performed in 299 patients (30%) and 77 patients (10%), respectively; 181 tumors (48%) were molecularly characterized. LTSs were more likely to harbor a HIST1H3B mutation (odds ratio, 1.28; 95% CI, 1.1 to 1.5; P = .002). Conclusion We report clinical, radiologic, and molecular factors that correlate with survival in children and young adults with DIPG, which are important for risk stratification in future clinical trials..
Mackay, A.
Burford, A.
Molinari, V.
Jones, D.T.
Izquierdo, E.
Brouwer-Visser, J.
Giangaspero, F.
Haberler, C.
Pietsch, T.
Jacques, T.S.
Figarella-Branger, D.
Rodriguez, D.
Morgan, P.S.
Raman, P.
Waanders, A.J.
Resnick, A.C.
Massimino, M.
Garrè, M.L.
Smith, H.
Capper, D.
Pfister, S.M.
Würdinger, T.
Tam, R.
Garcia, J.
Thakur, M.D.
Vassal, G.
Grill, J.
Jaspan, T.
Varlet, P.
Jones, C.
(2018). Molecular, Pathological, Radiological, and Immune Profiling of Non-brainstem Pediatric High-Grade Glioma from the HERBY Phase II Randomized Trial. Cancer cell,
Vol.33
(5),
pp. 829-842.e5.
show abstract
full text
The HERBY trial was a phase II open-label, randomized, multicenter trial evaluating bevacizumab (BEV) in addition to temozolomide/radiotherapy in patients with newly diagnosed non-brainstem high-grade glioma (HGG) between the ages of 3 and 18 years. We carried out comprehensive molecular analysis integrated with pathology, radiology, and immune profiling. In post-hoc subgroup analysis, hypermutator tumors (mismatch repair deficiency and somatic POLE/POLD1 mutations) and those biologically resembling pleomorphic xanthoastrocytoma ([PXA]-like, driven by BRAF_V600E or NF1 mutation) had significantly more CD8+ tumor-infiltrating lymphocytes, and longer survival with the addition of BEV. Histone H3 subgroups (hemispheric G34R/V and midline K27M) had a worse outcome and were immune cold. Future clinical trials will need to take into account the diversity represented by the term "HGG" in the pediatric population..
Burford, A.
Mackay, A.
Popov, S.
Vinci, M.
Carvalho, D.
Clarke, M.
Izquierdo, E.
Avery, A.
Jacques, T.S.
Ingram, W.J.
Moore, A.S.
Frawley, K.
Hassall, T.E.
Robertson, T.
Jones, C.
(2018). The ten-year evolutionary trajectory of a highly recurrent paediatric high grade neuroepithelial tumour with MN1:BEND2 fusion. Sci rep,
Vol.8
(1),
p. 1032.
show abstract
full text
Astroblastomas are rare brain tumours which predominate in children and young adults, and have a controversial claim as a distinct entity, with no established WHO grade. Reports suggest a better outcome than high grade gliomas, though they frequently recur. Recently, they have been described to overlap with a newly-discovered group of tumours described as'high grade neuroepithelial tumour with MN1 alteration' (CNS HGNET-MN1), defined by global methylation patterns and strongly associated with gene fusions targeting MN1. We have studied a unique case of astroblastoma arising in a 6 year-old girl, with multiple recurrences over a period of 10 years, with the pathognomonic MN1:BEND2 fusion. Exome sequencing allowed for a phylogenetic reconstruction of tumour evolution, which when integrated with clinical, pathological and radiological data provide for a detailed understanding of disease progression, with initial treatment driving tumour dissemination along four distinct trajectories. Infiltration of distant sites was associated with a later genome doubling, whilst there was evidence of convergent evolution of different lesions acquiring distinct alterations targeting NF-κB. These data represent an unusual opportunity to understand the evolutionary history of a highly recurrent childhood brain tumour, and provide novel therapeutic targets for astroblastoma/CNS HGNET-MN1..
Xavier-Magalhães, A.
Gonçalves, C.S.
Fogli, A.
Lourenço, T.
Pojo, M.
Pereira, B.
Rocha, M.
Lopes, M.C.
Crespo, I.
Rebelo, O.
Tão, H.
Lima, J.
Moreira, R.
Pinto, A.A.
Jones, C.
Reis, R.M.
Costello, J.F.
Arnaud, P.
Sousa, N.
Costa, B.M.
(2018). The long non-coding RNA HOTAIR is transcriptionally activated by HOXA9 and is an independent prognostic marker in patients with malignant glioma. Oncotarget,
Vol.9
(21),
pp. 15740-15756.
show abstract
full text
The lncRNA HOTAIR has been implicated in several human cancers. Here, we evaluated the molecular alterations and upstream regulatory mechanisms of HOTAIR in glioma, the most common primary brain tumors, and its clinical relevance. HOTAIR gene expression, methylation, copy-number and prognostic value were investigated in human gliomas integrating data from online datasets and our cohorts. High levels of HOTAIR were associated with higher grades of glioma, particularly IDH wild-type cases. Mechanistically, HOTAIR was overexpressed in a gene dosage-independent manner, while DNA methylation levels of particular CpGs in HOTAIR locus were associated with HOTAIR expression levels in GBM clinical specimens and cell lines. Concordantly, the demethylating agent 5-Aza-2'-deoxycytidine affected HOTAIR transcriptional levels in a cell line-dependent manner. Importantly, HOTAIR was frequently co-expressed with HOXA9 in high-grade gliomas from TCGA, Oncomine, and our Portuguese and French datasets. Integrated in silico analyses, chromatin immunoprecipitation, and qPCR data showed that HOXA9 binds directly to the promoter of HOTAIR. Clinically, GBM patients with high HOTAIR expression had a significantly reduced overall survival, independently of other prognostic variables. In summary, this work reveals HOXA9 as a novel direct regulator of HOTAIR, and establishes HOTAIR as an independent prognostic marker, providing new therapeutic opportunities to treat this highly aggressive cancer..
Capper, D.
Jones, D.T.
Sill, M.
Hovestadt, V.
Schrimpf, D.
Sturm, D.
Koelsche, C.
Sahm, F.
Chavez, L.
Reuss, D.E.
Kratz, A.
Wefers, A.K.
Huang, K.
Pajtler, K.W.
Schweizer, L.
Stichel, D.
Olar, A.
Engel, N.W.
Lindenberg, K.
Harter, P.N.
Braczynski, A.K.
Plate, K.H.
Dohmen, H.
Garvalov, B.K.
Coras, R.
Hölsken, A.
Hewer, E.
Bewerunge-Hudler, M.
Schick, M.
Fischer, R.
Beschorner, R.
Schittenhelm, J.
Staszewski, O.
Wani, K.
Varlet, P.
Pages, M.
Temming, P.
Lohmann, D.
Selt, F.
Witt, H.
Milde, T.
Witt, O.
Aronica, E.
Giangaspero, F.
Rushing, E.
Scheurlen, W.
Geisenberger, C.
Rodriguez, F.J.
Becker, A.
Preusser, M.
Haberler, C.
Bjerkvig, R.
Cryan, J.
Farrell, M.
Deckert, M.
Hench, J.
Frank, S.
Serrano, J.
Kannan, K.
Tsirigos, A.
Brück, W.
Hofer, S.
Brehmer, S.
Seiz-Rosenhagen, M.
Hänggi, D.
Hans, V.
Rozsnoki, S.
Hansford, J.R.
Kohlhof, P.
Kristensen, B.W.
Lechner, M.
Lopes, B.
Mawrin, C.
Ketter, R.
Kulozik, A.
Khatib, Z.
Heppner, F.
Koch, A.
Jouvet, A.
Keohane, C.
Mühleisen, H.
Mueller, W.
Pohl, U.
Prinz, M.
Benner, A.
Zapatka, M.
Gottardo, N.G.
Driever, P.H.
Kramm, C.M.
Müller, H.L.
Rutkowski, S.
von Hoff, K.
Frühwald, M.C.
Gnekow, A.
Fleischhack, G.
Tippelt, S.
Calaminus, G.
Monoranu, C.-.
Perry, A.
Jones, C.
Jacques, T.S.
Radlwimmer, B.
Gessi, M.
Pietsch, T.
Schramm, J.
Schackert, G.
Westphal, M.
Reifenberger, G.
Wesseling, P.
Weller, M.
Collins, V.P.
Blümcke, I.
Bendszus, M.
Debus, J.
Huang, A.
Jabado, N.
Northcott, P.A.
Paulus, W.
Gajjar, A.
Robinson, G.W.
Taylor, M.D.
Jaunmuktane, Z.
Ryzhova, M.
Platten, M.
Unterberg, A.
Wick, W.
Karajannis, M.A.
Mittelbronn, M.
Acker, T.
Hartmann, C.
Aldape, K.
Schüller, U.
Buslei, R.
Lichter, P.
Kool, M.
Herold-Mende, C.
Ellison, D.W.
Hasselblatt, M.
Snuderl, M.
Brandner, S.
Korshunov, A.
von Deimling, A.
Pfister, S.M.
(2018). DNA methylation-based classification of central nervous system tumours. Nature,
Vol.555
(7697),
pp. 469-474.
show abstract
full text
Accurate pathological diagnosis is crucial for optimal management of patients with cancer. For the approximately 100 known tumour types of the central nervous system, standardization of the diagnostic process has been shown to be particularly challenging-with substantial inter-observer variability in the histopathological diagnosis of many tumour types. Here we present a comprehensive approach for the DNA methylation-based classification of central nervous system tumours across all entities and age groups, and demonstrate its application in a routine diagnostic setting. We show that the availability of this method may have a substantial impact on diagnostic precision compared to standard methods, resulting in a change of diagnosis in up to 12% of prospective cases. For broader accessibility, we have designed a free online classifier tool, the use of which does not require any additional onsite data processing. Our results provide a blueprint for the generation of machine-learning-based tumour classifiers across other cancer entities, with the potential to fundamentally transform tumour pathology..
Hudson, L.
Mui, J.
Vázquez, S.
Carvalho, D.M.
Williams, E.
Jones, C.
Bullock, A.N.
Hoelder, S.
(2018). Novel Quinazolinone Inhibitors of ALK2 Flip between Alternate Binding Modes: Structure-Activity Relationship, Structural Characterization, Kinase Profiling, and Cellular Proof of Concept. J med chem,
Vol.61
(16),
pp. 7261-7272.
show abstract
full text
Structure-activity relationship and crystallographic data revealed that quinazolinone-containing fragments flip between two distinct modes of binding to activin receptor-like kinase-2 (ALK2). We explored both binding modes to discover potent inhibitors and characterized the chemical modifications that triggered the flip in binding mode. We report kinase selectivity and demonstrate that compounds of this series modulate ALK2 in cancer cells. These inhibitors are attractive starting points for the discovery of more advanced ALK2 inhibitors..
Jones, C.
Karajannis, M.A.
Jones, D.T.
Kieran, M.W.
Monje, M.
Baker, S.J.
Becher, O.J.
Cho, Y.-.
Gupta, N.
Hawkins, C.
Hargrave, D.
Haas-Kogan, D.A.
Jabado, N.
Li, X.-.
Mueller, S.
Nicolaides, T.
Packer, R.J.
Persson, A.I.
Phillips, J.J.
Simonds, E.F.
Stafford, J.M.
Tang, Y.
Pfister, S.M.
Weiss, W.A.
(2017). Pediatric high-grade glioma: biologically and clinically in need of new thinking. Neuro oncol,
Vol.19
(2),
pp. 153-161.
show abstract
full text
High-grade gliomas in children are different from those that arise in adults. Recent collaborative molecular analyses of these rare cancers have revealed previously unappreciated connections among chromatin regulation, developmental signaling, and tumorigenesis. As we begin to unravel the unique developmental origins and distinct biological drivers of this heterogeneous group of tumors, clinical trials need to keep pace. It is important to avoid therapeutic strategies developed purely using data obtained from studies on adult glioblastoma. This approach has resulted in repetitive trials and ineffective treatments being applied to these children, with limited improvement in clinical outcome. The authors of this perspective, comprising biology and clinical expertise in the disease, recently convened to discuss the most effective ways to translate the emerging molecular insights into patient benefit. This article reviews our current understanding of pediatric high-grade glioma and suggests approaches for innovative clinical management..
Baugh, J.
Bartels, U.
Leach, J.
Jones, B.
Chaney, B.
Warren, K.E.
Kirkendall, J.
Doughman, R.
Hawkins, C.
Miles, L.
Fuller, C.
Hassall, T.
Bouffet, E.
Lane, A.
Hargrave, D.
Grill, J.
Hoffman, L.M.
Jones, C.
Towbin, A.
Savage, S.A.
Monje, M.
Li, X.-.
Ziegler, D.S.
Veldhuijzen van Zanten, S.
Kramm, C.M.
van Vuurden, D.G.
Fouladi, M.
(2017). The international diffuse intrinsic pontine glioma registry: an infrastructure to accelerate collaborative research for an orphan disease. J neurooncol,
Vol.132
(2),
pp. 323-331.
show abstract
full text
Diffuse intrinsic pontine glioma (DIPG), a rare, often fatal childhood brain tumor, remains a major therapeutic challenge. In 2012, investigators, funded by the DIPG Collaborative (a philanthropic partnership among 29 private foundations), launched the International DIPG Registry (IDIPGR) to advance understanding of DIPG. Comprised of comprehensive deidentified but linked clinical, imaging, histopathological, and genomic repositories, the IDIPGR uses standardized case report forms for uniform data collection; serial imaging and histopathology are centrally reviewed by IDIPGR neuro-radiologists and neuro-pathologists, respectively. Tissue and genomic data, and cell cultures derived from autopsies coordinated by the IDIPGR are available to investigators for studies approved by the Scientific Advisory Committee. From April 2012 to December 2016, 670 patients diagnosed with DIPG have been enrolled from 55 participating institutions in the US, Canada, Australia and New Zealand. The radiology repository contains 3558 studies from 448 patients. The pathology repository contains tissue on 81 patients with another 98 samples available for submission. Fresh DIPG tissue from seven autopsies has been sent to investigators to develop primary cell cultures. The bioinformatics repository contains next-generation sequencing data on 66 tumors. Nine projects using data/tissue from the IDIPGR by 13 principle investigators from around the world are now underway. The IDIPGR, a successful alliance among philanthropic agencies and investigators, has developed and maintained a highly collaborative, hypothesis-driven research infrastructure for interdisciplinary and translational projects in DIPG to improve diagnosis, response assessment, treatment and outcome for patients..
Veldhuijzen van Zanten, S.E.
Baugh, J.
Chaney, B.
De Jongh, D.
Sanchez Aliaga, E.
Barkhof, F.
Noltes, J.
De Wolf, R.
Van Dijk, J.
Cannarozzo, A.
Damen-Korbijn, C.M.
Lieverst, J.A.
Colditz, N.
Hoffmann, M.
Warmuth-Metz, M.
Bison, B.
Jones, D.T.
Sturm, D.
Gielen, G.H.
Jones, C.
Hulleman, E.
Calmon, R.
Castel, D.
Varlet, P.
Giraud, G.
Slavc, I.
Van Gool, S.
Jacobs, S.
Jadrijevic-Cvrlje, F.
Sumerauer, D.
Nysom, K.
Pentikainen, V.
Kivivuori, S.-.
Leblond, P.
Entz-Werle, N.
von Bueren, A.O.
Kattamis, A.
Hargrave, D.R.
Hauser, P.
Garami, M.
Thorarinsdottir, H.K.
Pears, J.
Gandola, L.
Rutkauskiene, G.
Janssens, G.O.
Torsvik, I.K.
Perek-Polnik, M.
Gil-da-Costa, M.J.
Zheludkova, O.
Shats, L.
Deak, L.
Kitanovski, L.
Cruz, O.
Morales La Madrid, A.
Holm, S.
Gerber, N.
Kebudi, R.
Grundy, R.
Lopez-Aguilar, E.
Zapata-Tarres, M.
Emmerik, J.
Hayden, T.
Bailey, S.
Biassoni, V.
Massimino, M.
Grill, J.
Vandertop, W.P.
Kaspers, G.J.
Fouladi, M.
Kramm, C.M.
van Vuurden, D.G.
members of the SIOPE DIPG Network,
(2017). Development of the SIOPE DIPG network, registry and imaging repository: a collaborative effort to optimize research into a rare and lethal disease. J neurooncol,
Vol.132
(2),
pp. 255-266.
show abstract
full text
Diffuse intrinsic pontine glioma (DIPG) is a rare and deadly childhood malignancy. After 40 years of mostly single-center, often non-randomized trials with variable patient inclusions, there has been no improvement in survival. It is therefore time for international collaboration in DIPG research, to provide new hope for children, parents and medical professionals fighting DIPG. In a first step towards collaboration, in 2011, a network of biologists and clinicians working in the field of DIPG was established within the European Society for Paediatric Oncology (SIOPE) Brain Tumour Group: the SIOPE DIPG Network. By bringing together biomedical professionals and parents as patient representatives, several collaborative DIPG-related projects have been realized. With help from experts in the fields of information technology, and legal advisors, an international, web-based comprehensive database was developed, The SIOPE DIPG Registry and Imaging Repository, to centrally collect data of DIPG patients. As for April 2016, clinical data as well as MR-scans of 694 patients have been entered into the SIOPE DIPG Registry/Imaging Repository. The median progression free survival is 6.0 months (95% Confidence Interval (CI) 5.6-6.4 months) and the median overall survival is 11.0 months (95% CI 10.5-11.5 months). At two and five years post-diagnosis, 10 and 2% of patients are alive, respectively. The establishment of the SIOPE DIPG Network and SIOPE DIPG Registry means a paradigm shift towards collaborative research into DIPG. This is seen as an essential first step towards understanding the disease, improving care and (ultimately) cure for children with DIPG..
Trabelsi, S.
Chabchoub, I.
Ksira, I.
Karmeni, N.
Mama, N.
Kanoun, S.
Burford, A.
Jury, A.
Mackay, A.
Popov, S.
Bouaouina, N.
Ben Ahmed, S.
Mokni, M.
Tlili, K.
Krifa, H.
Yacoubi, M.T.
Jones, C.
Saad, A.
H'mida Ben Brahim, D.
(2017). Molecular Diagnostic and Prognostic Subtyping of Gliomas in Tunisian Population. Mol neurobiol,
Vol.54
(4),
pp. 2381-2394.
show abstract
It has become increasingly evident that morphologically similar gliomas may have distinct clinical phenotypes arising from diverse genetic signatures. To date, glial tumours from the Tunisian population have not been investigated. To address this, we correlated the clinico-pathology with molecular data of 110 gliomas by a combination of HM450K array, MLPA and TMA-IHC. PTEN loss and EGFR amplification were distributed in different glioma histological groups. However, 1p19q co-deletion and KIAA1549:BRAF fusion were, respectively, restricted to Oligodendroglioma and Pilocytic Astrocytoma. CDKN2A loss and EGFR overexpression were more common within high-grade gliomas. Furthermore, survival statistical correlations led us to identify Glioblastoma (GB) prognosis subtypes. In fact, significant lower overall survival (OS) was detected within GB that overexpressed EGFR and Cox2. In addition, IDH1R132H mutation seemed to provide a markedly survival advantage. Interestingly, the association of IDHR132H mutation and EGFR normal status, as well as the association of differentiation markers, defined GB subtypes with good prognosis. By contrast, poor survival GB subtypes were defined by the combination of PTEN loss with PDGFRa expression and/or EGFR amplification. Additionally, GB presenting p53-negative staining associated with CDKN2A loss or p21 positivity represented a subtype with short survival. Thus, distinct molecular subtypes with individualised prognosis were identified. Interestingly, we found a unique histone mutation in a poor survival young adult GB case. This tumour exceptionally associated the H3F3A G34R mutation and MYCN amplification as well as 1p36 loss and 10q loss. Furthermore, by exhibiting a remarkable methylation profile, it emphasised the oncogenic power of G34R mutation connecting gliomagenesis and chromatin regulation..
Haque, F.
Varlet, P.
Puntonet, J.
Storer, L.
Bountali, A.
Rahman, R.
Grill, J.
Carcaboso, A.M.
Jones, C.
Layfield, R.
Grundy, R.G.
(2017). Evaluation of a novel antibody to define histone 3 3 G34R mutant brain tumours. Acta neuropathol commun,
Vol.5
(1),
p. 45.
show abstract
full text
Missense somatic mutations affecting histone H3.1 and H3.3 proteins are now accepted as the hallmark of paediatric diffuse intrinsic pontine gliomas (DIPG), non-brain stem paediatric high grade gliomas (pHGG) as well as a subset of adult glioblastoma multiforme (GBM). Different mutations give rise to one of three amino acid substitutions at two critical positions within the histone tails, K27M, G34R/V. Several studies have highlighted gene expression and epigenetic changes associated with histone H3 mutations; however their precise roles in tumourigenesis remain incompletely understood. Determining how such amino acid substitutions in a protein affect its properties can be challenging because of difficulties in detecting and tracking mutant proteins within cells and tissues. Here we describe a strategy for the generation of antibodies to discriminate G34R and G34V mutant histone H3 proteins from their wild-type counterparts. Antibodies were validated by western blotting and immunocytochemistry, using recombinant H3.3 proteins and paediatric GBM cell lines. The H3-G34R antibody demonstrated a high degree of selectivity towards its target sequence. Accordingly, immunostaining on a cohort of 22 formalin-fixed paraffin embedded tumours with a previously known H3.3 G34R mutation status, detected successfully the corresponding mutant protein in 11/11 G34R cases. Since there was a high concordance between genotype and immunohistochemical analysis of G34R mutant tumour samples, we analysed a series of tissue microarrays (TMAs) to assess the specificity of the antibody in a range of paediatric brain tumours, and noted immunoreactivity in 2/634 cases. Importantly, we describe the generation and validation of highly specific antibodies for G34 mutations. Overall our work adds to an extremely valuable portfolio of antibodies, not only for histopathologic detection of tumour-associated mutant histone sequences, but also facilitating the study of spatial/anatomical aspects of tumour formation and the identification of downstream targets and pathways in malignant glioma progression..
Mackay, A.
Burford, A.
Carvalho, D.
Izquierdo, E.
Fazal-Salom, J.
Taylor, K.R.
Bjerke, L.
Clarke, M.
Vinci, M.
Nandhabalan, M.
Temelso, S.
Popov, S.
Molinari, V.
Raman, P.
Waanders, A.J.
Han, H.J.
Gupta, S.
Marshall, L.
Zacharoulis, S.
Vaidya, S.
Mandeville, H.C.
Bridges, L.R.
Martin, A.J.
Al-Sarraj, S.
Chandler, C.
Ng, H.-.
Li, X.
Mu, K.
Trabelsi, S.
Brahim, D.H.
Kisljakov, A.N.
Konovalov, D.M.
Moore, A.S.
Carcaboso, A.M.
Sunol, M.
de Torres, C.
Cruz, O.
Mora, J.
Shats, L.I.
Stavale, J.N.
Bidinotto, L.T.
Reis, R.M.
Entz-Werle, N.
Farrell, M.
Cryan, J.
Crimmins, D.
Caird, J.
Pears, J.
Monje, M.
Debily, M.-.
Castel, D.
Grill, J.
Hawkins, C.
Nikbakht, H.
Jabado, N.
Baker, S.J.
Pfister, S.M.
Jones, D.T.
Fouladi, M.
von Bueren, A.O.
Baudis, M.
Resnick, A.
Jones, C.
(2017). Integrated Molecular Meta-Analysis of 1,000 Pediatric High-Grade and Diffuse Intrinsic Pontine Glioma. Cancer cell,
Vol.32
(4),
pp. 520-537.e5.
show abstract
full text
We collated data from 157 unpublished cases of pediatric high-grade glioma and diffuse intrinsic pontine glioma and 20 publicly available datasets in an integrated analysis of >1,000 cases. We identified co-segregating mutations in histone-mutant subgroups including loss of FBXW7 in H3.3G34R/V, TOP3A rearrangements in H3.3K27M, and BCOR mutations in H3.1K27M. Histone wild-type subgroups are refined by the presence of key oncogenic events or methylation profiles more closely resembling lower-grade tumors. Genomic aberrations increase with age, highlighting the infant population as biologically and clinically distinct. Uncommon pathway dysregulation is seen in small subsets of tumors, further defining the molecular diversity of the disease, opening up avenues for biological study and providing a basis for functionally defined future treatment stratification..
Boult, J.K.
Box, G.
Vinci, M.
Perryman, L.
Eccles, S.A.
Jones, C.
Robinson, S.P.
(2017). Evaluation of the Response of Intracranial Xenografts to VEGF Signaling Inhibition Using Multiparametric MRI. Neoplasia,
Vol.19
(9),
pp. 684-694.
show abstract
full text
Vascular endothelial growth factor A (VEGF-A) is considered one of the most important factors in tumor angiogenesis, and consequently, a number of therapeutics have been developed to inhibit VEGF signaling. Therapeutic strategies to target brain malignancies, both primary brain tumors, particularly in pediatric patients, and metastases, are lacking, but targeting angiogenesis may be a promising approach. Multiparametric MRI was used to investigate the response of orthotopic SF188luc pediatric glioblastoma xenografts to small molecule pan-VEGFR inhibitor cediranib and the effects of both cediranib and cross-reactive human/mouse anti-VEGF-A antibody B20-4.1.1 in intracranial MDA-MB-231 LM2-4 breast cancer xenografts over 48 hours. All therapeutic regimens resulted in significant tumor growth delay. In cediranib-treated SF188luc tumors, this was associated with lower Ktrans (compound biomarker of perfusion and vascular permeability) than in vehicle-treated controls. Cediranib also induced significant reductions in both Ktrans and apparent diffusion coefficient (ADC) in MDA-MB-231 LM2-4 tumors associated with decreased histologically assessed perfusion. B20-4.1.1 treatment resulted in decreased Ktrans, but in the absence of a change in perfusion; a non-significant reduction in vascular permeability, assessed by Evans blue extravasation, was observed in treated tumors. The imaging responses of intracranial MDA-MB-231 LM2-4 tumors to VEGF/VEGFR pathway inhibitors with differing mechanisms of action are subtly different. We show that VEGF pathway blockade resulted in tumor growth retardation and inhibition of tumor vasculature in preclinical models of pediatric glioblastoma and breast cancer brain metastases, suggesting that multiparametric MRI can provide a powerful adjunct to accelerate the development of antiangiogenic therapies for use in these patient populations..
Korshunov, A.
Schrimpf, D.
Ryzhova, M.
Sturm, D.
Chavez, L.
Hovestadt, V.
Sharma, T.
Habel, A.
Burford, A.
Jones, C.
Zheludkova, O.
Kumirova, E.
Kramm, C.M.
Golanov, A.
Capper, D.
von Deimling, A.
Pfister, S.M.
Jones, D.T.
(2017). H3-/IDH-wild type pediatric glioblastoma is comprised of molecularly and prognostically distinct subtypes with associated oncogenic drivers. Acta neuropathol,
Vol.134
(3),
pp. 507-516.
show abstract
Pediatric glioblastoma (pedGBM) is an extremely aggressive pediatric brain tumor, accounting for ~6% of all central nervous system neoplasms in children. Approximately half of pedGBM harbor recurrent somatic mutations in histone 3 variants or, infrequently, IDH1/2. The remaining subset of pedGBM is highly heterogeneous, and displays a variety of genomic and epigenetic features. In the current study, we aimed to further stratify an H3-/IDH-wild type (wt) pedGBM cohort assessed through genome-wide molecular profiling. As a result, we identified three molecular subtypes of these tumors, differing in their genomic and epigenetic signatures as well as in their clinical behavior. We designated these subtypes 'pedGBM_MYCN' (enriched for MYCN amplification), 'pedGBM_RTK1' (enriched for PDGFRA amplification) and 'pedGBM_RTK2' (enriched for EGFR amplification). These molecular subtypes were associated with significantly different outcomes, i.e. pedGBM_RTK2 tumors show a significantly longer survival time (median OS 44 months), pedGBM_MYCN display extremely poor outcomes (median OS 14 months), and pedGBM_RTK1 tumors harbor an intermediate prognosis. In addition, the various molecular subtypes of H3-/IDH-wt pedGBM were clearly distinguishable from their adult counterparts, underlining their biological distinctiveness. In conclusion, our study demonstrates significant molecular heterogeneity of H3-/IDH-wt pedGBM in terms of DNA methylation and cytogenetic alterations. The recognition of three molecular subtypes of H3-/IDH-wt pedGBM further revealed close correlations with biological parameters and clinical outcomes and may therefore, be predictive of response to standard treatment protocols, but could also be useful for stratification for novel, molecularly based therapies..
Amodeo, V.
A, D.
Betts, J.
Bartesaghi, S.
Zhang, Y.
Richard-Londt, A.
Ellis, M.
Roshani, R.
Vouri, M.
Galavotti, S.
Oberndorfer, S.
Leite, A.P.
Mackay, A.
Lampada, A.
Stratford, E.W.
Li, N.
Dinsdale, D.
Grimwade, D.
Jones, C.
Nicotera, P.
Michod, D.
Brandner, S.
Salomoni, P.
(2017). A PML/Slit Axis Controls Physiological Cell Migration and Cancer Invasion in the CNS. Cell rep,
Vol.20
(2),
pp. 411-426.
show abstract
Cell migration through the brain parenchyma underpins neurogenesis and glioblastoma (GBM) development. Since GBM cells and neuroblasts use the same migratory routes, mechanisms underlying migration during neurogenesis and brain cancer pathogenesis may be similar. Here, we identify a common pathway controlling cell migration in normal and neoplastic cells in the CNS. The nuclear scaffold protein promyelocytic leukemia (PML), a regulator of forebrain development, promotes neural progenitor/stem cell (NPC) and neuroblast migration in the adult mouse brain. The PML pro-migratory role is active also in transformed mouse NPCs and in human primary GBM cells. In both normal and neoplastic settings, PML controls cell migration via Polycomb repressive complex 2 (PRC2)-mediated repression of Slits, key regulators of axon guidance. Finally, a PML/SLIT1 axis regulates sensitivity to the PML-targeting drug arsenic trioxide in primary GBM cells. Taken together, these findings uncover a drug-targetable molecular axis controlling cell migration in both normal and neoplastic cells..
Agliano, A.
Balarajah, G.
Ciobota, D.M.
Sidhu, J.
Clarke, P.A.
Jones, C.
Workman, P.
Leach, M.O.
Al-Saffar, N.M.
(2017). Pediatric and adult glioblastoma radiosensitization induced by PI3K/mTOR inhibition causes early metabolic alterations detected by nuclear magnetic resonance spectroscopy. Oncotarget,
Vol.8
(29),
pp. 47969-47983.
show abstract
full text
Poor outcome for patients with glioblastomas is often associated with radioresistance. PI3K/mTOR pathway deregulation has been correlated with radioresistance; therefore, PI3K/mTOR inhibition could render tumors radiosensitive. In this study, we show that NVP-BEZ235, a dual PI3K/mTOR inhibitor, potentiates the effects of irradiation in both adult and pediatric glioblastoma cell lines, resulting in early metabolic changes detected by nuclear magnetic resonance (NMR) spectroscopy. NVP-BEZ235 radiosensitises cells to X ray exposure, inducing cell death through the inhibition of CDC25A and the activation of p21cip1(CDKN1A). Lactate and phosphocholine levels, increased with radiation, are decreased after NVP-BEZ235 and combination treatment, suggesting that inhibiting the PI3K/mTOR pathway reverses radiation induced metabolic changes. Importantly, NVP-BEZ235 potentiates the effects of irradiation in a xenograft model of adult glioblastoma, where we observed a decrease in lactate and phosphocholine levels after seven days of combination treatment. Although tumor size was not affected due to the short length of the treatment, a significant increase in CASP3 mRNA was observed in the combination group. Taken together, our data suggest that NMR metabolites could be used as biomarkers to detect an early response to combination therapy with PI3K/mTOR inhibitors and radiotherapy in adult and pediatric glioblastoma patients..
Izquierdo, E.
Yuan, L.
George, S.
Hubank, M.
Jones, C.
Proszek, P.
Shipley, J.
Gatz, S.A.
Stinson, C.
Moore, A.S.
Clifford, S.C.
Hicks, D.
Lindsey, J.C.
Hill, R.M.
Jacques, T.S.
Chalker, J.
Thway, K.
O'Connor, S.
Marshall, L.
Moreno, L.
Pearson, A.
Chesler, L.
Walker, B.A.
De Castro, D.G.
(2017). Development of a targeted sequencing approach to identify prognostic, predictive and diagnostic markers in paediatric solid tumours. Oncotarget,
Vol.8
(67),
pp. 112036-112050.
show abstract
full text
The implementation of personalised medicine in childhood cancers has been limited by a lack of clinically validated multi-target sequencing approaches specific for paediatric solid tumours. In order to support innovative clinical trials in high-risk patients with unmet need, we have developed a clinically relevant targeted sequencing panel spanning 311 kb and comprising 78 genes involved in childhood cancers. A total of 132 samples were used for the validation of the panel, including Horizon Discovery cell blends (n=4), cell lines (n=15), formalin-fixed paraffin embedded (FFPE, n=83) and fresh frozen tissue (FF, n=30) patient samples. Cell blends containing known single nucleotide variants (SNVs, n=528) and small insertion-deletions (indels n=108) were used to define panel sensitivities of ≥98% for SNVs and ≥83% for indels [95% CI] and panel specificity of ≥98% [95% CI] for SNVs. FFPE samples performed comparably to FF samples (n=15 paired). Of 95 well-characterised genetic abnormalities in 33 clinical specimens and 13 cell lines (including SNVs, indels, amplifications, rearrangements and chromosome losses), 94 (98.9%) were detected by our approach. We have validated a robust and practical methodology to guide clinical management of children with solid tumours based on their molecular profiles. Our work demonstrates the value of targeted gene sequencing in the development of precision medicine strategies in paediatric oncology..
Nguyen, A.
Moussallieh, F.M.
Mackay, A.
Cicek, A.E.
Coca, A.
Pierre Chenard, M.
Weingertner, N.
Lhermitte, B.
Letouzé, E.
Guérin, E.
Pencreach, E.
Jannier, S.
Guenot, D.
Jacques Namer, I.
Jones, C.
Entz-Werlé, N.
(2017). Characterization of the transcriptional and metabolic responses of pediatric high grade gliomas to mTOR-HIF-1a axis inhibition. Oncotarget,
.
show abstract
full text
Pediatric high grade glioma (pHGGs), including sus-tentorial and diffuse intrinsic pontine gliomas, are known to have a very dismal prognosis. For instance, even an increased knowledge on molecular biology driving this brain tumor entity, there is no treatment able to cure those patients. Therefore, we were focusing on a translational pathway able to increase the cell resistance to treatment and to reprogram metabolically tumor cells, which are, then, adapting easily to a hypoxic microenvironment. To establish, the crucial role of the hypoxic pathways in pHGGs, we, first, assessed their protein and transcriptomic deregulations in a pediatric cohort of pHGGs and in pHGG's cell lines, cultured in both normoxic and hypoxic conditions. Secondly, based on the concept of a bi-therapy targeting in pHGGs mTORC1 (rapamycin) and HIF-1α (irinotecan), we hypothesized that the balanced expressions between RAS/ERK, PI3K/AKT and HIF-1α/HIF-2α/MYC proteins or genes may provide a modulation of the cell response to this double targeting. Finally, we could evidence three protein, genomic and metabolomic profiles of response to rapamycin combined with irinotecan. The pattern of highly sensitive cells to mTOR/HIF-1α targeting was linked to a MYC/ERK/HIF-1α over-expression and the cell resistance to a major hyper-expression of HIF-2α..
Qin, E.Y.
Cooper, D.D.
Abbott, K.L.
Lennon, J.
Nagaraja, S.
Mackay, A.
Jones, C.
Vogel, H.
Jackson, P.K.
Monje, M.
(2017). Neural Precursor-Derived Pleiotrophin Mediates Subventricular Zone Invasion by Glioma. Cell,
Vol.170
(5),
pp. 845-859.e19.
show abstract
full text
The lateral ventricle subventricular zone (SVZ) is a frequent and consequential site of pediatric and adult glioma spread, but the cellular and molecular mechanisms mediating this are poorly understood. We demonstrate that neural precursor cell (NPC):glioma cell communication underpins this propensity of glioma to colonize the SVZ through secretion of chemoattractant signals toward which glioma cells home. Biochemical, proteomic, and functional analyses of SVZ NPC-secreted factors revealed the neurite outgrowth-promoting factor pleiotrophin, along with required binding partners SPARC/SPARCL1 and HSP90B, as key mediators of this chemoattractant effect. Pleiotrophin expression is strongly enriched in the SVZ, and pleiotrophin knock down starkly reduced glioma invasion of the SVZ in the murine brain. Pleiotrophin, in complex with the binding partners, activated glioma Rho/ROCK signaling, and ROCK inhibition decreased invasion toward SVZ NPC-secreted factors. These findings demonstrate a pathogenic role for NPC:glioma interactions and potential therapeutic targets to limit glioma invasion. PAPERCLIP..
Al-Saffar, N.M.
Agliano, A.
Marshall, L.V.
Jackson, L.E.
Balarajah, G.
Sidhu, J.
Clarke, P.A.
Jones, C.
Workman, P.
Pearson, A.D.
Leach, M.O.
(2017). In vitro nuclear magnetic resonance spectroscopy metabolic biomarkers for the combination of temozolomide with PI3K inhibition in paediatric glioblastoma cells. Plos one,
Vol.12
(7),
p. e0180263.
show abstract
full text
Recent experimental data showed that the PI3K pathway contributes to resistance to temozolomide (TMZ) in paediatric glioblastoma and that this effect is reversed by combination treatment of TMZ with a PI3K inhibitor. Our aim is to assess whether this combination results in metabolic changes that are detectable by nuclear magnetic resonance (NMR) spectroscopy, potentially providing metabolic biomarkers for PI3K inhibition and TMZ combination treatment. Using two genetically distinct paediatric glioblastoma cell lines, SF188 and KNS42, in vitro 1H-NMR analysis following treatment with the dual pan-Class I PI3K/mTOR inhibitor PI-103 resulted in a decrease in lactate and phosphocholine (PC) levels (P<0.02) relative to control. In contrast, treatment with TMZ caused an increase in glycerolphosphocholine (GPC) levels (P≤0.05). Combination of PI-103 with TMZ showed metabolic effects of both agents including a decrease in the levels of lactate and PC (P<0.02) while an increase in GPC (P<0.05). We also report a decrease in the protein expression levels of HK2, LDHA and CHKA providing likely mechanisms for the depletion of lactate and PC, respectively. Our results show that our in vitro NMR-detected changes in lactate and choline metabolites may have potential as non-invasive biomarkers for monitoring response to combination of PI3K/mTOR inhibitors with TMZ during clinical trials in children with glioblastoma, subject to further in vivo validation..
Korshunov, A.
Capper, D.
Reuss, D.
Schrimpf, D.
Ryzhova, M.
Hovestadt, V.
Sturm, D.
Meyer, J.
Jones, C.
Zheludkova, O.
Kumirova, E.
Golanov, A.
Kool, M.
Schüller, U.
Mittelbronn, M.
Hasselblatt, M.
Schittenhelm, J.
Reifenberger, G.
Herold-Mende, C.
Lichter, P.
von Deimling, A.
Pfister, S.M.
Jones, D.T.
(2016). Histologically distinct neuroepithelial tumors with histone 3 G34 mutation are molecularly similar and comprise a single nosologic entity. Acta neuropathol,
Vol.131
(1),
pp. 137-146.
show abstract
In contrast to the relative morphological uniformity of histone H3 K27-mutant high-grade gliomas, H3 G34-mutant tumors present as a histopathologically heterogeneous group of neoplasms, with microscopic characteristics typical of either glioblastoma (GBM) or central nervous system primitive neuroectodermal tumors (CNS-PNET). In the current study, we performed an integrative clinical, histopathological and molecular analysis of 81 G34-mutant CNS tumors. Routinely prepared tumor tissues were investigated for genomic and epigenomic alterations. Despite their divergent histopathological appearance, CNS tumors with H3.3 G34 mutations displayed uniform epigenetic signatures, suggesting a single biological origin. Comparative cytogenetic analysis with other GBM subtypes disclosed a high frequency and high specificity of 3q and 4q loss across G34-mutant tumors. PDGFRA amplification was more common in cases with GBM than with PNET morphology (36 vs. 5 %, respectively), while CCND2 amplifications showed the opposite trend (5 vs. 27 %). Survival analysis revealed the presence of amplified oncogene(s) and MGMT methylation as independent prognostic markers for poor and favorable outcomes, respectively. No difference in outcome was found between morphological variants (GBM vs. PNET). Thus, different histological variants of G34-mutant CNS tumors likely comprise a single biological entity (high-grade glioma with H3 G34 mutation, HGG_G34), which should be outlined in future diagnostic and therapeutic classifications. Screening for H3.3 G34 mutation should therefore be recommended as a routine diagnostic marker for supratentorial CNS tumors across a broad histological spectrum..
Koschmann, C.
Calinescu, A.-.
Nunez, F.J.
Mackay, A.
Fazal-Salom, J.
Thomas, D.
Mendez, F.
Kamran, N.
Dzaman, M.
Mulpuri, L.
Krasinkiewicz, J.
Doherty, R.
Lemons, R.
Brosnan-Cashman, J.A.
Li, Y.
Roh, S.
Zhao, L.
Appelman, H.
Ferguson, D.
Gorbunova, V.
Meeker, A.
Jones, C.
Lowenstein, P.R.
Castro, M.G.
(2016). ATRX loss promotes tumor growth and impairs nonhomologous end joining DNA repair in glioma. Sci transl med,
Vol.8
(328),
p. 328ra28.
show abstract
Recent work in human glioblastoma (GBM) has documented recurrent mutations in the histone chaperone protein ATRX. We developed an animal model of ATRX-deficient GBM and showed that loss of ATRX reduces median survival and increases genetic instability. Further, analysis of genome-wide data for human gliomas showed that ATRX mutation is associated with increased mutation rate at the single-nucleotide variant (SNV) level. In mouse tumors, ATRX deficiency impairs nonhomologous end joining and increases sensitivity to DNA-damaging agents that induce double-stranded DNA breaks. We propose that ATRX loss results in a genetically unstable tumor, which is more aggressive when left untreated but is more responsive to double-stranded DNA-damaging agents, resulting in improved overall survival..
Koschmann, C.
Zamler, D.
MacKay, A.
Robinson, D.
Wu, Y.-.
Doherty, R.
Marini, B.
Tran, D.
Garton, H.
Muraszko, K.
Robertson, P.
Leonard, M.
Zhao, L.
Bixby, D.
Peterson, L.
Camelo-Piragua, S.
Jones, C.
Mody, R.
Lowenstein, P.R.
Castro, M.G.
(2016). Characterizing and targeting PDGFRA alterations in pediatric high-grade glioma. Oncotarget,
Vol.7
(40),
pp. 65696-65706.
show abstract
full text
Pediatric high-grade glioma (HGG, WHO Grade III and IV) is a devastating brain tumor with a median survival of less than two years. PDGFRA is frequently mutated/ amplified in pediatric HGG, but the significance of this finding has not been fully characterized. We hypothesize that alterations of PDGFRA will promote distinct prognostic and treatment implications in pediatric HGG. In order to characterize the impact of PDGFR pathway alterations, we integrated genomic data from pediatric HGG patients (n=290) from multiple pediatric datasets and sequencing platforms. Integration of multiple human datasets showed that PDGFRA mutation, but not amplification, was associated with older age in pediatric HGG (P= <0.0001). In multivariate analysis, PDGFRA mutation was correlated with worse prognosis (P = 0.026), while PDGFRA amplification was not (P = 0.11). By Kaplan-Meier analysis, non-brainstem HGG with PDGFRA amplification carried a worse prognosis than non-brainstem HGG without PDGFRA amplification (P = 0.021). There were no pediatric patients with PDGFRA-amplified HGG that survived longer than two years. Additionally, we performed paired molecular profiling (germline / tumor / primary cell culture) and targeting of an infant thalamic HGG with amplification and outlier increased expression of PDGFRA. Dasatinib inhibited proliferation most effectively. In summary, integration of the largest genomic dataset of pediatric HGG to date, allowed us to highlight that PDGFRA mutation is found in older pediatric patients and that PDGFRA amplification is prognostic in non-brainstem HGG. Future precision-medicine based clinical trials for pediatric patients with PDGFRA-altered HGG should consider the optimized delivery of dasatinib..
Bidinotto, L.T.
Torrieri, R.
Mackay, A.
Almeida, G.C.
Viana-Pereira, M.
Cruvinel-Carloni, A.
Spina, M.L.
Campanella, N.C.
Pereira de Menezes, W.
Clara, C.A.
Becker, A.P.
Jones, C.
Reis, R.M.
(2016). Copy Number Profiling of Brazilian Astrocytomas. G3 (bethesda),
Vol.6
(7),
pp. 1867-1878.
show abstract
full text
Copy number alterations (CNA) are one of the driving mechanisms of glioma tumorigenesis, and are currently used as important biomarkers in the routine setting. Therefore, we performed CNA profiling of 65 astrocytomas of distinct malignant grades (WHO grade I-IV) of Brazilian origin, using array-CGH and microsatellite instability analysis (MSI), and investigated their correlation with TERT and IDH1 mutational status and clinico-pathological features. Furthermore, in silico analysis using the Oncomine database was performed to validate our findings and extend the findings to gene expression level. We found that the number of genomic alterations increases in accordance with glioma grade. In glioblastomas (GBM), the most common alterations were gene amplifications (PDGFRA, KIT, KDR, EGFR, and MET) and deletions (CDKN2A and PTEN) Log-rank analysis correlated EGFR amplification and/or chr7 gain with better survival of the patients. MSI was observed in 11% of GBMs. A total of 69% of GBMs presented TERT mutation, whereas IDH1 mutation was most frequent in diffuse (85.7%) and anaplastic (100%) astrocytomas. The combination of 1p19q deletion and TERT and IDH1 mutational status separated tumor groups that showed distinct age of diagnosis and outcome. In silico validation pointed to less explored genes that may be worthy of future investigation, such as CDK2, DMRTA1, and MTAP Herein, using an extensive integrated analysis, we indicated potentially important genes, not extensively studied in gliomas, that could be further explored to assess their biological and clinical impact in astrocytomas..
Carceller, F.
Fowkes, L.A.
Khabra, K.
Moreno, L.
Saran, F.
Burford, A.
Mackay, A.
Jones, D.T.
Hovestadt, V.
Marshall, L.V.
Vaidya, S.
Mandeville, H.
Jerome, N.
Bridges, L.R.
Laxton, R.
Al-Sarraj, S.
Pfister, S.M.
Leach, M.O.
Pearson, A.D.
Jones, C.
Koh, D.-.
Zacharoulis, S.
(2016). Pseudoprogression in children, adolescents and young adults with non-brainstem high grade glioma and diffuse intrinsic pontine glioma. J neurooncol,
Vol.129
(1),
pp. 109-121.
show abstract
Pseudoprogression (PsP) is a treatment-related phenomenon which hinders response interpretation. Its prevalence and clinical impact have not been evaluated in children/adolescents. We assessed the characteristics, risk factors and prognosis of PsP in children/adolescents and young-adults diagnosed with non-brainstem high grade gliomas (HGG) and diffuse intrinsic pontine gliomas (DIPG). Patients aged 1-21 years diagnosed with HGG or DIPG between 1995 and 2012 who had completed radiotherapy were eligible. PsP was assessed according to study-specific criteria and correlated with first-line treatment, molecular biomarkers and survival. Ninety-one patients (47 HGG, 44 DIPG) were evaluable. Median age: 10 years (range, 2-20). Eleven episodes of PsP were observed in 10 patients (4 HGG, 6 DIPG). Rates of PsP: 8.5 % (HGG); 13.6 % (DIPG). Two episodes of PsP were based on clinical findings alone; nine episodes had concurrent radiological changes: increased size of lesions (n = 5), new focal enhancement (n = 4). Temozolomide, MGMT methylation or H3F3A mutations were not found to be associated with increased occurrence of PsP. For HGG, 1-year progression-free survival (PFS) was 41.9 % no-PsP versus 100 % PsP (p = 0.041); differences in 1-year overall survival (OS) were not significant. For DIPG, differences in 1-year PFS and OS were not statistically significant. Hazard ratio (95 %CI) of PsP for OS was 0.551 (0.168-1.803; p = 0.325) in HGG; and 0.308 (0.107-0.882; p = 0.028) in DIPG. PsP occurred in both pediatric HGG and DIPG patients at a comparable rate to adult HGG. PsP was associated with improved 1-yr PFS in HGG patients. PsP had a protective effect upon OS in DIPG patients..
Xipell, E.
Aragón, T.
Martínez-Velez, N.
Vera, B.
Idoate, M.A.
Martínez-Irujo, J.J.
Garzón, A.G.
Gonzalez-Huarriz, M.
Acanda, A.M.
Jones, C.
Lang, F.F.
Fueyo, J.
Gomez-Manzano, C.
Alonso, M.M.
(2016). Endoplasmic reticulum stress-inducing drugs sensitize glioma cells to temozolomide through downregulation of MGMT, MPG, and Rad51. Neuro oncol,
Vol.18
(8),
pp. 1109-1119.
show abstract
BACKGROUND: Endoplasmic reticulum (ER) stress results from protein misfolding imbalance and has been postulated as a therapeutic strategy. ER stress activates the unfolded protein response which leads to a complex cellular response, including the upregulation of aberrant protein degradation in the ER, with the goal of resolving that stress. O(6)-methylguanine DNA methyltransferase (MGMT), N-methylpurine DNA glycosylase (MPG), and Rad51 are DNA damage repair proteins that mediate resistance to temozolomide in glioblastoma. In this work we sought to evaluate whether ER stress-inducing drugs were able to downmodulate DNA damage repair proteins and become candidates to combine with temozolomide. METHODS: MTT assays were performed to evaluate the cytotoxicity of the treatments. The expression of proteins was evaluated using western blot and immunofluorescence. In vivo studies were performed using 2 orthotopic glioblastoma models in nude mice to evaluate the efficacy of the treatments. All statistical tests were 2-sided. RESULTS: Treatment of glioblastoma cells with ER stress-inducing drugs leads to downregulation of MGMT, MPG, and Rad51. Inhibition of ER stress through pharmacological treatment resulted in rescue of MGMT, MPG, and Rad51 protein levels. Moreover, treatment of glioblastoma cells with salinomycin, an ER stress-inducing drug, and temozolomide resulted in enhanced DNA damage and a synergistic antitumor effect in vitro. Of importance, treatment with salinomycin/temozolomide resulted in a significant antiglioma effect in 2 aggressive orthotopic intracranial brain tumor models. CONCLUSIONS: These findings provide a strong rationale for combining temozolomide with ER stress-inducing drugs as an alternative therapeutic strategy for glioblastoma..
Boult, J.K.
Borri, M.
Jury, A.
Popov, S.
Box, G.
Perryman, L.
Eccles, S.A.
Jones, C.
Robinson, S.P.
(2016). Investigating intracranial tumour growth patterns with multiparametric MRI incorporating Gd-DTPA and USPIO-enhanced imaging. Nmr biomed,
Vol.29
(11),
pp. 1608-1617.
show abstract
full text
High grade and metastatic brain tumours exhibit considerable spatial variations in proliferation, angiogenesis, invasion, necrosis and oedema. Vascular heterogeneity arising from vascular co-option in regions of invasive growth (in which the blood-brain barrier remains intact) and neoangiogenesis is a major challenge faced in the assessment of brain tumours by conventional MRI. A multiparametric MRI approach, incorporating native measurements and both Gd-DTPA (Magnevist) and ultrasmall superparamagnetic iron oxide (P904)-enhanced imaging, was used in combination with histogram and unsupervised cluster analysis using a k-means algorithm to examine the spatial distribution of vascular parameters, water diffusion characteristics and invasion in intracranially propagated rat RG2 gliomas and human MDA-MB-231 LM2-4 breast adenocarcinomas in mice. Both tumour models presented with higher ΔR1 (the change in transverse relaxation rate R1 induced by Gd-DTPA), fractional blood volume (fBV) and apparent diffusion coefficient than uninvolved regions of the brain. MDA-MB-231 LM2-4 tumours were less densely cellular than RG2 tumours and exhibited substantial local invasion, associated with oedema, whereas invasion in RG2 tumours was minimal. These additional features were reflected in the more heterogeneous appearance of MDA-MB-231 LM2-4 tumours on T2 -weighted images and maps of functional MRI parameters. Unsupervised cluster analysis separated subregions with distinct functional properties; areas with a low fBV and relatively impermeable blood vessels (low ΔR1 ) were predominantly located at the tumour margins, regions of MDA-MB-231 LM2-4 tumours with relatively high levels of water diffusion and low vascular permeability and/or fBV corresponded to histologically identified regions of invasion and oedema, and areas of mismatch between vascular permeability and blood volume were identified. We demonstrate that dual contrast MRI and evaluation of tissue diffusion properties, coupled with cluster analysis, allows for the assessment of heterogeneity within invasive brain tumours and the designation of functionally diverse subregions that may provide more informative predictive biomarkers..
International Cancer Genome Consortium PedBrain Tumor Project,
(2016). Recurrent MET fusion genes represent a drug target in pediatric glioblastoma. Nat med,
Vol.22
(11),
pp. 1314-1320.
show abstract
full text
Pediatric glioblastoma is one of the most common and most deadly brain tumors in childhood. Using an integrative genetic analysis of 53 pediatric glioblastomas and five in vitro model systems, we identified previously unidentified gene fusions involving the MET oncogene in ∼10% of cases. These MET fusions activated mitogen-activated protein kinase (MAPK) signaling and, in cooperation with lesions compromising cell cycle regulation, induced aggressive glial tumors in vivo. MET inhibitors suppressed MET tumor growth in xenograft models. Finally, we treated a pediatric patient bearing a MET-fusion-expressing glioblastoma with the targeted inhibitor crizotinib. This therapy led to substantial tumor shrinkage and associated relief of symptoms, but new treatment-resistant lesions appeared, indicating that combination therapies are likely necessary to achieve a durable clinical response..
Yogev, O.
Barker, K.
Sikka, A.
Almeida, G.S.
Hallsworth, A.
Smith, L.M.
Jamin, Y.
Ruddle, R.
Koers, A.
Webber, H.T.
Raynaud, F.I.
Popov, S.
Jones, C.
Petrie, K.
Robinson, S.P.
Keun, H.C.
Chesler, L.
(2016). p53 Loss in MYC-Driven Neuroblastoma Leads to Metabolic Adaptations Supporting Radioresistance. Cancer res,
Vol.76
(10),
pp. 3025-3035.
show abstract
full text
Neuroblastoma is the most common childhood extracranial solid tumor. In high-risk cases, many of which are characterized by amplification of MYCN, outcome remains poor. Mutations in the p53 (TP53) tumor suppressor are rare at diagnosis, but evidence suggests that p53 function is often impaired in relapsed, treatment-resistant disease. To address the role of p53 loss of function in the development and pathogenesis of high-risk neuroblastoma, we generated a MYCN-driven genetically engineered mouse model in which the tamoxifen-inducible p53ER(TAM) fusion protein was expressed from a knock-in allele (Th-MYCN/Trp53(KI)). We observed no significant differences in tumor-free survival between Th-MYCN mice heterozygous for Trp53(KI) (n = 188) and Th-MYCN mice with wild-type p53 (n = 101). Conversely, the survival of Th-MYCN/Trp53(KI/KI) mice lacking functional p53 (n = 60) was greatly reduced. We found that Th-MYCN/Trp53(KI/KI) tumors were resistant to ionizing radiation (IR), as expected. However, restoration of functional p53ER(TAM) reinstated sensitivity to IR in only 50% of Th-MYCN/Trp53(KI/KI) tumors, indicating the acquisition of additional resistance mechanisms. Gene expression and metabolic analyses indicated that the principal acquired mechanism of resistance to IR in the absence of functional p53 was metabolic adaptation in response to chronic oxidative stress. Tumors exhibited increased antioxidant metabolites and upregulation of glutathione S-transferase pathway genes, including Gstp1 and Gstz1, which are associated with poor outcome in human neuroblastoma. Accordingly, glutathione depletion by buthionine sulfoximine together with restoration of p53 activity resensitized tumors to IR. Our findings highlight the complex pathways operating in relapsed neuroblastomas and the need for combination therapies that target the diverse resistance mechanisms at play. Cancer Res; 76(10); 3025-35. ©2016 AACR..
Sturm, D.
Orr, B.A.
Toprak, U.H.
Hovestadt, V.
Jones, D.T.
Capper, D.
Sill, M.
Buchhalter, I.
Northcott, P.A.
Leis, I.
Ryzhova, M.
Koelsche, C.
Pfaff, E.
Allen, S.J.
Balasubramanian, G.
Worst, B.C.
Pajtler, K.W.
Brabetz, S.
Johann, P.D.
Sahm, F.
Reimand, J.
Mackay, A.
Carvalho, D.M.
Remke, M.
Phillips, J.J.
Perry, A.
Cowdrey, C.
Drissi, R.
Fouladi, M.
Giangaspero, F.
Łastowska, M.
Grajkowska, W.
Scheurlen, W.
Pietsch, T.
Hagel, C.
Gojo, J.
Lötsch, D.
Berger, W.
Slavc, I.
Haberler, C.
Jouvet, A.
Holm, S.
Hofer, S.
Prinz, M.
Keohane, C.
Fried, I.
Mawrin, C.
Scheie, D.
Mobley, B.C.
Schniederjan, M.J.
Santi, M.
Buccoliero, A.M.
Dahiya, S.
Kramm, C.M.
von Bueren, A.O.
von Hoff, K.
Rutkowski, S.
Herold-Mende, C.
Frühwald, M.C.
Milde, T.
Hasselblatt, M.
Wesseling, P.
Rößler, J.
Schüller, U.
Ebinger, M.
Schittenhelm, J.
Frank, S.
Grobholz, R.
Vajtai, I.
Hans, V.
Schneppenheim, R.
Zitterbart, K.
Collins, V.P.
Aronica, E.
Varlet, P.
Puget, S.
Dufour, C.
Grill, J.
Figarella-Branger, D.
Wolter, M.
Schuhmann, M.U.
Shalaby, T.
Grotzer, M.
van Meter, T.
Monoranu, C.-.
Felsberg, J.
Reifenberger, G.
Snuderl, M.
Forrester, L.A.
Koster, J.
Versteeg, R.
Volckmann, R.
van Sluis, P.
Wolf, S.
Mikkelsen, T.
Gajjar, A.
Aldape, K.
Moore, A.S.
Taylor, M.D.
Jones, C.
Jabado, N.
Karajannis, M.A.
Eils, R.
Schlesner, M.
Lichter, P.
von Deimling, A.
Pfister, S.M.
Ellison, D.W.
Korshunov, A.
Kool, M.
(2016). New Brain Tumor Entities Emerge from Molecular Classification of CNS-PNETs. Cell,
Vol.164
(5),
pp. 1060-1072.
show abstract
Primitive neuroectodermal tumors of the central nervous system (CNS-PNETs) are highly aggressive, poorly differentiated embryonal tumors occurring predominantly in young children but also affecting adolescents and adults. Herein, we demonstrate that a significant proportion of institutionally diagnosed CNS-PNETs display molecular profiles indistinguishable from those of various other well-defined CNS tumor entities, facilitating diagnosis and appropriate therapy for patients with these tumors. From the remaining fraction of CNS-PNETs, we identify four new CNS tumor entities, each associated with a recurrent genetic alteration and distinct histopathological and clinical features. These new molecular entities, designated "CNS neuroblastoma with FOXR2 activation (CNS NB-FOXR2)," "CNS Ewing sarcoma family tumor with CIC alteration (CNS EFT-CIC)," "CNS high-grade neuroepithelial tumor with MN1 alteration (CNS HGNET-MN1)," and "CNS high-grade neuroepithelial tumor with BCOR alteration (CNS HGNET-BCOR)," will enable meaningful clinical trials and the development of therapeutic strategies for patients affected by poorly differentiated CNS tumors..
Klionsky, D.J.
Abdelmohsen, K.
Abe, A.
Abedin, M.J.
Abeliovich, H.
Acevedo Arozena, A.
Adachi, H.
Adams, C.M.
Adams, P.D.
Adeli, K.
Adhihetty, P.J.
Adler, S.G.
Agam, G.
Agarwal, R.
Aghi, M.K.
Agnello, M.
Agostinis, P.
Aguilar, P.V.
Aguirre-Ghiso, J.
Airoldi, E.M.
Ait-Si-Ali, S.
Akematsu, T.
Akporiaye, E.T.
Al-Rubeai, M.
Albaiceta, G.M.
Albanese, C.
Albani, D.
Albert, M.L.
Aldudo, J.
Algül, H.
Alirezaei, M.
Alloza, I.
Almasan, A.
Almonte-Beceril, M.
Alnemri, E.S.
Alonso, C.
Altan-Bonnet, N.
Altieri, D.C.
Alvarez, S.
Alvarez-Erviti, L.
Alves, S.
Amadoro, G.
Amano, A.
Amantini, C.
Ambrosio, S.
Amelio, I.
Amer, A.O.
Amessou, M.
Amon, A.
An, Z.
Anania, F.A.
Andersen, S.U.
Andley, U.P.
Andreadi, C.K.
Andrieu-Abadie, N.
Anel, A.
Ann, D.K.
Anoopkumar-Dukie, S.
Antonioli, M.
Aoki, H.
Apostolova, N.
Aquila, S.
Aquilano, K.
Araki, K.
Arama, E.
Aranda, A.
Araya, J.
Arcaro, A.
Arias, E.
Arimoto, H.
Ariosa, A.R.
Armstrong, J.L.
Arnould, T.
Arsov, I.
Asanuma, K.
Askanas, V.
Asselin, E.
Atarashi, R.
Atherton, S.S.
Atkin, J.D.
Attardi, L.D.
Auberger, P.
Auburger, G.
Aurelian, L.
Autelli, R.
Avagliano, L.
Avantaggiati, M.L.
Avrahami, L.
Awale, S.
Azad, N.
Bachetti, T.
Backer, J.M.
Bae, D.-.
Bae, J.-.
Bae, O.-.
Bae, S.H.
Baehrecke, E.H.
Baek, S.-.
Baghdiguian, S.
Bagniewska-Zadworna, A.
Bai, H.
Bai, J.
Bai, X.-.
Bailly, Y.
Balaji, K.N.
Balduini, W.
Ballabio, A.
Balzan, R.
Banerjee, R.
Bánhegyi, G.
Bao, H.
Barbeau, B.
Barrachina, M.D.
Barreiro, E.
Bartel, B.
Bartolomé, A.
Bassham, D.C.
Bassi, M.T.
Bast, R.C.
Basu, A.
Batista, M.T.
Batoko, H.
Battino, M.
Bauckman, K.
Baumgarner, B.L.
Bayer, K.U.
Beale, R.
Beaulieu, J.-.
Beck, G.R.
Becker, C.
Beckham, J.D.
Bédard, P.-.
Bednarski, P.J.
Begley, T.J.
Behl, C.
Behrends, C.
Behrens, G.M.
Behrns, K.E.
Bejarano, E.
Belaid, A.
Belleudi, F.
Bénard, G.
Berchem, G.
Bergamaschi, D.
Bergami, M.
Berkhout, B.
Berliocchi, L.
Bernard, A.
Bernard, M.
Bernassola, F.
Bertolotti, A.
Bess, A.S.
Besteiro, S.
Bettuzzi, S.
Bhalla, S.
Bhattacharyya, S.
Bhutia, S.K.
Biagosch, C.
Bianchi, M.W.
Biard-Piechaczyk, M.
Billes, V.
Bincoletto, C.
Bingol, B.
Bird, S.W.
Bitoun, M.
Bjedov, I.
Blackstone, C.
Blanc, L.
Blanco, G.A.
Blomhoff, H.K.
Boada-Romero, E.
Böckler, S.
Boes, M.
Boesze-Battaglia, K.
Boise, L.H.
Bolino, A.
Boman, A.
Bonaldo, P.
Bordi, M.
Bosch, J.
Botana, L.M.
Botti, J.
Bou, G.
Bouché, M.
Bouchecareilh, M.
Boucher, M.-.
Boulton, M.E.
Bouret, S.G.
Boya, P.
Boyer-Guittaut, M.
Bozhkov, P.V.
Brady, N.
Braga, V.M.
Brancolini, C.
Braus, G.H.
Bravo-San Pedro, J.M.
Brennan, L.A.
Bresnick, E.H.
Brest, P.
Bridges, D.
Bringer, M.-.
Brini, M.
Brito, G.C.
Brodin, B.
Brookes, P.S.
Brown, E.J.
Brown, K.
Broxmeyer, H.E.
Bruhat, A.
Brum, P.C.
Brumell, J.H.
Brunetti-Pierri, N.
Bryson-Richardson, R.J.
Buch, S.
Buchan, A.M.
Budak, H.
Bulavin, D.V.
Bultman, S.J.
Bultynck, G.
Bumbasirevic, V.
Burelle, Y.
Burke, R.E.
Burmeister, M.
Bütikofer, P.
Caberlotto, L.
Cadwell, K.
Cahova, M.
Cai, D.
Cai, J.
Cai, Q.
Calatayud, S.
Camougrand, N.
Campanella, M.
Campbell, G.R.
Campbell, M.
Campello, S.
Candau, R.
Caniggia, I.
Cantoni, L.
Cao, L.
Caplan, A.B.
Caraglia, M.
Cardinali, C.
Cardoso, S.M.
Carew, J.S.
Carleton, L.A.
Carlin, C.R.
Carloni, S.
Carlsson, S.R.
Carmona-Gutierrez, D.
Carneiro, L.A.
Carnevali, O.
Carra, S.
Carrier, A.
Carroll, B.
Casas, C.
Casas, J.
Cassinelli, G.
Castets, P.
Castro-Obregon, S.
Cavallini, G.
Ceccherini, I.
Cecconi, F.
Cederbaum, A.I.
Ceña, V.
Cenci, S.
Cerella, C.
Cervia, D.
Cetrullo, S.
Chaachouay, H.
Chae, H.-.
Chagin, A.S.
Chai, C.-.
Chakrabarti, G.
Chamilos, G.
Chan, E.Y.
Chan, M.T.
Chandra, D.
Chandra, P.
Chang, C.-.
Chang, R.C.
Chang, T.Y.
Chatham, J.C.
Chatterjee, S.
Chauhan, S.
Che, Y.
Cheetham, M.E.
Cheluvappa, R.
Chen, C.-.
Chen, G.
Chen, G.-.
Chen, G.
Chen, H.
Chen, J.W.
Chen, J.-.
Chen, M.
Chen, M.
Chen, P.
Chen, Q.
Chen, Q.
Chen, S.-.
Chen, S.
Chen, S.S.
Chen, W.
Chen, W.-.
Chen, W.Q.
Chen, W.
Chen, X.
Chen, Y.-.
Chen, Y.-.
Chen, Y.
Chen, Y.
Chen, Y.
Chen, Y.-.
Chen, Y.-.
Chen, Y.
Chen, Z.
Chen, Z.
Cheng, A.
Cheng, C.H.
Cheng, H.
Cheong, H.
Cherry, S.
Chesney, J.
Cheung, C.H.
Chevet, E.
Chi, H.C.
Chi, S.-.
Chiacchiera, F.
Chiang, H.-.
Chiarelli, R.
Chiariello, M.
Chieppa, M.
Chin, L.-.
Chiong, M.
Chiu, G.N.
Cho, D.-.
Cho, S.-.
Cho, W.C.
Cho, Y.-.
Cho, Y.-.
Choi, A.M.
Choi, E.-.
Choi, E.-.
Choi, J.
Choi, M.E.
Choi, S.-.
Chou, T.-.
Chouaib, S.
Choubey, D.
Choubey, V.
Chow, K.-.
Chowdhury, K.
Chu, C.T.
Chuang, T.-.
Chun, T.
Chung, H.
Chung, T.
Chung, Y.-.
Chwae, Y.-.
Cianfanelli, V.
Ciarcia, R.
Ciechomska, I.A.
Ciriolo, M.R.
Cirone, M.
Claerhout, S.
Clague, M.J.
Clària, J.
Clarke, P.G.
Clarke, R.
Clementi, E.
Cleyrat, C.
Cnop, M.
Coccia, E.M.
Cocco, T.
Codogno, P.
Coers, J.
Cohen, E.E.
Colecchia, D.
Coletto, L.
Coll, N.S.
Colucci-Guyon, E.
Comincini, S.
Condello, M.
Cook, K.L.
Coombs, G.H.
Cooper, C.D.
Cooper, J.M.
Coppens, I.
Corasaniti, M.T.
Corazzari, M.
Corbalan, R.
Corcelle-Termeau, E.
Cordero, M.D.
Corral-Ramos, C.
Corti, O.
Cossarizza, A.
Costelli, P.
Costes, S.
Cotman, S.L.
Coto-Montes, A.
Cottet, S.
Couve, E.
Covey, L.R.
Cowart, L.A.
Cox, J.S.
Coxon, F.P.
Coyne, C.B.
Cragg, M.S.
Craven, R.J.
Crepaldi, T.
Crespo, J.L.
Criollo, A.
Crippa, V.
Cruz, M.T.
Cuervo, A.M.
Cuezva, J.M.
Cui, T.
Cutillas, P.R.
Czaja, M.J.
Czyzyk-Krzeska, M.F.
Dagda, R.K.
Dahmen, U.
Dai, C.
Dai, W.
Dai, Y.
Dalby, K.N.
Dalla Valle, L.
Dalmasso, G.
D'Amelio, M.
Damme, M.
Darfeuille-Michaud, A.
Dargemont, C.
Darley-Usmar, V.M.
Dasarathy, S.
Dasgupta, B.
Dash, S.
Dass, C.R.
Davey, H.M.
Davids, L.M.
Dávila, D.
Davis, R.J.
Dawson, T.M.
Dawson, V.L.
Daza, P.
de Belleroche, J.
de Figueiredo, P.
de Figueiredo, R.C.
de la Fuente, J.
De Martino, L.
De Matteis, A.
De Meyer, G.R.
De Milito, A.
De Santi, M.
de Souza, W.
De Tata, V.
De Zio, D.
Debnath, J.
Dechant, R.
Decuypere, J.-.
Deegan, S.
Dehay, B.
Del Bello, B.
Del Re, D.P.
Delage-Mourroux, R.
Delbridge, L.M.
Deldicque, L.
Delorme-Axford, E.
Deng, Y.
Dengjel, J.
Denizot, M.
Dent, P.
Der, C.J.
Deretic, V.
Derrien, B.
Deutsch, E.
Devarenne, T.P.
Devenish, R.J.
Di Bartolomeo, S.
Di Daniele, N.
Di Domenico, F.
Di Nardo, A.
Di Paola, S.
Di Pietro, A.
Di Renzo, L.
DiAntonio, A.
Díaz-Araya, G.
Díaz-Laviada, I.
Diaz-Meco, M.T.
Diaz-Nido, J.
Dickey, C.A.
Dickson, R.C.
Diederich, M.
Digard, P.
Dikic, I.
Dinesh-Kumar, S.P.
Ding, C.
Ding, W.-.
Ding, Z.
Dini, L.
Distler, J.H.
Diwan, A.
Djavaheri-Mergny, M.
Dmytruk, K.
Dobson, R.C.
Doetsch, V.
Dokladny, K.
Dokudovskaya, S.
Donadelli, M.
Dong, X.C.
Dong, X.
Dong, Z.
Donohue, T.M.
Doran, K.S.
D'Orazi, G.
Dorn, G.W.
Dosenko, V.
Dridi, S.
Drucker, L.
Du, J.
Du, L.-.
Du, L.
du Toit, A.
Dua, P.
Duan, L.
Duann, P.
Dubey, V.K.
Duchen, M.R.
Duchosal, M.A.
Duez, H.
Dugail, I.
Dumit, V.I.
Duncan, M.C.
Dunlop, E.A.
Dunn, W.A.
Dupont, N.
Dupuis, L.
Durán, R.V.
Durcan, T.M.
Duvezin-Caubet, S.
Duvvuri, U.
Eapen, V.
Ebrahimi-Fakhari, D.
Echard, A.
Eckhart, L.
Edelstein, C.L.
Edinger, A.L.
Eichinger, L.
Eisenberg, T.
Eisenberg-Lerner, A.
Eissa, N.T.
El-Deiry, W.S.
El-Khoury, V.
Elazar, Z.
Eldar-Finkelman, H.
Elliott, C.J.
Emanuele, E.
Emmenegger, U.
Engedal, N.
Engelbrecht, A.-.
Engelender, S.
Enserink, J.M.
Erdmann, R.
Erenpreisa, J.
Eri, R.
Eriksen, J.L.
Erman, A.
Escalante, R.
Eskelinen, E.-.
Espert, L.
Esteban-Martínez, L.
Evans, T.J.
Fabri, M.
Fabrias, G.
Fabrizi, C.
Facchiano, A.
Færgeman, N.J.
Faggioni, A.
Fairlie, W.D.
Fan, C.
Fan, D.
Fan, J.
Fang, S.
Fanto, M.
Fanzani, A.
Farkas, T.
Faure, M.
Favier, F.B.
Fearnhead, H.
Federici, M.
Fei, E.
Felizardo, T.C.
Feng, H.
Feng, Y.
Feng, Y.
Ferguson, T.A.
Fernández, Á.F.
Fernandez-Barrena, M.G.
Fernandez-Checa, J.C.
Fernández-López, A.
Fernandez-Zapico, M.E.
Feron, O.
Ferraro, E.
Ferreira-Halder, C.V.
Fesus, L.
Feuer, R.
Fiesel, F.C.
Filippi-Chiela, E.C.
Filomeni, G.
Fimia, G.M.
Fingert, J.H.
Finkbeiner, S.
Finkel, T.
Fiorito, F.
Fisher, P.B.
Flajolet, M.
Flamigni, F.
Florey, O.
Florio, S.
Floto, R.A.
Folini, M.
Follo, C.
Fon, E.A.
Fornai, F.
Fortunato, F.
Fraldi, A.
Franco, R.
Francois, A.
François, A.
Frankel, L.B.
Fraser, I.D.
Frey, N.
Freyssenet, D.G.
Frezza, C.
Friedman, S.L.
Frigo, D.E.
Fu, D.
Fuentes, J.M.
Fueyo, J.
Fujitani, Y.
Fujiwara, Y.
Fujiya, M.
Fukuda, M.
Fulda, S.
Fusco, C.
Gabryel, B.
Gaestel, M.
Gailly, P.
Gajewska, M.
Galadari, S.
Galili, G.
Galindo, I.
Galindo, M.F.
Galliciotti, G.
Galluzzi, L.
Galluzzi, L.
Galy, V.
Gammoh, N.
Gandy, S.
Ganesan, A.K.
Ganesan, S.
Ganley, I.G.
Gannagé, M.
Gao, F.-.
Gao, F.
Gao, J.-.
García Nannig, L.
García Véscovi, E.
Garcia-Macía, M.
Garcia-Ruiz, C.
Garg, A.D.
Garg, P.K.
Gargini, R.
Gassen, N.C.
Gatica, D.
Gatti, E.
Gavard, J.
Gavathiotis, E.
Ge, L.
Ge, P.
Ge, S.
Gean, P.-.
Gelmetti, V.
Genazzani, A.A.
Geng, J.
Genschik, P.
Gerner, L.
Gestwicki, J.E.
Gewirtz, D.A.
Ghavami, S.
Ghigo, E.
Ghosh, D.
Giammarioli, A.M.
Giampieri, F.
Giampietri, C.
Giatromanolaki, A.
Gibbings, D.J.
Gibellini, L.
Gibson, S.B.
Ginet, V.
Giordano, A.
Giorgini, F.
Giovannetti, E.
Girardin, S.E.
Gispert, S.
Giuliano, S.
Gladson, C.L.
Glavic, A.
Gleave, M.
Godefroy, N.
Gogal, R.M.
Gokulan, K.
Goldman, G.H.
Goletti, D.
Goligorsky, M.S.
Gomes, A.V.
Gomes, L.C.
Gomez, H.
Gomez-Manzano, C.
Gómez-Sánchez, R.
Gonçalves, D.A.
Goncu, E.
Gong, Q.
Gongora, C.
Gonzalez, C.B.
Gonzalez-Alegre, P.
Gonzalez-Cabo, P.
González-Polo, R.A.
Goping, I.S.
Gorbea, C.
Gorbunov, N.V.
Goring, D.R.
Gorman, A.M.
Gorski, S.M.
Goruppi, S.
Goto-Yamada, S.
Gotor, C.
Gottlieb, R.A.
Gozes, I.
Gozuacik, D.
Graba, Y.
Graef, M.
Granato, G.E.
Grant, G.D.
Grant, S.
Gravina, G.L.
Green, D.R.
Greenhough, A.
Greenwood, M.T.
Grimaldi, B.
Gros, F.
Grose, C.
Groulx, J.-.
Gruber, F.
Grumati, P.
Grune, T.
Guan, J.-.
Guan, K.-.
Guerra, B.
Guillen, C.
Gulshan, K.
Gunst, J.
Guo, C.
Guo, L.
Guo, M.
Guo, W.
Guo, X.-.
Gust, A.A.
Gustafsson, Å.B.
Gutierrez, E.
Gutierrez, M.G.
Gwak, H.-.
Haas, A.
Haber, J.E.
Hadano, S.
Hagedorn, M.
Hahn, D.R.
Halayko, A.J.
Hamacher-Brady, A.
Hamada, K.
Hamai, A.
Hamann, A.
Hamasaki, M.
Hamer, I.
Hamid, Q.
Hammond, E.M.
Han, F.
Han, W.
Handa, J.T.
Hanover, J.A.
Hansen, M.
Harada, M.
Harhaji-Trajkovic, L.
Harper, J.W.
Harrath, A.H.
Harris, A.L.
Harris, J.
Hasler, U.
Hasselblatt, P.
Hasui, K.
Hawley, R.G.
Hawley, T.S.
He, C.
He, C.Y.
He, F.
He, G.
He, R.-.
He, X.-.
He, Y.-.
He, Y.-.
Heath, J.K.
Hébert, M.-.
Heinzen, R.A.
Helgason, G.V.
Hensel, M.
Henske, E.P.
Her, C.
Herman, P.K.
Hernández, A.
Hernandez, C.
Hernández-Tiedra, S.
Hetz, C.
Hiesinger, P.R.
Higaki, K.
Hilfiker, S.
Hill, B.G.
Hill, J.A.
Hill, W.D.
Hino, K.
Hofius, D.
Hofman, P.
Höglinger, G.U.
Höhfeld, J.
Holz, M.K.
Hong, Y.
Hood, D.A.
Hoozemans, J.J.
Hoppe, T.
Hsu, C.
Hsu, C.-.
Hsu, L.-.
Hu, D.
Hu, G.
Hu, H.-.
Hu, H.
Hu, M.C.
Hu, Y.-.
Hu, Z.-.
Hua, F.
Hua, Y.
Huang, C.
Huang, H.-.
Huang, K.-.
Huang, K.-.
Huang, S.
Huang, S.
Huang, W.-.
Huang, Y.-.
Huang, Y.
Huang, Y.
Huber, T.B.
Huebbe, P.
Huh, W.-.
Hulmi, J.J.
Hur, G.M.
Hurley, J.H.
Husak, Z.
Hussain, S.N.
Hussain, S.
Hwang, J.J.
Hwang, S.
Hwang, T.I.
Ichihara, A.
Imai, Y.
Imbriano, C.
Inomata, M.
Into, T.
Iovane, V.
Iovanna, J.L.
Iozzo, R.V.
Ip, N.Y.
Irazoqui, J.E.
Iribarren, P.
Isaka, Y.
Isakovic, A.J.
Ischiropoulos, H.
Isenberg, J.S.
Ishaq, M.
Ishida, H.
Ishii, I.
Ishmael, J.E.
Isidoro, C.
Isobe, K.-.
Isono, E.
Issazadeh-Navikas, S.
Itahana, K.
Itakura, E.
Ivanov, A.I.
Iyer, A.K.
Izquierdo, J.M.
Izumi, Y.
Izzo, V.
Jäättelä, M.
Jaber, N.
Jackson, D.J.
Jackson, W.T.
Jacob, T.G.
Jacques, T.S.
Jagannath, C.
Jain, A.
Jana, N.R.
Jang, B.K.
Jani, A.
Janji, B.
Jannig, P.R.
Jansson, P.J.
Jean, S.
Jendrach, M.
Jeon, J.-.
Jessen, N.
Jeung, E.-.
Jia, K.
Jia, L.
Jiang, H.
Jiang, H.
Jiang, L.
Jiang, T.
Jiang, X.
Jiang, X.
Jiang, X.
Jiang, Y.
Jiang, Y.
Jiménez, A.
Jin, C.
Jin, H.
Jin, L.
Jin, M.
Jin, S.
Jinwal, U.K.
Jo, E.-.
Johansen, T.
Johnson, D.E.
Johnson, G.V.
Johnson, J.D.
Jonasch, E.
Jones, C.
Joosten, L.A.
Jordan, J.
Joseph, A.-.
Joseph, B.
Joubert, A.M.
Ju, D.
Ju, J.
Juan, H.-.
Juenemann, K.
Juhász, G.
Jung, H.S.
Jung, J.U.
Jung, Y.-.
Jungbluth, H.
Justice, M.J.
Jutten, B.
Kaakoush, N.O.
Kaarniranta, K.
Kaasik, A.
Kabuta, T.
Kaeffer, B.
Kågedal, K.
Kahana, A.
Kajimura, S.
Kakhlon, O.
Kalia, M.
Kalvakolanu, D.V.
Kamada, Y.
Kambas, K.
Kaminskyy, V.O.
Kampinga, H.H.
Kandouz, M.
Kang, C.
Kang, R.
Kang, T.-.
Kanki, T.
Kanneganti, T.-.
Kanno, H.
Kanthasamy, A.G.
Kantorow, M.
Kaparakis-Liaskos, M.
Kapuy, O.
Karantza, V.
Karim, M.R.
Karmakar, P.
Kaser, A.
Kaushik, S.
Kawula, T.
Kaynar, A.M.
Ke, P.-.
Ke, Z.-.
Kehrl, J.H.
Keller, K.E.
Kemper, J.K.
Kenworthy, A.K.
Kepp, O.
Kern, A.
Kesari, S.
Kessel, D.
Ketteler, R.
Kettelhut, I.D.
Khambu, B.
Khan, M.M.
Khandelwal, V.K.
Khare, S.
Kiang, J.G.
Kiger, A.A.
Kihara, A.
Kim, A.L.
Kim, C.H.
Kim, D.R.
Kim, D.-.
Kim, E.K.
Kim, H.Y.
Kim, H.-.
Kim, J.-.
Kim, J.H.
Kim, J.C.
Kim, J.H.
Kim, K.W.
Kim, M.D.
Kim, M.-.
Kim, P.K.
Kim, S.W.
Kim, S.-.
Kim, Y.-.
Kim, Y.
Kimchi, A.
Kimmelman, A.C.
Kimura, T.
King, J.S.
Kirkegaard, K.
Kirkin, V.
Kirshenbaum, L.A.
Kishi, S.
Kitajima, Y.
Kitamoto, K.
Kitaoka, Y.
Kitazato, K.
Kley, R.A.
Klimecki, W.T.
Klinkenberg, M.
Klucken, J.
Knævelsrud, H.
Knecht, E.
Knuppertz, L.
Ko, J.-.
Kobayashi, S.
Koch, J.C.
Koechlin-Ramonatxo, C.
Koenig, U.
Koh, Y.H.
Köhler, K.
Kohlwein, S.D.
Koike, M.
Komatsu, M.
Kominami, E.
Kong, D.
Kong, H.J.
Konstantakou, E.G.
Kopp, B.T.
Korcsmaros, T.
Korhonen, L.
Korolchuk, V.I.
Koshkina, N.V.
Kou, Y.
Koukourakis, M.I.
Koumenis, C.
Kovács, A.L.
Kovács, T.
Kovacs, W.J.
Koya, D.
Kraft, C.
Krainc, D.
Kramer, H.
Kravic-Stevovic, T.
Krek, W.
Kretz-Remy, C.
Krick, R.
Krishnamurthy, M.
Kriston-Vizi, J.
Kroemer, G.
Kruer, M.C.
Kruger, R.
Ktistakis, N.T.
Kuchitsu, K.
Kuhn, C.
Kumar, A.P.
Kumar, A.
Kumar, A.
Kumar, D.
Kumar, D.
Kumar, R.
Kumar, S.
Kundu, M.
Kung, H.-.
Kuno, A.
Kuo, S.-.
Kuret, J.
Kurz, T.
Kwok, T.
Kwon, T.K.
Kwon, Y.T.
Kyrmizi, I.
La Spada, A.R.
Lafont, F.
Lahm, T.
Lakkaraju, A.
Lam, T.
Lamark, T.
Lancel, S.
Landowski, T.H.
Lane, D.J.
Lane, J.D.
Lanzi, C.
Lapaquette, P.
Lapierre, L.R.
Laporte, J.
Laukkarinen, J.
Laurie, G.W.
Lavandero, S.
Lavie, L.
LaVoie, M.J.
Law, B.Y.
Law, H.K.
Law, K.B.
Layfield, R.
Lazo, P.A.
Le Cam, L.
Le Roch, K.G.
Le Stunff, H.
Leardkamolkarn, V.
Lecuit, M.
Lee, B.-.
Lee, C.-.
Lee, E.F.
Lee, G.M.
Lee, H.-.
Lee, H.
Lee, J.K.
Lee, J.
Lee, J.-.
Lee, J.H.
Lee, M.
Lee, M.-.
Lee, P.J.
Lee, S.W.
Lee, S.-.
Lee, S.-.
Lee, S.Y.
Lee, S.H.
Lee, S.S.
Lee, S.-.
Lee, S.
Lee, Y.-.
Lee, Y.J.
Lee, Y.H.
Leeuwenburgh, C.
Lefort, S.
Legouis, R.
Lei, J.
Lei, Q.-.
Leib, D.A.
Leibowitz, G.
Lekli, I.
Lemaire, S.D.
Lemasters, J.J.
Lemberg, M.K.
Lemoine, A.
Leng, S.
Lenz, G.
Lenzi, P.
Lerman, L.O.
Lettieri Barbato, D.
Leu, J.I.
Leung, H.Y.
Levine, B.
Lewis, P.A.
Lezoualc'h, F.
Li, C.
Li, F.
Li, F.-.
Li, J.
Li, K.
Li, L.
Li, M.
Li, M.
Li, Q.
Li, R.
Li, S.
Li, W.
Li, W.
Li, X.
Li, Y.
Lian, J.
Liang, C.
Liang, Q.
Liao, Y.
Liberal, J.
Liberski, P.P.
Lie, P.
Lieberman, A.P.
Lim, H.J.
Lim, K.-.
Lim, K.
Lima, R.T.
Lin, C.-.
Lin, C.-.
Lin, F.
Lin, F.
Lin, F.-.
Lin, K.
Lin, K.-.
Lin, P.-.
Lin, T.
Lin, W.-.
Lin, Y.-.
Lin, Y.
Linden, R.
Lindholm, D.
Lindqvist, L.M.
Lingor, P.
Linkermann, A.
Liotta, L.A.
Lipinski, M.M.
Lira, V.A.
Lisanti, M.P.
Liton, P.B.
Liu, B.
Liu, C.
Liu, C.-.
Liu, F.
Liu, H.-.
Liu, J.
Liu, J.-.
Liu, J.-.
Liu, K.
Liu, L.
Liu, L.
Liu, Q.
Liu, R.-.
Liu, S.
Liu, S.
Liu, W.
Liu, X.-.
Liu, X.
Liu, X.-.
Liu, X.
Liu, X.
Liu, X.
Liu, Y.
Liu, Y.
Liu, Z.
Liu, Z.
Liuzzi, J.P.
Lizard, G.
Ljujic, M.
Lodhi, I.J.
Logue, S.E.
Lokeshwar, B.L.
Long, Y.C.
Lonial, S.
Loos, B.
López-Otín, C.
López-Vicario, C.
Lorente, M.
Lorenzi, P.L.
Lõrincz, P.
Los, M.
Lotze, M.T.
Lovat, P.E.
Lu, B.
Lu, B.
Lu, J.
Lu, Q.
Lu, S.-.
Lu, S.
Lu, Y.
Luciano, F.
Luckhart, S.
Lucocq, J.M.
Ludovico, P.
Lugea, A.
Lukacs, N.W.
Lum, J.J.
Lund, A.H.
Luo, H.
Luo, J.
Luo, S.
Luparello, C.
Lyons, T.
Ma, J.
Ma, Y.
Ma, Y.
Ma, Z.
Machado, J.
Machado-Santelli, G.M.
Macian, F.
MacIntosh, G.C.
MacKeigan, J.P.
Macleod, K.F.
MacMicking, J.D.
MacMillan-Crow, L.A.
Madeo, F.
Madesh, M.
Madrigal-Matute, J.
Maeda, A.
Maeda, T.
Maegawa, G.
Maellaro, E.
Maes, H.
Magariños, M.
Maiese, K.
Maiti, T.K.
Maiuri, L.
Maiuri, M.C.
Maki, C.G.
Malli, R.
Malorni, W.
Maloyan, A.
Mami-Chouaib, F.
Man, N.
Mancias, J.D.
Mandelkow, E.-.
Mandell, M.A.
Manfredi, A.A.
Manié, S.N.
Manzoni, C.
Mao, K.
Mao, Z.
Mao, Z.-.
Marambaud, P.
Marconi, A.M.
Marelja, Z.
Marfe, G.
Margeta, M.
Margittai, E.
Mari, M.
Mariani, F.V.
Marin, C.
Marinelli, S.
Mariño, G.
Markovic, I.
Marquez, R.
Martelli, A.M.
Martens, S.
Martin, K.R.
Martin, S.J.
Martin, S.
Martin-Acebes, M.A.
Martín-Sanz, P.
Martinand-Mari, C.
Martinet, W.
Martinez, J.
Martinez-Lopez, N.
Martinez-Outschoorn, U.
Martínez-Velázquez, M.
Martinez-Vicente, M.
Martins, W.K.
Mashima, H.
Mastrianni, J.A.
Matarese, G.
Matarrese, P.
Mateo, R.
Matoba, S.
Matsumoto, N.
Matsushita, T.
Matsuura, A.
Matsuzawa, T.
Mattson, M.P.
Matus, S.
Maugeri, N.
Mauvezin, C.
Mayer, A.
Maysinger, D.
Mazzolini, G.D.
McBrayer, M.K.
McCall, K.
McCormick, C.
McInerney, G.M.
McIver, S.C.
McKenna, S.
McMahon, J.J.
McNeish, I.A.
Mechta-Grigoriou, F.
Medema, J.P.
Medina, D.L.
Megyeri, K.
Mehrpour, M.
Mehta, J.L.
Mei, Y.
Meier, U.-.
Meijer, A.J.
Meléndez, A.
Melino, G.
Melino, S.
de Melo, E.J.
Mena, M.A.
Meneghini, M.D.
Menendez, J.A.
Menezes, R.
Meng, L.
Meng, L.-.
Meng, S.
Menghini, R.
Menko, A.S.
Menna-Barreto, R.F.
Menon, M.B.
Meraz-Ríos, M.A.
Merla, G.
Merlini, L.
Merlot, A.M.
Meryk, A.
Meschini, S.
Meyer, J.N.
Mi, M.-.
Miao, C.-.
Micale, L.
Michaeli, S.
Michiels, C.
Migliaccio, A.R.
Mihailidou, A.S.
Mijaljica, D.
Mikoshiba, K.
Milan, E.
Miller-Fleming, L.
Mills, G.B.
Mills, I.G.
Minakaki, G.
Minassian, B.A.
Ming, X.-.
Minibayeva, F.
Minina, E.A.
Mintern, J.D.
Minucci, S.
Miranda-Vizuete, A.
Mitchell, C.H.
Miyamoto, S.
Miyazawa, K.
Mizushima, N.
Mnich, K.
Mograbi, B.
Mohseni, S.
Moita, L.F.
Molinari, M.
Molinari, M.
Møller, A.B.
Mollereau, B.
Mollinedo, F.
Mongillo, M.
Monick, M.M.
Montagnaro, S.
Montell, C.
Moore, D.J.
Moore, M.N.
Mora-Rodriguez, R.
Moreira, P.I.
Morel, E.
Morelli, M.B.
Moreno, S.
Morgan, M.J.
Moris, A.
Moriyasu, Y.
Morrison, J.L.
Morrison, L.A.
Morselli, E.
Moscat, J.
Moseley, P.L.
Mostowy, S.
Motori, E.
Mottet, D.
Mottram, J.C.
Moussa, C.E.
Mpakou, V.E.
Mukhtar, H.
Mulcahy Levy, J.M.
Muller, S.
Muñoz-Moreno, R.
Muñoz-Pinedo, C.
Münz, C.
Murphy, M.E.
Murray, J.T.
Murthy, A.
Mysorekar, I.U.
Nabi, I.R.
Nabissi, M.
Nader, G.A.
Nagahara, Y.
Nagai, Y.
Nagata, K.
Nagelkerke, A.
Nagy, P.
Naidu, S.R.
Nair, S.
Nakano, H.
Nakatogawa, H.
Nanjundan, M.
Napolitano, G.
Naqvi, N.I.
Nardacci, R.
Narendra, D.P.
Narita, M.
Nascimbeni, A.C.
Natarajan, R.
Navegantes, L.C.
Nawrocki, S.T.
Nazarko, T.Y.
Nazarko, V.Y.
Neill, T.
Neri, L.M.
Netea, M.G.
Netea-Maier, R.T.
Neves, B.M.
Ney, P.A.
Nezis, I.P.
Nguyen, H.T.
Nguyen, H.P.
Nicot, A.-.
Nilsen, H.
Nilsson, P.
Nishimura, M.
Nishino, I.
Niso-Santano, M.
Niu, H.
Nixon, R.A.
Njar, V.C.
Noda, T.
Noegel, A.A.
Nolte, E.M.
Norberg, E.
Norga, K.K.
Noureini, S.K.
Notomi, S.
Notterpek, L.
Nowikovsky, K.
Nukina, N.
Nürnberger, T.
O'Donnell, V.B.
O'Donovan, T.
O'Dwyer, P.J.
Oehme, I.
Oeste, C.L.
Ogawa, M.
Ogretmen, B.
Ogura, Y.
Oh, Y.J.
Ohmuraya, M.
Ohshima, T.
Ojha, R.
Okamoto, K.
Okazaki, T.
Oliver, F.J.
Ollinger, K.
Olsson, S.
Orban, D.P.
Ordonez, P.
Orhon, I.
Orosz, L.
O'Rourke, E.J.
Orozco, H.
Ortega, A.L.
Ortona, E.
Osellame, L.D.
Oshima, J.
Oshima, S.
Osiewacz, H.D.
Otomo, T.
Otsu, K.
Ou, J.-.
Outeiro, T.F.
Ouyang, D.-.
Ouyang, H.
Overholtzer, M.
Ozbun, M.A.
Ozdinler, P.H.
Ozpolat, B.
Pacelli, C.
Paganetti, P.
Page, G.
Pages, G.
Pagnini, U.
Pajak, B.
Pak, S.C.
Pakos-Zebrucka, K.
Pakpour, N.
Palková, Z.
Palladino, F.
Pallauf, K.
Pallet, N.
Palmieri, M.
Paludan, S.R.
Palumbo, C.
Palumbo, S.
Pampliega, O.
Pan, H.
Pan, W.
Panaretakis, T.
Pandey, A.
Pantazopoulou, A.
Papackova, Z.
Papademetrio, D.L.
Papassideri, I.
Papini, A.
Parajuli, N.
Pardo, J.
Parekh, V.V.
Parenti, G.
Park, J.-.
Park, J.
Park, O.K.
Parker, R.
Parlato, R.
Parys, J.B.
Parzych, K.R.
Pasquet, J.-.
Pasquier, B.
Pasumarthi, K.B.
Patschan, D.
Patterson, C.
Pattingre, S.
Pattison, S.
Pause, A.
Pavenstädt, H.
Pavone, F.
Pedrozo, Z.
Peña, F.J.
Peñalva, M.A.
Pende, M.
Peng, J.
Penna, F.
Penninger, J.M.
Pensalfini, A.
Pepe, S.
Pereira, G.J.
Pereira, P.C.
Pérez-de la Cruz, V.
Pérez-Pérez, M.E.
Pérez-Rodríguez, D.
Pérez-Sala, D.
Perier, C.
Perl, A.
Perlmutter, D.H.
Perrotta, I.
Pervaiz, S.
Pesonen, M.
Pessin, J.E.
Peters, G.J.
Petersen, M.
Petrache, I.
Petrof, B.J.
Petrovski, G.
Phang, J.M.
Piacentini, M.
Pierdominici, M.
Pierre, P.
Pierrefite-Carle, V.
Pietrocola, F.
Pimentel-Muiños, F.X.
Pinar, M.
Pineda, B.
Pinkas-Kramarski, R.
Pinti, M.
Pinton, P.
Piperdi, B.
Piret, J.M.
Platanias, L.C.
Platta, H.W.
Plowey, E.D.
Pöggeler, S.
Poirot, M.
Polčic, P.
Poletti, A.
Poon, A.H.
Popelka, H.
Popova, B.
Poprawa, I.
Poulose, S.M.
Poulton, J.
Powers, S.K.
Powers, T.
Pozuelo-Rubio, M.
Prak, K.
Prange, R.
Prescott, M.
Priault, M.
Prince, S.
Proia, R.L.
Proikas-Cezanne, T.
Prokisch, H.
Promponas, V.J.
Przyklenk, K.
Puertollano, R.
Pugazhenthi, S.
Puglielli, L.
Pujol, A.
Puyal, J.
Pyeon, D.
Qi, X.
Qian, W.-.
Qin, Z.-.
Qiu, Y.
Qu, Z.
Quadrilatero, J.
Quinn, F.
Raben, N.
Rabinowich, H.
Radogna, F.
Ragusa, M.J.
Rahmani, M.
Raina, K.
Ramanadham, S.
Ramesh, R.
Rami, A.
Randall-Demllo, S.
Randow, F.
Rao, H.
Rao, V.A.
Rasmussen, B.B.
Rasse, T.M.
Ratovitski, E.A.
Rautou, P.-.
Ray, S.K.
Razani, B.
Reed, B.H.
Reggiori, F.
Rehm, M.
Reichert, A.S.
Rein, T.
Reiner, D.J.
Reits, E.
Ren, J.
Ren, X.
Renna, M.
Reusch, J.E.
Revuelta, J.L.
Reyes, L.
Rezaie, A.R.
Richards, R.I.
Richardson, D.R.
Richetta, C.
Riehle, M.A.
Rihn, B.H.
Rikihisa, Y.
Riley, B.E.
Rimbach, G.
Rippo, M.R.
Ritis, K.
Rizzi, F.
Rizzo, E.
Roach, P.J.
Robbins, J.
Roberge, M.
Roca, G.
Roccheri, M.C.
Rocha, S.
Rodrigues, C.M.
Rodríguez, C.I.
de Cordoba, S.R.
Rodriguez-Muela, N.
Roelofs, J.
Rogov, V.V.
Rohn, T.T.
Rohrer, B.
Romanelli, D.
Romani, L.
Romano, P.S.
Roncero, M.I.
Rosa, J.L.
Rosello, A.
Rosen, K.V.
Rosenstiel, P.
Rost-Roszkowska, M.
Roth, K.A.
Roué, G.
Rouis, M.
Rouschop, K.M.
Ruan, D.T.
Ruano, D.
Rubinsztein, D.C.
Rucker, E.B.
Rudich, A.
Rudolf, E.
Rudolf, R.
Ruegg, M.A.
Ruiz-Roldan, C.
Ruparelia, A.A.
Rusmini, P.
Russ, D.W.
Russo, G.L.
Russo, G.
Russo, R.
Rusten, T.E.
Ryabovol, V.
Ryan, K.M.
Ryter, S.W.
Sabatini, D.M.
Sacher, M.
Sachse, C.
Sack, M.N.
Sadoshima, J.
Saftig, P.
Sagi-Eisenberg, R.
Sahni, S.
Saikumar, P.
Saito, T.
Saitoh, T.
Sakakura, K.
Sakoh-Nakatogawa, M.
Sakuraba, Y.
Salazar-Roa, M.
Salomoni, P.
Saluja, A.K.
Salvaterra, P.M.
Salvioli, R.
Samali, A.
Sanchez, A.M.
Sánchez-Alcázar, J.A.
Sanchez-Prieto, R.
Sandri, M.
Sanjuan, M.A.
Santaguida, S.
Santambrogio, L.
Santoni, G.
Dos Santos, C.N.
Saran, S.
Sardiello, M.
Sargent, G.
Sarkar, P.
Sarkar, S.
Sarrias, M.R.
Sarwal, M.M.
Sasakawa, C.
Sasaki, M.
Sass, M.
Sato, K.
Sato, M.
Satriano, J.
Savaraj, N.
Saveljeva, S.
Schaefer, L.
Schaible, U.E.
Scharl, M.
Schatzl, H.M.
Schekman, R.
Scheper, W.
Schiavi, A.
Schipper, H.M.
Schmeisser, H.
Schmidt, J.
Schmitz, I.
Schneider, B.E.
Schneider, E.M.
Schneider, J.L.
Schon, E.A.
Schönenberger, M.J.
Schönthal, A.H.
Schorderet, D.F.
Schröder, B.
Schuck, S.
Schulze, R.J.
Schwarten, M.
Schwarz, T.L.
Sciarretta, S.
Scotto, K.
Scovassi, A.I.
Screaton, R.A.
Screen, M.
Seca, H.
Sedej, S.
Segatori, L.
Segev, N.
Seglen, P.O.
Seguí-Simarro, J.M.
Segura-Aguilar, J.
Seki, E.
Sell, C.
Seiliez, I.
Semenkovich, C.F.
Semenza, G.L.
Sen, U.
Serra, A.L.
Serrano-Puebla, A.
Sesaki, H.
Setoguchi, T.
Settembre, C.
Shacka, J.J.
Shajahan-Haq, A.N.
Shapiro, I.M.
Sharma, S.
She, H.
Shen, C.-.
Shen, C.-.
Shen, H.-.
Shen, S.
Shen, W.
Sheng, R.
Sheng, X.
Sheng, Z.-.
Shepherd, T.G.
Shi, J.
Shi, Q.
Shi, Q.
Shi, Y.
Shibutani, S.
Shibuya, K.
Shidoji, Y.
Shieh, J.-.
Shih, C.-.
Shimada, Y.
Shimizu, S.
Shin, D.W.
Shinohara, M.L.
Shintani, M.
Shintani, T.
Shioi, T.
Shirabe, K.
Shiri-Sverdlov, R.
Shirihai, O.
Shore, G.C.
Shu, C.-.
Shukla, D.
Sibirny, A.A.
Sica, V.
Sigurdson, C.J.
Sigurdsson, E.M.
Sijwali, P.S.
Sikorska, B.
Silveira, W.A.
Silvente-Poirot, S.
Silverman, G.A.
Simak, J.
Simmet, T.
Simon, A.K.
Simon, H.-.
Simone, C.
Simons, M.
Simonsen, A.
Singh, R.
Singh, S.V.
Singh, S.K.
Sinha, D.
Sinha, S.
Sinicrope, F.A.
Sirko, A.
Sirohi, K.
Sishi, B.J.
Sittler, A.
Siu, P.M.
Sivridis, E.
Skwarska, A.
Slack, R.
Slaninová, I.
Slavov, N.
Smaili, S.S.
Smalley, K.S.
Smith, D.R.
Soenen, S.J.
Soleimanpour, S.A.
Solhaug, A.
Somasundaram, K.
Son, J.H.
Sonawane, A.
Song, C.
Song, F.
Song, H.K.
Song, J.-.
Song, W.
Soo, K.Y.
Sood, A.K.
Soong, T.W.
Soontornniyomkij, V.
Sorice, M.
Sotgia, F.
Soto-Pantoja, D.R.
Sotthibundhu, A.
Sousa, M.J.
Spaink, H.P.
Span, P.N.
Spang, A.
Sparks, J.D.
Speck, P.G.
Spector, S.A.
Spies, C.D.
Springer, W.
Clair, D.S.
Stacchiotti, A.
Staels, B.
Stang, M.T.
Starczynowski, D.T.
Starokadomskyy, P.
Steegborn, C.
Steele, J.W.
Stefanis, L.
Steffan, J.
Stellrecht, C.M.
Stenmark, H.
Stepkowski, T.M.
Stern, S.T.
Stevens, C.
Stockwell, B.R.
Stoka, V.
Storchova, Z.
Stork, B.
Stratoulias, V.
Stravopodis, D.J.
Strnad, P.
Strohecker, A.M.
Ström, A.-.
Stromhaug, P.
Stulik, J.
Su, Y.-.
Su, Z.
Subauste, C.S.
Subramaniam, S.
Sue, C.M.
Suh, S.W.
Sui, X.
Sukseree, S.
Sulzer, D.
Sun, F.-.
Sun, J.
Sun, J.
Sun, S.-.
Sun, Y.
Sun, Y.
Sun, Y.
Sundaramoorthy, V.
Sung, J.
Suzuki, H.
Suzuki, K.
Suzuki, N.
Suzuki, T.
Suzuki, Y.J.
Swanson, M.S.
Swanton, C.
Swärd, K.
Swarup, G.
Sweeney, S.T.
Sylvester, P.W.
Szatmari, Z.
Szegezdi, E.
Szlosarek, P.W.
Taegtmeyer, H.
Tafani, M.
Taillebourg, E.
Tait, S.W.
Takacs-Vellai, K.
Takahashi, Y.
Takáts, S.
Takemura, G.
Takigawa, N.
Talbot, N.J.
Tamagno, E.
Tamburini, J.
Tan, C.-.
Tan, L.
Tan, M.L.
Tan, M.
Tan, Y.-.
Tanaka, K.
Tanaka, M.
Tang, D.
Tang, D.
Tang, G.
Tanida, I.
Tanji, K.
Tannous, B.A.
Tapia, J.A.
Tasset-Cuevas, I.
Tatar, M.
Tavassoly, I.
Tavernarakis, N.
Taylor, A.
Taylor, G.S.
Taylor, G.A.
Taylor, J.P.
Taylor, M.J.
Tchetina, E.V.
Tee, A.R.
Teixeira-Clerc, F.
Telang, S.
Tencomnao, T.
Teng, B.-.
Teng, R.-.
Terro, F.
Tettamanti, G.
Theiss, A.L.
Theron, A.E.
Thomas, K.J.
Thomé, M.P.
Thomes, P.G.
Thorburn, A.
Thorner, J.
Thum, T.
Thumm, M.
Thurston, T.L.
Tian, L.
Till, A.
Ting, J.P.
Titorenko, V.I.
Toker, L.
Toldo, S.
Tooze, S.A.
Topisirovic, I.
Torgersen, M.L.
Torosantucci, L.
Torriglia, A.
Torrisi, M.R.
Tournier, C.
Towns, R.
Trajkovic, V.
Travassos, L.H.
Triola, G.
Tripathi, D.N.
Trisciuoglio, D.
Troncoso, R.
Trougakos, I.P.
Truttmann, A.C.
Tsai, K.-.
Tschan, M.P.
Tseng, Y.-.
Tsukuba, T.
Tsung, A.
Tsvetkov, A.S.
Tu, S.
Tuan, H.-.
Tucci, M.
Tumbarello, D.A.
Turk, B.
Turk, V.
Turner, R.F.
Tveita, A.A.
Tyagi, S.C.
Ubukata, M.
Uchiyama, Y.
Udelnow, A.
Ueno, T.
Umekawa, M.
Umemiya-Shirafuji, R.
Underwood, B.R.
Ungermann, C.
Ureshino, R.P.
Ushioda, R.
Uversky, V.N.
Uzcátegui, N.L.
Vaccari, T.
Vaccaro, M.I.
Váchová, L.
Vakifahmetoglu-Norberg, H.
Valdor, R.
Valente, E.M.
Vallette, F.
Valverde, A.M.
Van den Berghe, G.
Van Den Bosch, L.
van den Brink, G.R.
van der Goot, F.G.
van der Klei, I.J.
van der Laan, L.J.
van Doorn, W.G.
van Egmond, M.
van Golen, K.L.
Van Kaer, L.
van Lookeren Campagne, M.
Vandenabeele, P.
Vandenberghe, W.
Vanhorebeek, I.
Varela-Nieto, I.
Vasconcelos, M.H.
Vasko, R.
Vavvas, D.G.
Vega-Naredo, I.
Velasco, G.
Velentzas, A.D.
Velentzas, P.D.
Vellai, T.
Vellenga, E.
Vendelbo, M.H.
Venkatachalam, K.
Ventura, N.
Ventura, S.
Veras, P.S.
Verdier, M.
Vertessy, B.G.
Viale, A.
Vidal, M.
Vieira, H.L.
Vierstra, R.D.
Vigneswaran, N.
Vij, N.
Vila, M.
Villar, M.
Villar, V.H.
Villarroya, J.
Vindis, C.
Viola, G.
Viscomi, M.T.
Vitale, G.
Vogl, D.T.
Voitsekhovskaja, O.V.
von Haefen, C.
von Schwarzenberg, K.
Voth, D.E.
Vouret-Craviari, V.
Vuori, K.
Vyas, J.M.
Waeber, C.
Walker, C.L.
Walker, M.J.
Walter, J.
Wan, L.
Wan, X.
Wang, B.
Wang, C.
Wang, C.-.
Wang, C.
Wang, C.
Wang, C.
Wang, D.
Wang, F.
Wang, F.
Wang, G.
Wang, H.-.
Wang, H.
Wang, H.-.
Wang, H.
Wang, H.-.
Wang, J.
Wang, J.
Wang, M.
Wang, M.-.
Wang, P.-.
Wang, P.
Wang, R.C.
Wang, S.
Wang, T.-.
Wang, X.
Wang, X.-.
Wang, X.-.
Wang, X.
Wang, X.
Wang, Y.
Wang, Y.
Wang, Y.
Wang, Y.-.
Wang, Y.
Wang, Y.
Wang, Y.T.
Wang, Y.
Wang, Z.-.
Wappner, P.
Ward, C.
Ward, D.M.
Warnes, G.
Watada, H.
Watanabe, Y.
Watase, K.
Weaver, T.E.
Weekes, C.D.
Wei, J.
Weide, T.
Weihl, C.C.
Weindl, G.
Weis, S.N.
Wen, L.
Wen, X.
Wen, Y.
Westermann, B.
Weyand, C.M.
White, A.R.
White, E.
Whitton, J.L.
Whitworth, A.J.
Wiels, J.
Wild, F.
Wildenberg, M.E.
Wileman, T.
Wilkinson, D.S.
Wilkinson, S.
Willbold, D.
Williams, C.
Williams, K.
Williamson, P.R.
Winklhofer, K.F.
Witkin, S.S.
Wohlgemuth, S.E.
Wollert, T.
Wolvetang, E.J.
Wong, E.
Wong, G.W.
Wong, R.W.
Wong, V.K.
Woodcock, E.A.
Wright, K.L.
Wu, C.
Wu, D.
Wu, G.S.
Wu, J.
Wu, J.
Wu, M.
Wu, M.
Wu, S.
Wu, W.K.
Wu, Y.
Wu, Z.
Xavier, C.P.
Xavier, R.J.
Xia, G.-.
Xia, T.
Xia, W.
Xia, Y.
Xiao, H.
Xiao, J.
Xiao, S.
Xiao, W.
Xie, C.-.
Xie, Z.
Xie, Z.
Xilouri, M.
Xiong, Y.
Xu, C.
Xu, C.
Xu, F.
Xu, H.
Xu, H.
Xu, J.
Xu, J.
Xu, J.
Xu, L.
Xu, X.
Xu, Y.
Xu, Y.
Xu, Z.-.
Xu, Z.
Xue, Y.
Yamada, T.
Yamamoto, A.
Yamanaka, K.
Yamashina, S.
Yamashiro, S.
Yan, B.
Yan, B.
Yan, X.
Yan, Z.
Yanagi, Y.
Yang, D.-.
Yang, J.-.
Yang, L.
Yang, M.
Yang, P.-.
Yang, P.
Yang, Q.
Yang, W.
Yang, W.Y.
Yang, X.
Yang, Y.
Yang, Y.
Yang, Z.
Yang, Z.
Yao, M.-.
Yao, P.J.
Yao, X.
Yao, Z.
Yao, Z.
Yasui, L.S.
Ye, M.
Yedvobnick, B.
Yeganeh, B.
Yeh, E.S.
Yeyati, P.L.
Yi, F.
Yi, L.
Yin, X.-.
Yip, C.K.
Yoo, Y.-.
Yoo, Y.H.
Yoon, S.-.
Yoshida, K.-.
Yoshimori, T.
Young, K.H.
Yu, H.
Yu, J.J.
Yu, J.-.
Yu, J.
Yu, L.
Yu, W.H.
Yu, X.-.
Yu, Z.
Yuan, J.
Yuan, Z.-.
Yue, B.Y.
Yue, J.
Yue, Z.
Zacks, D.N.
Zacksenhaus, E.
Zaffaroni, N.
Zaglia, T.
Zakeri, Z.
Zecchini, V.
Zeng, J.
Zeng, M.
Zeng, Q.
Zervos, A.S.
Zhang, D.D.
Zhang, F.
Zhang, G.
Zhang, G.-.
Zhang, H.
Zhang, H.
Zhang, H.
Zhang, H.
Zhang, J.
Zhang, J.
Zhang, J.
Zhang, J.
Zhang, J.-.
Zhang, L.
Zhang, L.
Zhang, L.
Zhang, L.
Zhang, M.-.
Zhang, X.
Zhang, X.D.
Zhang, Y.
Zhang, Y.
Zhang, Y.
Zhang, Y.
Zhang, Y.
Zhao, M.
Zhao, W.-.
Zhao, X.
Zhao, Y.G.
Zhao, Y.
Zhao, Y.
Zhao, Y.-.
Zhao, Z.
Zhao, Z.J.
Zheng, D.
Zheng, X.-.
Zheng, X.
Zhivotovsky, B.
Zhong, Q.
Zhou, G.-.
Zhou, G.
Zhou, H.
Zhou, S.-.
Zhou, X.-.
Zhu, H.
Zhu, H.
Zhu, W.-.
Zhu, W.
Zhu, X.-.
Zhu, Y.
Zhuang, S.-.
Zhuang, X.
Ziparo, E.
Zois, C.E.
Zoladek, T.
Zong, W.-.
Zorzano, A.
Zughaier, S.M.
(2016). Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy,
Vol.12
(1),
pp. 1-222.
Jamin, Y.
Boult, J.K.
Li, J.
Popov, S.
Garteiser, P.
Ulloa, J.L.
Cummings, C.
Box, G.
Eccles, S.A.
Jones, C.
Waterton, J.C.
Bamber, J.C.
Sinkus, R.
Robinson, S.P.
(2015). Exploring the biomechanical properties of brain malignancies and their pathologic determinants in vivo with magnetic resonance elastography. Cancer res,
Vol.75
(7),
pp. 1216-1224.
show abstract
Malignant tumors are typically associated with altered rigidity relative to normal host tissue. Magnetic resonance elastography (MRE) enables the noninvasive quantitation of the mechanical properties of deep-seated tissue following application of an external vibrational mechanical stress to that tissue. In this preclinical study, we used MRE to quantify (kPa) the elasticity modulus Gd and viscosity modulus Gl of three intracranially implanted glioma and breast metastatic tumor models. In all these brain tumors, we found a notable softness characterized by lower elasticity and viscosity than normal brain parenchyma, enabling their detection on Gd and Gl parametric maps. The most circumscribed tumor (U-87 MG glioma) was the stiffest, whereas the most infiltrative tumor (MDA-MB-231 metastatic breast carcinoma) was the softest. Tumor cell density and microvessel density correlated significantly and positively with elasticity and viscosity, whereas there was no association with the extent of collagen deposition or myelin fiber entrapment. In conclusion, although malignant tumors tend to exhibit increased rigidity, intracranial tumors presented as remarkably softer than normal brain parenchyma. Our findings reinforce the case for MRE use in diagnosing and staging brain malignancies, based on the association of different tumor phenotypes with different mechanical properties..
Perryman, L.
Hoye, A.
Cox, T.
Lukram, B.
Jones, C.
Erler, J.
(2015). LOX expression is associated with infiltrative growth in glioblastoma. Clinical & experimental metastasis,
Vol.32
(3),
pp. 229-229.
Erice, O.
Smith, M.P.
White, R.
Goicoechea, I.
Barriuso, J.
Jones, C.
Margison, G.P.
Acosta, J.C.
Wellbrock, C.
Arozarena, I.
(2015). MGMT Expression Predicts PARP-Mediated Resistance to Temozolomide. Mol cancer ther,
Vol.14
(5),
pp. 1236-1246.
show abstract
Melanoma and other solid cancers are frequently resistant to chemotherapies based on DNA alkylating agents such as dacarbazine and temozolomide. As a consequence, clinical responses are generally poor. Such resistance is partly due to the ability of cancer cells to use a variety of DNA repair enzymes to maintain cell viability. Particularly, the expression of MGMT has been linked to temozolomide resistance, but cotargeting MGMT has proven difficult due to dose-limiting toxicities. Here, we show that the MGMT-mediated resistance of cancer cells is profoundly dependent on the DNA repair enzyme PARP. Both in vitro and in vivo, we observe that MGMT-positive cancer cells strongly respond to the combination of temozolomide and PARP inhibitors (PARPi), whereas MGMT-deficient cells do not. In melanoma cells, temozolomide induced an antiproliferative senescent response, which was greatly enhanced by PARPi in MGMT-positive cells. In summary, we provide compelling evidence to suggest that the stratification of patients with cancer upon the MGMT status would enhance the success of combination treatments using temozolomide and PARPi..
Trabelsi, S.
Mama, N.
Ladib, M.
Popov, S.
Burford, A.
Mokni, M.
Tlili, K.
Krifa, H.
Varella-Garcia, M.
Jones, C.
Tahar Yacoubi, M.
Saad, A.
H'mida Ben Brahim, D.
(2015). Adult recurrent pilocytic astrocytoma: Clinical, histopathological and molecular study. Neurochirurgie,
Vol.61
(6),
pp. 392-397.
show abstract
BACKGROUND: PA is a grade I glial tumor that mostly occurs in children. However, although apparently similar to paediatric PA, adult PA presents a different clinical follow-up that could arise from specific molecular alterations. A variety of genetic alterations have been identified as diagnostic or prognostic glioma molecular markers. MATERIAL AND METHODS: We describe a right infratentorial tumor that occurred in a 58-year-old man. Neuroimaging and neuropathological examination suggested PA as an initial diagnosis. The tumor was completely resected. Unexpectedly, two years later, a rapidly growing tumor on the operative site was observed with a second location in the pineal region. Immunohistochemical reactions (IHC), Multiplex ligation probe amplification (MLPA) and fluorescence in situ hybridization (FISH) was performed in both primary and relapse tumor. RESULTS: Neuroimaging and neuropathological examinations suggested an unusual diagnosis for adult patients: a recurrent PA. Both MLPA and FISH analysis contribute to diagnostic confirmation by KIAA1549: BRAF fusion detection. Additional genetic results revealed interesting findings that justified the tumor aggressivity. CONCLUSION: Molecular analysis of adult PA cases should be routinely combined with histopathological and neuroimaging examination to further refine prognostic diagnoses..
Castel, D.
Philippe, C.
Calmon, R.
Le Dret, L.
Truffaux, N.
Boddaert, N.
Pagès, M.
Taylor, K.R.
Saulnier, P.
Lacroix, L.
Mackay, A.
Jones, C.
Sainte-Rose, C.
Blauwblomme, T.
Andreiuolo, F.
Puget, S.
Grill, J.
Varlet, P.
Debily, M.-.
(2015). Histone H3F3A and HIST1H3B K27M mutations define two subgroups of diffuse intrinsic pontine gliomas with different prognosis and phenotypes. Acta neuropathol,
Vol.130
(6),
pp. 815-827.
show abstract
Diffuse intrinsic pontine glioma (DIPG) is the most severe paediatric solid tumour, with no significant therapeutic progress made in the past 50 years. Recent studies suggest that diffuse midline glioma, H3-K27M mutant, may comprise more than one biological entity. The aim of the study was to determine the clinical and biological variables that most impact their prognosis. Ninety-one patients with classically defined DIPG underwent a systematic stereotactic biopsy and were included in this observational retrospective study. Histone H3 genes mutations were assessed by immunochemistry and direct sequencing, whilst global gene expression profiling and chromosomal imbalances were determined by microarrays. A full description of the MRI findings at diagnosis and at relapse was integrated with the molecular profiling data and clinical outcome. All DIPG but one were found to harbour either a somatic H3-K27M mutation and/or loss of H3K27 trimethylation. We also discovered a novel K27M mutation in HIST2H3C, and a lysine-to-isoleucine substitution (K27I) in H3F3A, also creating a loss of trimethylation. Patients with tumours harbouring a K27M mutation in H3.3 (H3F3A) did not respond clinically to radiotherapy as well, relapsed significantly earlier and exhibited more metastatic recurrences than those in H3.1 (HIST1H3B/C). H3.3-K27M-mutated DIPG have a proneural/oligodendroglial phenotype and a pro-metastatic gene expression signature with PDGFRA activation, while H3.1-K27M-mutated tumours exhibit a mesenchymal/astrocytic phenotype and a pro-angiogenic/hypoxic signature supported by expression profiling and radiological findings. H3K27 alterations appear as the founding event in DIPG and the mutations in the two main histone H3 variants drive two distinct oncogenic programmes with potential specific therapeutic targets..
Bidinotto, L.T.
Scapulatempo-Neto, C.
Mackay, A.
de Almeida, G.C.
Scheithauer, B.W.
Berardinelli, G.N.
Torrieri, R.
Clara, C.A.
Feltrin, L.T.
Viana-Pereira, M.
Varella-Garcia, M.
Jones, C.
Reis, R.M.
(2015). Molecular Profiling of a Rare Rosette-Forming Glioneuronal Tumor Arising in the Spinal Cord. Plos one,
Vol.10
(9),
p. e0137690.
show abstract
Rosette-forming glioneuronal tumor (RGNT) of the IV ventricle is a rare and recently recognized brain tumor entity. It is histologically composed by two distinct features: a glial component, resembling pilocytic astrocytoma, and a component forming neurocytic rosettes and/or perivascular rosettes. Herein, we describe a 33-year-old man with RGNT arising in the spinal cord. Following an immunohistochemistry validation, we further performed an extensive genomic analysis, using array-CGH (aCGH), whole exome and cancer-related hotspot sequencing, in order to better understand its underlying biology. We observed the loss of 1p and gain of 1q, as well as gain of the whole chromosomes 7, 9 and 16. Local amplifications in 9q34.2 and 19p13.3 (encompassing the gene SBNO2) were identified. Moreover, we observed focal gains/losses in several chromosomes. Additionally, on chromosome 7, we identified the presence of the KIAA1549:BRAF gene fusion, which was further validated by RT-PCR and FISH. Across all mutational analyses, we detected and validated the somatic mutations of the genes MLL2, CNNM3, PCDHGC4 and SCN1A. Our comprehensive molecular profiling of this RGNT suggests that MAPK pathway and methylome changes, driven by KIAA1549:BRAF fusion and MLL2 mutation, respectively, could be associated with the development of this rare tumor entity..
Grill, J.
Hargrave, D.
Varlet, P.
Jaspan, T.
Jones, C.
Massimino, M.
Cañete, A.
Azizi, A.A.
Le Deley, M.
Saran, F.
Morgan, P.
Zahlmann, G.
Zheng, M.
Fuerst-Recktenwald, S.
Berger, C.
Bouffet, E.
Vassal, G.
(2014). 440TiPTHE HERBY STUDY: A PHASE 2 OPEN-LABEL, RANDOMIZED, MULTICENTER STUDY OF BEVACIZUMAB-BASED THERAPY IN PEDIATRIC PATIENTS WITH NEWLY DIAGNOSED HIGH-GRADE GLIOMA. Ann oncol,
Vol.25
(suppl_4),
p. iv145.
Taylor, K.R.
Vinci, M.
Bullock, A.N.
Jones, C.
(2014). ACVR1 Mutations in DIPG: Lessons Learned from FOP. Cancer research,
Vol.74
(17),
pp. 4565-4570.
Sturm, D.
Bender, S.
Jones, D.T.
Lichter, P.
Grill, J.
Becher, O.
Hawkins, C.
Majewski, J.
Jones, C.
Costello, J.F.
Iavarone, A.
Aldape, K.
Brennan, C.W.
Jabado, N.
Pfister, S.M.
(2014). Paediatric and adult glioblastoma: multiform (epi)genomic culprits emerge. Nat rev cancer,
Vol.14
(2),
pp. 92-107.
show abstract
full text
We have extended our understanding of the molecular biology that underlies adult glioblastoma over many years. By contrast, high-grade gliomas in children and adolescents have remained a relatively under-investigated disease. The latest large-scale genomic and epigenomic profiling studies have yielded an unprecedented abundance of novel data and provided deeper insights into gliomagenesis across all age groups, which has highlighted key distinctions but also some commonalities. As we are on the verge of dissecting glioblastomas into meaningful biological subgroups, this Review summarizes the hallmark genetic alterations that are associated with distinct epigenetic features and patient characteristics in both paediatric and adult disease, and examines the complex interplay between the glioblastoma genome and epigenome..
Wu, G.
Diaz, A.K.
Paugh, B.S.
Rankin, S.L.
Ju, B.
Li, Y.
Zhu, X.
Qui, C.
Chen, X.
Zhang, J.
Easton, J.
Edmonson, M.
Ma, X.
Lu, C.
Nagahawatte, P.
Hedlund, E.
Rusch, M.
Pounds, S.
Lin, T.
Onar-Thomas, A.
Huether, R.
Kriwacki, R.
Parker, M.
Gupta, P.
Becksfort, J.
Wei, L.
Mulder, H.L.
Boggs, K.
Vadodaria, B.
Yergeau, D.
Russell, J.C.
Ochoa, K.I.
Fulton, R.S.
Fulton, L.L.
Jones, C.
Boop, F.A.
Broniscer, A.
Wetmore, C.
Gajjar, A.
Ding, L.
Mardis, E.R.
Wilson, R.K.
Taylor, M.R.
Downing, J.R.
Ellison, D.W.
Zhang, J.
Baker, S.J.
Hosp, S.J.
Genome, W.U.
(2014). The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma. Nature genetics,
Vol.46
(5),
pp. 444-450.
full text
Buczkowicz, P.
Hoeman, C.
Rakopoulos, P.
Pajovic, S.
Letourneau, L.
Dzamba, M.
Morrison, A.
Lewis, P.
Bouffet, E.
Bartels, U.
Zuccaro, J.
Agnihotri, S.
Rya, S.
Barszczyk, M.
Chornenkyy, Y.
Bourgey, M.
Bourque, G.
Montpetit, A.
Cordero, F.
Castelo-Branco, P.
Mangere, J.
Tabori, U.
Ching, K.
Huang, A.
Taylor, K.R.
Mackay, A.
Bendell, A.E.
Nazarian, J.
Fangusaro, J.R.
Karajannis, M.A.
Zagzag, D.
Foreman, N.K.
Donson, A.
Hegert, J.V.
Smith, A.
Chan, J.
Lafay-Cousin, L.
Dunn, S.
Hukin, J.
Dunham, C.
Scheinemann, K.
Michaud, J.
Zelcer, S.
Ramsay, D.
Cain, J.
Brennan, C.
Souweidane, M.M.
Jones, C.
Allis, C.D.
Brudno, M.
Becher, O.
Hawkins, C.
(2014). Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations. Nature genetics,
Vol.46
(5),
pp. 451-456.
full text
Taylor, K.R.
Mackay, A.
Truffaux, N.
Butterfield, Y.S.
Morozova, O.
Philippe, C.
Castel, D.
Grasso, C.S.
Vinci, M.
Carvalho, D.
Carcaboso, A.M.
de Torres, C.
Cruz, O.
Mora, J.
Entz-Werle, N.
Ingram, W.J.
Monje, M.
Hargrave, D.
Bullock, A.N.
Puget, S.
Yip, S.
Jones, C.
Grill, J.
(2014). Recurrent activating ACVR1 mutations in diffuse intrinsic pontine glioma. Nature genetics,
Vol.46
(5),
pp. 457-461.
full text
Spence, T.
Sin-Chan, P.
Picard, D.
Barszczyk, M.
Hoss, K.
Lu, M.
Kim, S.-.
Ra, Y.-.
Nakamura, H.
Fangusaro, J.
Hwang, E.
Kiehna, E.
Toledano, H.
Wang, Y.
Shi, Q.
Johnston, D.
Michaud, J.
La Spina, M.
Buccoliero, A.M.
Adamek, D.
Camelo-Piragua, S.
Collins, V.P.
Jones, C.
Kabbara, N.
Jurdi, N.
Varlet, P.
Perry, A.
Scharnhorst, D.
Fan, X.
Muraszko, K.M.
Eberhart, C.G.
Ng, H.-.
Gururangan, S.
Van Meter, T.
Remke, M.
Lafay-Cousin, L.
Chan, J.A.
Sirachainan, N.
Pomeroy, S.L.
Clifford, S.C.
Gajjar, A.
Shago, M.
Halliday, W.
Taylor, M.D.
Grundy, R.
Lau, C.C.
Phillips, J.
Bouffet, E.
Dirks, P.B.
Hawkins, C.E.
Huang, A.
(2014). CNS-PNETs with C19MC amplification and/or LIN28 expression comprise a distinct histogenetic diagnostic and therapeutic entity. Acta neuropathologica,
Vol.128
(2),
pp. 291-303.
Jones, C.
Baker, S.J.
(2014). Unique genetic and epigenetic mechanisms driving paediatric diffuse high-grade glioma. Nat rev cancer,
Vol.14
(10).
show abstract
full text
Diffuse high-grade gliomas (HGGs) of childhood are a devastating spectrum of disease with no effective cures. The two-year survival for paediatric HGG ranges from 30%, for tumours arising in the cerebral cortex, to less than 10% for diffuse intrinsic pontine gliomas (DIPGs), which arise in the brainstem. Recent genome-wide studies provided abundant evidence that unique selective pressures drive HGG in children compared to adults, identifying novel oncogenic mutations connecting tumorigenesis and chromatin regulation, as well as developmental signalling pathways. These new genetic findings give insights into disease pathogenesis and the challenges and opportunities for improving patient survival in these mostly incurable childhood brain tumours..
Agnihotri, S.
Burrell, K.
Buczkowicz, P.
Remke, M.
Golbourn, B.
Chornenkyy, Y.
Gajadhar, A.
Fernandez, N.A.
Clarke, I.D.
Barszczyk, M.S.
Pajovic, S.
Ternamian, C.
Head, R.
Sabha, N.
Sobol, R.W.
Taylor, M.D.
Rutka, J.T.
Jones, C.
Dirks, P.B.
Zadeh, G.
Hawkins, C.
(2014). ATM Regulates 3-Methylpurine-DNA Glycosylase and Promotes Therapeutic Resistance to Alkylating Agents. Cancer discovery,
Vol.4
(10),
pp. 1198-1213.
Pereira, M.S.
Pinto, F.
Pardal, F.
Amorim, J.
Pires, M.
Pinheiro, C.
Lopes, J.M.
Pimentel, J.
Jones, C.
Viana-Pereira, M.
Reis, R.
(2014). UNRAVELING THE BIOLOGICAL ROLE OF SPINT2 IN GLIOMAS. Pediatric blood & cancer,
Vol.61,
pp. S182-S182.
Grill, J.
Varlet, P.
Jaspan, T.
Hargrave, D.R.
Massimino, M.
Bouffet, E.
Canete, A.
Azizi, A.A.
Saran, F.
Vassal, G.
Therapies, E.I.
Oncology, E.S.
Trials, A.C.
(2014). THE HERBY STUDY: A PHASE II OPEN LABEL, RANDOMIZED, MULTICENTER STUDY OF BEVACIZUMAB-BASED THERAPY IN PEDIATRIC PATIENTS WITH NEWLY DIAGNOSED HIGH-GRADE GLIOMA (HGG). Pediatric blood & cancer,
Vol.61,
pp. S280-S280.
Carvalho, D.
Mackay, A.
Bjerke, L.
Grundy, R.G.
Lopes, C.
Reis, R.M.
Jones, C.
(2014). The prognostic role of intragenic copy number breakpoints and identification of novel fusion genes in paediatric high grade glioma. Acta neuropathol commun,
Vol.2,
p. 23.
show abstract
BACKGROUND: Paediatric high grade glioma (pHGG) is a distinct biological entity to histologically similar tumours arising in older adults, and has differing copy number profiles and driver genetic alterations. As functionally important intragenic copy number aberrations (iCNA) and fusion genes begin to be identified in adult HGG, the same has not yet been done in the childhood setting. We applied an iCNA algorithm to our previously published dataset of DNA copy number profiling in pHGG with a view to identify novel intragenic breakpoints. RESULTS: We report a series of 288 iCNA events in pHGG, with the presence of intragenic breakpoints itself a negative prognostic factor. We identified an increased number of iCNA in older children compared to infants, and increased iCNA in H3F3A K27M mutant tumours compared to G34R/V and wild-type. We observed numerous gene disruptions by iCNA due to both deletions and amplifications, targeting known HGG-associated genes such as RB1 and NF1, putative tumour suppressors such as FAF1 and KIDINS220, and novel candidates such as PTPRE and KCND2. We further identified two novel fusion genes in pHGG - CSGALNACT2:RET and the complex fusion DHX57:TMEM178:MAP4K3. The latter was sequence-validated and appears to be an activating event in pHGG. CONCLUSIONS: These data expand upon our understanding of the genomic events driving these tumours and represent novel targets for therapeutic intervention in these poor prognosis cancers of childhood..
Al-Saffar, N.M.
Marshall, L.V.
Jackson, L.E.
Balarajah, G.
Eykyn, T.R.
Agliano, A.
Clarke, P.A.
Jones, C.
Workman, P.
Pearson, A.D.
Leach, M.O.
(2014). Lactate and choline metabolites detected in vitro by nuclear magnetic resonance spectroscopy are potential metabolic biomarkers for PI3K inhibition in pediatric glioblastoma. Plos one,
Vol.9
(8),
p. e103835.
show abstract
The phosphoinositide 3-kinase (PI3K) pathway is believed to be of key importance in pediatric glioblastoma. Novel inhibitors of the PI3K pathway are being developed and are entering clinical trials. Our aim is to identify potential non-invasive biomarkers of PI3K signaling pathway inhibition in pediatric glioblastoma using in vitro nuclear magnetic resonance (NMR) spectroscopy, to aid identification of target inhibition and therapeutic response in early phase clinical trials of PI3K inhibitors in childhood cancer. Treatment of SF188 and KNS42 human pediatric glioblastoma cell lines with the dual pan-Class I PI3K/mTOR inhibitor PI-103, inhibited the PI3K signaling pathway and resulted in a decrease in phosphocholine (PC), total choline (tCho) and lactate levels (p<0.02) as detected by phosphorus (31P)- and proton (1H)-NMR. Similar changes were also detected using the pan-Class I PI3K inhibitor GDC-0941 which lacks significant mTOR activity and is entering Phase II clinical trials. In contrast, the DNA damaging agent temozolomide (TMZ), which is used as current frontline therapy in the treatment of glioblastoma postoperatively (in combination with radiotherapy), increased PC, glycerophosphocholine (GPC) and tCho levels (p<0.04). PI-103-induced NMR changes were associated with alterations in protein expression levels of regulatory enzymes involved in glucose and choline metabolism including GLUT1, HK2, LDHA and CHKA. Our results show that by using NMR we can detect distinct biomarkers following PI3K pathway inhibition compared to treatment with the DNA-damaging anti-cancer agent TMZ. This is the first study reporting that lactate and choline metabolites are potential non-invasive biomarkers for monitoring response to PI3K pathway inhibitors in pediatric glioblastoma..
Ajeawung, N.F.
Maltais, R.
Jones, C.
Poirier, D.
Kamnasaran, D.
(2013). Viability screen on pediatric low grade glioma cell lines unveils a novel anti-cancer drug of the steroid biosynthesis inhibitor family. Cancer letters,
Vol.330
(1),
pp. 96-105.
Moniz, S.
Martinho, O.
Pinto, F.
Sousa, B.
Loureiro, C.
Oliveira, M.J.
Moita, L.F.
Honavar, M.
Pinheiro, C.
Pires, M.
Lopes, J.M.
Jones, C.
Costello, J.F.
Paredes, J.
Reis, R.M.
Jordan, P.
(2013). Loss of WNK2 expression by promoter gene methylation occurs in adult gliomas and triggers Rac1-mediated tumour cell invasiveness. Human molecular genetics,
Vol.22
(1),
pp. 84-95.
Moreno, L.
Popov, S.
Jury, A.
Al Sarraj, S.
Jones, C.
Zacharoulis, S.
(2013). Role of platelet derived growth factor receptor (PDGFR) over-expression and angiogenesis in ependymoma. J neurooncol,
Vol.111
(2),
pp. 169-176.
show abstract
New molecularly targeted therapies are needed for childhood ependymoma. Angiogenesis and the PDGFR pathway could be potential therapeutic targets. This study aimed to screen ependymomas for the expression and clinicopathological correlates of angiogenic factors and potential therapeutic targets including VEGFR, endoglin (CD105), CD34, CD31, c-Kit, PDGFR-α and PDGFR-β. Immunohistochemistry for angiogenesis factors and PDGFR-α and β was performed in 24 archival tumor samples from children and adults treated for ependymoma at our institution. CD31 density, CD105 density and pericyte coverage index (PCI) were calculated. These findings were correlated with clinical outcome. VEGFR2 was overexpressed in tumor cells in only one out of 24 cases, but was found overexpressed in the vessels in 6 cases. PDGFR-α and β were found to be over-expressed in the ependymoma tumor cells in seven out of 24 cases (29.2 %). CD31 density, CD105 density and PCI did not correlate with expression of PDGFRs. Overexpression of PDGFR-α and β in tumor cells and overexpression of PDGFR-α in tumor endothelium had prognostic significance and this was maintained in multivariate analysis for overexpression of PDGFR-α in tumor cells (2 year progression free survival was 16.7 ± 15.2 for cases with overexpression of PDGFR-α in the tumor vs. 74.5 ± 15.2 for those with low/no expression, hazard ratio = 5.78, p = 0.04). A number of angiogenic factors are expressed in ependymoma tumor cells and tumor endothelium. Preliminary evidence suggests that the expression of PDGFRs could have a prognostic significance in ependymoma. This data suggests that PDGFRs should be further evaluated as targets using novel PDGFR inhibitors..
Bjerke, L.
Mackay, A.
Nandhabalan, M.
Burford, A.
Jury, A.
Popov, S.
Bax, D.A.
Carvalho, D.
Taylor, K.R.
Vinci, M.
Bajrami, I.
McGonnell, I.M.
Lord, C.J.
Reis, R.M.
Hargrave, D.
Ashworth, A.
Workman, P.
Jones, C.
(2013). Histone H3 3 Mutations Drive Pediatric Glioblastoma through Upregulation of MYCN. Cancer discovery,
Vol.3
(5),
pp. 512-519.
Bjerke, L.
Mackay, A.
Nandhabalan, M.
Burford, A.
Jury, A.
Popov, S.
Bax, D.A.
Carvalho, D.
Taylor, K.R.
Vinci, M.
Bajrami, I.
McGonnell, I.M.
Lord, C.J.
Reis, R.M.
Hargrave, D.
Ashworth, A.
Workman, P.
Jones, C.
(2013). Histone H3 3 mutations drive pediatric glioblastoma through upregulation of MYCN. Cancer discov,
Vol.3
(5),
pp. 512-519.
show abstract
UNLABELLED: Children and young adults with glioblastoma (GBM) have a median survival rate of only 12 to 15 months, and these GBMs are clinically and biologically distinct from histologically similar cancers in older adults. They are defined by highly specific mutations in the gene encoding the histone H3.3 variant H3F3A , occurring either at or close to key residues marked by methylation for regulation of transcription—K27 and G34. Here, we show that the cerebral hemisphere-specific G34 mutation drives a distinct expression signature through differential genomic binding of the K36 trimethylation mark (H3K36me3). The transcriptional program induced recapitulates that of the developing forebrain, and involves numerous markers of stem-cell maintenance, cell-fate decisions, and self-renewal.Critically, H3F3A G34 mutations cause profound upregulation of MYCN , a potent oncogene that is causative of GBMs when expressed in the correct developmental context. This driving aberration is selectively targetable in this patient population through inhibiting kinases responsible for stabilization of the protein. SIGNIFICANCE: We provide the mechanistic explanation for how the fi rst histone gene mutation inhuman disease biology acts to deliver MYCN, a potent tumorigenic initiator, into a stem-cell compartment of the developing forebrain, selectively giving rise to incurable cerebral hemispheric GBM. Using synthetic lethal approaches to these mutant tumor cells provides a rational way to develop novel and highly selective treatment strategies.
Galavotti, S.
Bartesaghi, S.
Faccenda, D.
Shaked-Rabi, M.
Sanzone, S.
McEvoy, A.
Dinsdale, D.
Condorelli, F.
Brandner, S.
Campanella, M.
Grose, R.
Jones, C.
Salomoni, P.
(2013). The autophagy-associated factors DRAM1 and p62 regulate cell migration and invasion in glioblastoma stem cells. Oncogene,
Vol.32
(6),
pp. 699-712.
Ajeawung, N.F.
Faure, R.
Jones, C.
Kamnasaran, D.
(2013). Preclinical evaluation of dipotassium bisperoxo (picolinato) oxovanadate V for the treatment of pediatric low-grade gliomas. Future oncology,
Vol.9
(8),
pp. 1215-1229.
Grill, J.
Hargrave, D.
Massimino, M.
Jaspan, T.
Varlet, P.
Jones, C.
Morgan, P.
Le Deley, M.C.
Azizi, A.
Canete, A.
Bouffet, E.
Saran, F.
Bachir, J.
Bubuteishvili-Pacaud, L.
Rousseau, R.
Vassal, G.
(2013). HERBY: A PHASE II COMPARATIVE STUDY OF BEVACIZUMAB-BASED THERAPY IN CHILDREN WITH HIGH-GRADE GLIOMA. Pediatric blood & cancer,
Vol.60,
pp. 205-205.
Xavier-Magalhães, A.
Nandhabalan, M.
Jones, C.
Costa, B.M.
(2013). Molecular prognostic factors in glioblastoma: state of the art and future challenges. Cns oncol,
Vol.2
(6),
pp. 495-510.
show abstract
full text
Gliomas account for the majority of primary tumors of the CNS, of which glioblastoma (GBM) is the most common and malignant, and for which survival is very poor. Despite significant inter- and intra-tumor heterogeneity, all patients are treated with a standardized therapeutic approach. While some clinical features of GBM patients have already been established as classic prognostic factors (e.g., patient age at diagnosis and Karnofsky performance status), one of the most important research fields in neuro-oncology today is the identification of novel molecular determinants of patient survival and tumor response to therapy. Here, we aim to review and discuss some of the most relevant and novel prognostic biomarkers in adult and pediatric GBM patients that may aid in stratifying subgroups of GBMs and rationalizing treatment decisions..
Bender, S.
Tang, Y.
Lindroth, A.M.
Hovestadt, V.
Jones, D.T.
Kool, M.
Zapatka, M.
Northcott, P.A.
Sturm, D.
Wang, W.
Radlwimmer, B.
Hojfeldt, J.W.
Truffaux, N.
Castel, D.
Schubert, S.
Ryzhova, M.
Seker-Cin, H.
Gronych, J.
Johann, P.D.
Stark, S.
Meyer, J.
Milde, T.
Schuhmann, M.
Ebinger, M.
Monoranu, C.-.
Ponnuswami, A.
Chen, S.
Jones, C.
Witt, O.
Collins, V.P.
von Deimling, A.
Jabado, N.
Puget, S.
Grill, J.
Helin, K.
Korshunov, A.
Lichter, P.
Monje, M.
Plass, C.
Cho, Y.-.
Pfister, S.M.
(2013). Reduced H3K27me3 and DNA Hypomethylation Are Major Drivers of Gene Expression in K27M Mutant Pediatric High-Grade Gliomas. Cancer cell,
Vol.24
(5),
pp. 660-672.
Laxton, R.C.
Popov, S.
Doey, L.
Jury, A.
Bhangoo, R.
Gullan, R.
Chandler, C.
Brazil, L.
Sadler, G.
Beaney, R.
Sibtain, N.
King, A.
Bodi, I.
Jones, C.
Ashkan, K.
Al-Sarraj, S.
(2013). Primary glioblastoma with oligodendroglial differentiation has better clinical outcome but no difference in common biological markers compared with other types of glioblastoma. Neuro oncol,
Vol.15
(12),
pp. 1635-1643.
show abstract
BACKGROUND: Glioblastoma multiforme with an oligodendroglial component (GBMO) has been recognized in the World Health Organization classification-however, the diagnostic criteria, molecular biology, and clinical outcome of primary GBMO remain unclear. Our aim was to investigate whether primary GBMO is a distinct clinicopathological subgroup of GBM and to determine the relative frequency of prognostic markers such as loss of heterozygosity (LOH) on 1p and/or 19q, O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation, and isocitrate dehydrogenase 1 (IDH1) mutation. METHODS: We examined 288 cases of primary GBM and assessed the molecular markers in 57 GBMO and 50 cases of other primary GBM, correlating the data with clinical parameters and outcome. RESULTS: GBMO comprised 21.5% of our GBM specimens and showed significantly longer survival compared with our other GBM (12 mo vs 5.8 mo, P = .006); there was also a strong correlation with younger age at diagnosis (56.4 y vs 60.6 y, P = .005). Singular LOH of 19q (P = .04) conferred a 1.9-fold increased hazard of shorter survival. There was no difference in the frequencies of 1p or 19q deletion, MGMT promoter methylation, or IDH1 mutation (P = .8, P = 1.0, P = 1.0, respectively). CONCLUSIONS: Primary GBMO is a subgroup of GBM associated with longer survival and a younger age group but shows no difference in the frequency of LOH of 1p/19q, MGMT, and IDH1 mutation compared with other primary GBM..
Paugh, B.S.
Zhu, X.
Qu, C.
Endersby, R.
Diaz, A.K.
Zhang, J.
Bax, D.A.
Carvalho, D.
Reis, R.M.
Onar-Thomas, A.
Broniscer, A.
Wetmore, C.
Zhang, J.
Jones, C.
Ellison, D.W.
Baker, S.J.
(2013). Novel Oncogenic PDGFRA Mutations in Pediatric High-Grade Gliomas. Cancer research,
Vol.73
(20),
pp. 6219-6229.
Burford, A.
Little, S.E.
Jury, A.
Popov, S.
Laxton, R.
Doey, L.
Al-Sarraj, S.
Jürgensmeier, J.M.
Jones, C.
(2013). Distinct phenotypic differences associated with differential amplification of receptor tyrosine kinase genes at 4q12 in glioblastoma. Plos one,
Vol.8
(8),
p. e71777.
show abstract
Gene amplification at chromosome 4q12 is a common alteration in human high grade gliomas including glioblastoma, a CNS tumour with consistently poor prognosis. This locus harbours the known oncogenes encoding the receptor tyrosine kinases PDGFRA, KIT, and VEGFR2. These receptors are potential targets for novel therapeutic intervention in these diseases, with expression noted in tumour cells and/or associated vasculature. Despite this, a detailed assessment of their relative contributions to different high grade glioma histologies and the underlying heterogeneity within glioblastoma has been lacking. We studied 342 primary high grade gliomas for individual gene amplification using specific FISH probes, as well as receptor expression in the tumour and endothelial cells by immunohistochemistry, and correlated our findings with specific tumour cell morphological types and patterns of vasculature. We identified amplicons which encompassed PDGFRA only, PDGFRA/KIT, and PDGFRA/KIT/VEGFR2, with distinct phenotypic correlates. Within glioblastoma specimens, PDGFRA amplification alone was linked to oligodendroglial, small cell and sarcomatous tumour cell morphologies, and rare MGMT promoter methylation. A younger age at diagnosis and better clinical outcome in glioblastoma patients is only seen when PDGFRA and KIT are co-amplified. IDH1 mutation was only found when all three genes are amplified; this is a subgroup which also harbours extensive MGMT promoter methylation. Whilst PDGFRA amplification was tightly linked to tumour expression of the receptor, this was not the case for KIT or VEGFR2. Thus we have identified differential patterns of gene amplification and expression of RTKs at the 4q12 locus to be associated with specific phenotypes which may reflect their distinct underlying mechanisms..
Popov, S.
Jury, A.
Laxton, R.
Doey, L.
Kandasamy, N.
Al-Sarraj, S.
Jürgensmeier, J.M.
Jones, C.
(2013). IDH1-associated primary glioblastoma in young adults displays differential patterns of tumour and vascular morphology. Plos one,
Vol.8
(2),
p. e56328.
show abstract
Glioblastoma is a highly aggressive tumour with marked heterogeneity at the morphological level in both the tumour cells and the associated highly prominent vasculature. As we begin to develop an increased biological insight into the underlying processes driving the disease, fewer attempts have thus far been made to understand these phenotypic differences. We sought to address this by carefully assessing the morphological characteristics of both the tumour cells and the associated vasculature, relating these observations to the IDH1/MGMT status, with a particular focus on the early onset population of young adults who develop primary glioblastoma. 276 primary glioblastoma specimens were classified into their predominant cell morphological type (fibrillary, gemistocytic, giant cell, small cell, oligodendroglial, sarcomatous), and assessed for specific tumour (cellularity, necrosis, palisades) and vascular features (glomeruloid structures, arcades, pericyte proliferation). IDH1 positive glioblastomas were associated with a younger age at diagnosis, better clinical outcome, prominent oligodendroglial and small cell tumour cell morphology, pallisading necrosis and glomeruloid vascular proliferation in the absence of arcade-like structures. These features widen the phenotype of IDH1 mutation-positive primary glioblastoma in young adults and provide correlative evidence for a functional role of mutant IDH1 in the differential nature of neo-angiogenesis in different subtypes of glioblastoma..
Little, S.E.
Popov, S.
Jury, A.
Bax, D.A.
Doey, L.
Al-Sarraj, S.
Jurgensmeier, J.M.
Jones, C.
(2012). Receptor tyrosine kinase genes amplified in glioblastoma exhibit a mutual exclusivity in variable proportions reflective of individual tumor heterogeneity. Cancer res,
Vol.72
(7),
pp. 1614-1620.
show abstract
Intratumoral heterogeneity in human solid tumors represents a major barrier for the development of effective molecular treatment strategies, as treatment efficacies will reflect the molecular variegation in individual tumors. In glioblastoma, the generation of composite genomic profiles from bulk tumor samples has allowed one to map the genomic amplifications of putative genetic drivers and to prioritize therapeutic targeting strategies aimed at eradicating the tumor burden. Notably, amplification of multiple receptor tyrosine kinases (RTK) within a single tumor specimen obtained from patients is frequently observed. In this study, use of a detailed multicolor FISH mapping procedure in pathologic specimens revealed a mutual exclusivity of gene amplification in the majority of glioblastoma tumors examined. In particular, the two most commonly amplified RTK genes, EGFR and PDGFRA, were found to be present in variable proportions across the tumors, with one or the other gene predominating in certain areas of the same specimen. Our findings have profound implications for designing efficacious therapeutic regimens, as it remains unclear that how the cells with different gene amplification events contribute to disease propagation or the response to molecular targeted therapies..
Fong, A.-.
Eykyn, T.R.
Parkes, H.G.
Bielen, A.
Jones, C.
Judson, I.R.
Griffiths, J.R.
Leach, M.O.
Chung, Y.-.
(2012). Picropodophyllin (PPP) increases glucose metabolism and lactate production in paediatric glioblastoma cells. Clinical cancer research,
Vol.18
(10).
Crocker, M.
Saadoun, S.
Jury, A.
Jones, C.
Zacharoulis, S.
Thomas, S.
Zwiggelaar, R.
Bridges, L.R.
Bell, B.A.
Papadopoulos, M.C.
(2012). Glioblastoma blood flow measured with stable xenon CT indicates tumor necrosis, vascularity, and brain invasion. Neuro oncol,
Vol.14
(5),
pp. 641-648.
show abstract
Tumor vasculature is a promising therapeutic target in glioblastoma. Imaging tumor blood flow may help assess the efficacy of anti-angiogenic treatments. We determined the clinical usefulness of stable xenon CT performed preoperatively in patients with glioblastoma. This is a prospective cohort study. We determined absolute tumor blood flow before surgery in 38 patients with glioblastoma using stable xenon CT. We also histologically examined tumor specimens obtained from surgery and quantified their vascularity (by CD31 and CD105 immunostain), necrosis (by hematoxylin and eosin stain), and the presence of neuronal processes (by neurofilament immunostain). According to the xenon CT blood flow map, there are 3 types of glioblastoma. Type I glioblastomas have unimodal high blood flow histograms; histologically there is little necrosis and vascular proliferation. Type II glioblastomas have unimodal low blood flow histograms; histologically there is prominent necrosis and vascular proliferation. We propose that in type II glioblastoma, the abnormal vessels induced by hypoxia are inefficient at promoting blood flow. Type III glioblastomas have multimodal blood flow histograms. Histologically there is significant neuronal tissue within the tumor. Patients with type III glioblastomas were more likely to develop a post-surgical deficit, consistent with the inclusion of normal tissue within the tumor. Preoperative measurement of absolute blood flow with stable xenon CT in patients with glioblastoma predicts key biological features of the tumor and may aid surgical planning..
Reid, A.H.
Attard, G.
Brewer, D.
Miranda, S.
Riisnaes, R.
Clark, J.
Hylands, L.
Merson, S.
Vergis, R.
Jameson, C.
Høyer, S.
Sørenson, K.D.
Borre, M.
Jones, C.
de Bono, J.S.
Cooper, C.S.
(2012). Novel, gross chromosomal alterations involving PTEN cooperate with allelic loss in prostate cancer. Mod pathol,
Vol.25
(6),
pp. 902-910.
show abstract
There is increasing evidence that multiple chromosomal rearrangements occur in prostate cancer. PTEN loss is considered to be a key event in prostate carcinogenesis but the mechanisms of loss remain to be fully elucidated. We hypothesised that gross rearrangements may exist that cause disruption of the PTEN gene in the absence of genomic deletion. We therefore designed a novel fluorescence in situ hybridisation (FISH) assay with probes overlying regions 3' and 5' of PTEN and a third probe overlying the gene. We aimed to identify both genomic deletions and gross rearrangements of PTEN that would be overlooked by previously reported single-probe FISH assays. We proceeded to evaluate a tissue microarray with radical prostatectomy and trans-urethral resection of the prostate specimens from 187 patients. We identified PTEN genomic loss in 45/150 (30%) radical prostatectomy patients and 16/37 (43%) trans-urethral resection of the prostate patients. Importantly, our assay detected novel chromosomal alterations in the PTEN gene (characterised by splitting of FISH signals) in 13 tumours (6.9% of all prostate cancers; 21% of PTEN-lost cancers). All PTEN-rearranged tumours had genomic loss at the other allele and had no expression of PTEN by immunohistochemistry. PTEN-rearranged tumours were significantly more likely to have an underlying ERG rearrangement. Our assay differentiated loss of the probe overlying PTEN in isolation or in combination with either one of or both the probes overlying the 3' and 5' regions. This gave an indication of the size of genomic loss and we observed considerable inter-tumoural heterogeneity in the extent of genomic loss in PTEN-lost tumours. In summary, gross rearrangements of the PTEN locus occur in prostate cancer and can be detected by a 'break-apart' FISH assay. This observation could explain the absence of PTEN protein expression in a subgroup of tumours previously classified as having heterozygous genomic loss using single-probe traditional FISH assays..
Lee, C.
Fotovati, A.
Triscott, J.
Chen, J.
Venugopal, C.
Singhal, A.
Dunham, C.
Kerr, J.M.
Verreault, M.
Yip, S.
Wakimoto, H.
Jones, C.
Jayanthan, A.
Narendran, A.
Singh, S.K.
Dunn, S.E.
(2012). Polo-Like Kinase 1 Inhibition Kills Glioblastoma Multiforme Brain Tumor Cells in Part Through Loss of SOX2 and Delays Tumor Progression in Mice. Stem cells,
Vol.30
(6),
pp. 1064-1075.
Picard, D.
Miller, S.
Hawkins, C.E.
Bouffet, E.
Rogers, H.A.
Chan, T.S.
Kim, S.-.
Ra, Y.-.
Fangusaro, J.
Korshunov, A.
Toledano, H.
Nakamura, H.
Hayden, J.T.
Chan, J.
Lafay-Cousin, L.
Hu, P.
Fan, X.
Muraszko, K.M.
Pomeroy, S.L.
Lau, C.C.
Ng, H.-.
Jones, C.
Van Meter, T.
Clifford, S.C.
Eberhart, C.
Gajjar, A.
Pfister, S.M.
Grundy, R.G.
Huang, A.
(2012). Markers of survival and metastatic potential in childhood CNS primitive neuro-ectodermal brain tumours: an integrative genomic analysis. Lancet oncology,
Vol.13
(8),
pp. 838-848.
full text
Bielen, A.
Box, G.
Perryman, L.
Bjerke, L.
Popov, S.
Jamin, Y.
Jury, A.
Valenti, M.
Brandon, A.D.
Martins, V.
Romanet, V.
Jeay, S.
Raynaud, F.I.
Hofmann, F.
Robinson, S.P.
Eccles, S.A.
Jones, C.
(2012). Dependence of Wilms tumor cells on signaling through insulin-like growth factor 1 in an orthotopic xenograft model targetable by specific receptor inhibition. Proc natl acad sci u s a,
Vol.109
(20),
pp. E1267-E1276.
show abstract
We have previously demonstrated an increased DNA copy number and expression of IGF1R to be associated with poor outcome in Wilms tumors. We have now tested whether inhibiting this receptor may be a useful therapeutic strategy by using a panel of Wilms tumor cell lines. Both genetic and pharmacological targeting resulted in inhibition of downstream signaling through PI3 and MAP kinases, G(1) cell cycle arrest, and cell death, with drug efficacy dependent on the levels of phosphorylated IGF1R. These effects were further associated with specific gene expression signatures reflecting pathway inhibition, and conferred synergistic chemosensitisation to doxorubicin and topotecan. In the in vivo setting, s.c. xenografts of WiT49 cells resembled malignant rhabdoid tumors rather than Wilms tumors. Treatment with an IGF1R inhibitor (NVP-AEW541) showed no discernable antitumor activity and no downstream pathway inactivation. By contrast, Wilms tumor cells established orthotopically within the kidney were histologically accurate and exhibited significantly elevated insulin-like growth factor-mediated signaling, and growth was significantly reduced on treatment with NVP-AEW541 in parallel with signaling pathway ablation. As a result of the paracrine effects of enhanced IGF2 expression in Wilms tumor, this disease may be acutely dependent on signaling through the IGF1 receptor, and thus treatment strategies aimed at its inhibition may be useful in the clinic. Such efficacy may be missed if only standard ectopic models are considered as a result of an imperfect recapitulation of the specific tumor microenvironment..
Little, S.
Jury, A.
Bax, D.
Doey, L.
Al-Sarraj, S.
Jurgensmeier, J.
Jones, C.
(2012). Intratumoral mutual exclusivity of dual amplified receptor tyrosine kinase genes in glioblastoma. Cancer research,
Vol.72.
Puget, S.
Philippe, C.
Bax, D.A.
Job, B.
Varlet, P.
Junier, M.-.
Andreiuolo, F.
Carvalho, D.
Reis, R.
Guerrini-Rousseau, L.
Roujeau, T.
Dessen, P.
Richon, C.
Lazar, V.
Le Teuff, G.
Sainte-Rose, C.
Geoerger, B.
Vassal, G.
Jones, C.
Grill, J.
(2012). Mesenchymal Transition and PDGFRA Amplification/Mutation Are Key Distinct Oncogenic Events in Pediatric Diffuse Intrinsic Pontine Gliomas. Plos one,
Vol.7
(2).
Jones, C.
Perryman, L.
Hargrave, D.
(2012). Paediatric and adult malignant glioma: close relatives or distant cousins?. Nat rev clin oncol,
Vol.9
(7),
pp. 400-413.
show abstract
Gliomas in children differ from their adult counterparts by their distribution of histological grade, site of presentation and rate of malignant transformation. Although rare in the paediatric population, patients with high-grade gliomas have, for the most part, a comparably dismal clinical outcome to older patients with morphologically similar lesions. Molecular profiling data have begun to reveal the major genetic alterations underpinning these malignant tumours in children. Indeed, the accumulation of large datasets on adult high-grade glioma has revealed key biological differences between the adult and paediatric disease. Furthermore, subclassifications within the childhood age group can be made depending on age at diagnosis and tumour site. However, challenges remain on how to reconcile clinical data from adult patients to tailor novel treatment strategies specifically for paediatric patients..
Puget, S.
Philippe, C.
Bax, D.A.
Job, B.
Varlet, P.
Junier, M.P.
Andreiuolo, F.
Jubert, C.
Opolon, P.
Carvalho, D.
Reis, R.M.
Guerrini-Rousseau, L.
Roujeau, T.
Dessen, P.
Richon, C.
Lazor, V.
Le Teuff, G.
Sainte-Rose, C.
Vassal, G.
Jones, C.
Geoerger, B.
Grill, J.
(2011). MESENCHYMAL TRANSITION AND PDGFRA AMPLIFICATION/MUTATION ARE KEY DISTINCT ONCOGENIC EVENTS IN PEDIATRIC DIFFUSE INTRINSIC PONTINE GLIOMAS. Pediatric blood & cancer,
Vol.57
(5),
pp. 806-2.
Bielen, A.
Perryman, L.
Box, G.M.
Valenti, M.
de Haven Brandon, A.
Martins, V.
Jury, A.
Popov, S.
Gowan, S.
Jeay, S.
Raynaud, F.I.
Hofmann, F.
Hargrave, D.
Eccles, S.A.
Jones, C.
(2011). Enhanced efficacy of IGF1R inhibition in pediatric glioblastoma by combinatorial targeting of PDGFRα/β. Mol cancer ther,
Vol.10
(8),
pp. 1407-1418.
show abstract
Pediatric glioblastoma (pGBM), although rare, is one of the leading causes of cancer-related deaths in children, with tumors essentially refractory to existing treatments. We have identified IGF1R to be a potential therapeutic target in pGBM due to gene amplification and high levels of IGF2 expression in some tumor samples, as well as constitutive receptor activation in pGBM cell lines. To evaluate the therapeutic potential of strategies targeting the receptor, we have carried out in vitro and in vivo preclinical studies using the specific IGF1R inhibitor NVP-AEW541. A modest inhibitory effect was seen in vitro, with GI(50) values of 5 to 6 μmol/L, and concurrent inhibition of receptor phosphorylation. Specific targeting of IGF1R with short interfering RNA decreased cell viability, diminished downstream signaling through phosphoinositide 3-kinase (PI3K), and induced G(1) arrest, effects mimicked by NVP-AEW541, both in the absence and presence of IGF2. Hallmarks of PI3K inhibition were observed after treatment with NVP-AEW541 by expression profiling and Western blot analysis. Phospho-receptor tyrosine kinase (RTK) arrays showed phosphorylation of platelet-derived growth factor receptor (PDGFR) α/β in pGBM cells, suggesting coactivation of an alternative RTK pathway. Treatment of KNS42 with the PDGFR inhibitor imatinib showed additional effects targeting the mitogen-activated protein kinase pathway, and cotreatment of the PDGFR inhibitor imatinib with NVP-AEW541 resulted in a highly synergistic interaction in vitro and increased efficacy after 14 days therapy in vivo compared with either agent alone. These data provide evidence that inhibition of IGF1R, in combination with other targeted agents, may be a useful and novel therapeutic strategy in pGBM..
Nicolaides, T.
Hariono, S.
Barkovich, K.
Hashizume, R.
Rowitch, D.
Weiss, W.
Sheer, D.
Baker, S.
Paugh, B.
Waldman, T.
Li, H.
Jones, C.
Forshew, T.
James, D.
(2011). TARGETED THERAPY FOR BRAFV600E MUTANT MALIGNANT ASTROCYTOMA. Neuro-oncology,
Vol.13,
pp. 112-112.
Williams, R.D.
Al-Saadi, R.
Natrajan, R.
Mackay, A.
Chagtai, T.
Little, S.
Hing, S.N.
Fenwick, K.
Ashworth, A.
Grundy, P.
Anderson, J.R.
Dome, J.S.
Perlman, E.J.
Jones, C.
Pritchard-Jones, K.
(2011). Molecular Profiling Reveals Frequent Gain of MYCN and Anaplasia-Specific Loss of 4q and 14q in Wilms Tumor. Genes chromosomes & cancer,
Vol.50
(12),
pp. 982-14.
Fotovati, A.
Abu-Ali, S.
Wang, P.-.
Deleyrolle, L.P.
Lee, C.
Triscott, J.
Chen, J.Y.
Franciosi, S.
Nakamura, Y.
Sugita, Y.
Uchiumi, T.
Kuwano, M.
Leavitt, B.R.
Singh, S.K.
Jury, A.
Jones, C.
Wakimoto, H.
Reynolds, B.A.
Pallen, C.J.
Dunn, S.E.
(2011). YB-1 bridges neural stem cells and brain tumor-initiating cells via its roles in differentiation and cell growth. Cancer res,
Vol.71
(16),
pp. 5569-5578.
show abstract
The Y-box binding protein 1 (YB-1) is upregulated in many human malignancies including glioblastoma (GBM). It is also essential for normal brain development, suggesting that YB-1 is part of a neural stem cell (NSC) network. Here, we show that YB-1 was highly expressed in the subventricular zone (SVZ) of mouse fetal brain tissues but not in terminally differentiated primary astrocytes. Conversely, YB-1 knockout mice had reduced Sox-2, nestin, and musashi-1 expression in the SVZ. Although primary murine neurospheres were rich in YB-1, its expression was lost during glial differentiation. Glial tumors often express NSC markers and tend to loose the cellular control that governs differentiation; therefore, we addressed whether YB-1 served a similar role in cancer cells. YB-1, Sox-2, musashi-1, Bmi-1, and nestin are coordinately expressed in SF188 cells and 9/9 GBM patient-derived primary brain tumor-initiating cells (BTIC). Silencing YB-1 with siRNA attenuated the expression of these NSC markers, reduced neurosphere growth, and triggered differentiation via coordinate loss of GSK3-β. Furthermore, differentiation of BTIC with 1% serum or bone morphogenetic protein-4 suppressed YB-1 protein expression. Likewise, YB-1 expression was lost during differentiation of normal human NSCs. Consistent with these observations, YB-1 expression increased with tumor grade (n = 49 cases). YB-1 was also coexpressed with Bmi-1 (Spearmans 0.80, P > 0.001) and Sox-2 (Spearmans 0.66, P > 0.001) based on the analysis of 282 cases of high-grade gliomas. These proteins were highly expressed in 10/15 (67%) of GBM patients that subsequently relapsed. In conclusion, YB-1 correlatively expresses with NSC markers where it functions to promote cell growth and inhibit differentiation..
Nicolaides, T.P.
Li, H.
Solomon, D.A.
Hariono, S.
Hashizume, R.
Barkovich, K.
Baker, S.J.
Paugh, B.S.
Jones, C.
Forshew, T.
Hindley, G.F.
Hodgson, J.G.
Kim, J.-.
Rowitch, D.H.
Weiss, W.A.
Waldman, T.A.
James, C.D.
(2011). Targeted therapy for BRAFV600E malignant astrocytoma. Clin cancer res,
Vol.17
(24),
pp. 7595-7604.
show abstract
PURPOSE: Malignant astrocytomas (MA) are aggressive central nervous system tumors with poor prognosis. Activating mutation of BRAF (BRAF(V600E)) has been reported in a subset of these tumors, especially in children. We have investigated the incidence of BRAF(V600E) in additional pediatric patient cohorts and examined the effects of BRAF blockade in preclinical models of BRAF(V600E) and wild-type BRAF MA. EXPERIMENTAL DESIGN: BRAF(V600E) mutation status was examined in two pediatric MA patient cohorts. For functional studies, BRAF(V600E) MA cell lines were used to investigate the effects of BRAF shRNA knockdown in vitro, and to investigate BRAF pharmacologic inhibition in vitro and in vivo. RESULTS: BRAF(V600E) mutations were identified in 11 and 10% of MAs from two distinct series of tumors (six of 58 cases total). BRAF was expressed in all MA cell lines examined, among which BRAF(V600E) was identified in four instances. Using the BRAF(V600E)-specific inhibitor PLX4720, pharmacologic blockade of BRAF revealed preferential antiproliferative activity against BRAF(V600E) mutant cells in vitro, in contrast to the use of shRNA-mediated knockdown of BRAF, which inhibited cell growth of glioma cell lines regardless of BRAF mutation status. Using orthotopic MA xenografts, we show that PLX4720 treatment decreases tumor growth and increases overall survival in mice-bearing BRAF(V600E) mutant xenografts, while being ineffective, and possibly tumor promoting, against xenografts with wild-type BRAF. CONCLUSIONS: Our results indicate a 10% incidence of activating BRAF(V600E) among pediatric MAs. With regard to implications for therapy, our results support evaluation of BRAF(V600E)-specific inhibitors for treating BRAF(V600E) MA patients..
Viana-Pereira, M.
Lee, A.
Popov, S.
Bax, D.A.
Al-Sarraj, S.
Bridges, L.R.
Stavale, J.N.
Hargrave, D.
Jones, C.
Reis, R.M.
(2011). Microsatellite Instability in Pediatric High Grade Glioma Is Associated with Genomic Profile and Differential Target Gene Inactivation. Plos one,
Vol.6
(5),
p. 10.
Williams, R.D.
Al-Saadi, R.
Chagtai, T.
Popov, S.
Messahel, B.
Sebire, N.
Gessler, M.
Wegert, J.
Graf, N.
Leuschner, I.
Hubank, M.
Jones, C.
Vujanic, G.
Pritchard-Jones, K.
Children's Cancer and Leukaemia Group,
SIOP Wilms' Tumour Biology Group,
(2010). Subtype-specific FBXW7 mutation and MYCN copy number gain in Wilms' tumor. Clin cancer res,
Vol.16
(7),
pp. 2036-2045.
show abstract
PURPOSE: Wilms' tumor (WT), the most common pediatric renal malignancy, is associated with mutations in several well-characterized genes, most notably WT1, CTNNB1, WTX, and TP53. However, the majority of cases do not harbor mutations in these genes. We hypothesized that additional drivers of tumor behavior would be contained within areas of consistent genomic copy number change, especially those associated with the WT risk groups defined by the International Society of Paediatric Oncology (SIOP). EXPERIMENTAL DESIGN: We analyzed high-resolution (Affymetrix 250K single nucleotide polymorphism array) genomic copy number profiles of over 100 tumors from selected risk groups treated under the SIOP protocols, further characterizing genes of interest by sequencing, Multiplex Ligation-dependent Probe Amplification, or fluorescence in situ hybridization. RESULTS: We identified FBXW7, an E3 ubiquitin ligase component, as a novel Wilms' tumor gene, mutated or deleted in approximately 4% of tumors examined. Strikingly, 3 of 14 (21%) of tumors with epithelial type histology after neoadjuvant chemotherapy had FBXW7 aberrations, whereas a fourth WT patient had germline mutations in both FBXW7 and WT1. We also showed that MYCN copy number gain, detected in 9 of 104 (8.7%) of cases, is relatively common in WT and significantly more so in tumors of the high risk diffuse anaplastic subtype (6 of 19, 32%). CONCLUSIONS: Because MYCN is itself a target of FBXW7-mediated ubiquitination and degradation, these results suggest that a common pathway is dysregulated by different mechanisms in various WT subtypes. Emerging therapies that target MYCN, which is amplified in several other pediatric cancers, may therefore be of value in high risk Wilms' tumor..
Bax, D.A.
Mackay, A.
Little, S.E.
Carvalho, D.
Viana-Pereira, M.
Tamber, N.
Grigoriadis, A.E.
Ashworth, A.
Reis, R.M.
Ellison, D.W.
Al-Sarraj, S.
Hargrave, D.
Jones, C.
(2010). A distinct spectrum of copy number aberrations in pediatric high-grade gliomas. Clin cancer res,
Vol.16
(13),
pp. 3368-3377.
show abstract
PURPOSE: As genome-scale technologies begin to unravel the complexity of the equivalent tumors in adults, we can attempt detailed characterization of high-grade gliomas in children, that have until recently been lacking. Toward this end, we sought to validate and extend investigations of the differences between pediatric and adult tumors. EXPERIMENTAL DESIGN: We carried out copy number profiling by array comparative genomic hybridization using a 32K bacterial artificial chromosome platform on 63 formalin-fixed paraffin-embedded cases of high-grade glioma arising in children and young people (<23 years). RESULTS: The genomic profiles of these tumors could be subclassified into four categories: those with stable genomes, which were associated with a better prognosis; those with aneuploid and those with highly rearranged genomes; and those with an amplifier genotype, which had a significantly worse clinical outcome. Independent of this was a clear segregation of cases with 1q gain (more common in children) from those with concurrent 7 gain/10q loss (a defining feature of adults). Detailed mapping of all the amplification and deletion events revealed numerous low-frequency amplifications, including IGF1R, PDGFRB, PIK3CA, CDK6, CCND1, and CCNE1, and novel homozygous deletions encompassing unknown genes, including those at 5q35, 10q25, and 22q13. Despite this, aberrations targeting the "core signaling pathways" in adult glioblastomas are significantly underrepresented in the pediatric setting. CONCLUSIONS: These data highlight that although there are overlaps in the genomic events driving gliomagenesis of all ages, the pediatric disease harbors a distinct spectrum of copy number aberrations compared with adults..
Gaspar, N.
Sharp, S.Y.
Eccles, S.A.
Gowan, S.
Popov, S.
Jones, C.
Pearson, A.
Vassal, G.
Workman, P.
(2010). Mechanistic evaluation of the novel HSP90 inhibitor NVP-AUY922 in adult and pediatric glioblastoma. Mol cancer ther,
Vol.9
(5),
pp. 1219-1233.
show abstract
The dismal prognosis of glioblastoma (GB) indicates the urgent need for new therapies for these tumors. Heat shock protein 90 (HSP90) inhibitors induce the proteasome-mediated degradation of many oncogenic client proteins involved in all of the hallmark characteristics of cancer. Here, we explored the mechanistic potential of the potent synthetic diarylisoxazole amide resorcinol HSP90 inhibitor, NVP-AUY922, in adult and pediatric GB. In vitro antiproliferative potency (nanomolar range) was seen in both adult and pediatric human GB cell lines with different molecular pathologies. A cytostatic effect was observed in all GB lines; more apoptosis was observed at lower concentrations in the SF188 pediatric GB line and at 144 hours in the slower growing KNS42 pediatric GB line, as compared with the adult GB lines U87MG and SF268. In vitro combination studies with inhibitors of phosphoinositide 3-kinase/mammalian target of rapamycin (PI-103) or mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase (PD-0325901) supported the hypothesis that sustained inhibition of ERK up to 72 hours and at least temporary inhibition of AKT were necessary to induce apoptosis in GB lines. In athymic mice bearing established s.c U87MG GB xenografts, NVP-AUY922 (50 mg/kg i.p x 3 days) caused the inhibition of ERK1/2 and AKT phosphorylation and induced apoptosis, whereas 17-AAG used at maximum tolerated dose was less effective. NVP-AUY922 antitumor activity with objective tumor regression resulted from antiproliferative, proapoptotic, and antiangiogenic effects, the latter shown by decreased microvessel density and HIF1alpha levels. Our results have established a mechanistic proof of concept for the potential of novel synthetic HSP90 inhibitors in adult and pediatric GB, alone or in combination with phosphoinositide 3-kinase/mammalian target of rapamycin and mitogen-activated protein/ERK kinase inhibitors..
Natrajan, R.
Weigelt, B.
Mackay, A.
Geyer, F.C.
Grigoriadis, A.
Tan, D.S.
Jones, C.
Lord, C.J.
Vatcheva, R.
Rodriguez-Pinilla, S.M.
Palacios, J.
Ashworth, A.
Reis-Filho, J.S.
(2010). An integrative genomic and transcriptomic analysis reveals molecular pathways and networks regulated by copy number aberrations in basal-like, HER2 and luminal cancers. Breast cancer res treat,
Vol.121
(3),
pp. 575-589.
show abstract
Breast cancer is a heterogeneous disease caused by the accumulation of genetic changes in neoplastic cells. We hypothesised that molecular subtypes of breast cancer may be driven by specific constellations of genes whose expression is regulated by gene copy number aberrations. To address this question, we analysed a series of 48 microdissected grade III ductal carcinomas using high resolution microarray comparative genomic hybridisation and mRNA expression arrays. There were 5,931 genes whose expression significantly correlates with copy number identified; out of these, 1,897 genes were significantly differentially expressed between basal-like, HER2 and luminal tumours. Ingenuity Pathway Analysis (IPA) revealed that 'G1/S cell cycle regulation' and 'BRCA1 in DNA damage control' pathways were significantly enriched for genes whose expression correlates with copy number and are differentially expressed between the molecular subtypes of breast cancer. IPA of genes whose expression significantly correlates with copy number in each molecular subtype individually revealed that canonical pathways involved in oestrogen receptor (ER) signalling and DNA repair are enriched for these genes. We also identified 32, 157 and 265 genes significantly overexpressed when amplified in basal-like, HER2 and luminal cancers, respectively. These lists include known and novel potential therapeutic targets (e.g. HER2 and PPM1D in HER2 cancers). Our results provide strong circumstantial evidence that different patterns of genetic aberrations in distinct molecular subtypes of breast cancer contribute to their specific transcriptomic profiles and that biological phenomena characteristic of each subtype (e.g. proliferation, HER2 and ER signalling) may be driven by specific patterns of copy number aberrations..
Gaspar, N.
Marshall, L.
Perryman, L.
Bax, D.A.
Little, S.E.
Viana-Pereira, M.
Sharp, S.Y.
Vassal, G.
Pearson, A.D.
Reis, R.M.
Hargrave, D.
Workman, P.
Jones, C.
(2010). MGMT-Independent Temozolomide Resistance in Pediatric Glioblastoma Cells Associated with a PI3-Kinase-Mediated HOX/Stem Cell Gene Signature. Cancer research,
Vol.70
(22),
pp. 9243-10.
Paugh, B.S.
Qu, C.
Jones, C.
Liu, Z.
Adamowicz-Brice, M.
Zhang, J.
Bax, D.A.
Coyle, B.
Barrow, J.
Hargrave, D.
Lowe, J.
Gajjar, A.
Zhao, W.
Broniscer, A.
Ellison, D.W.
Grundy, R.G.
Baker, S.J.
(2010). Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease. J clin oncol,
Vol.28
(18),
pp. 3061-3068.
show abstract
PURPOSE: To define copy number alterations and gene expression signatures underlying pediatric high-grade glioma (HGG). PATIENTS AND METHODS: We conducted a high-resolution analysis of genomic imbalances in 78 de novo pediatric HGGs, including seven diffuse intrinsic pontine gliomas, and 10 HGGs arising in children who received cranial irradiation for a previous cancer using single nucleotide polymorphism microarray analysis. Gene expression was analyzed with gene expression microarrays for 53 tumors. Results were compared with publicly available data from adult tumors. RESULTS: Significant differences in copy number alterations distinguish childhood and adult glioblastoma. PDGFRA was the predominant target of focal amplification in childhood HGG, including diffuse intrinsic pontine gliomas, and gene expression analyses supported an important role for deregulated PDGFRalpha signaling in pediatric HGG. No IDH1 hotspot mutations were found in pediatric tumors, highlighting molecular differences with adult secondary glioblastoma. Pediatric and adult glioblastomas were clearly distinguished by frequent gain of chromosome 1q (30% v 9%, respectively) and lower frequency of chromosome 7 gain (13% v 74%, respectively) and 10q loss (35% v 80%, respectively). PDGFRA amplification and 1q gain occurred at significantly higher frequency in irradiation-induced tumors, suggesting that these are initiating events in childhood gliomagenesis. A subset of pediatric HGGs showed minimal copy number changes. CONCLUSION: Integrated molecular profiling showed substantial differences in the molecular features underlying pediatric and adult HGG, indicating that findings in adult tumors cannot be simply extrapolated to younger patients. PDGFRalpha may be a useful target for pediatric HGG, including diffuse pontine gliomas..
Huijbers, I.J.
Iravani, M.
Popov, S.
Robertson, D.
Al-Sarraj, S.
Jones, C.
Isacke, C.M.
(2010). A role for fibrillar collagen deposition and the collagen internalization receptor endo180 in glioma invasion. Plos one,
Vol.5
(3),
p. e9808.
show abstract
BACKGROUND: Glioblastoma multiforme (GBM, WHO grade IV) is the most common and most malignant of astrocytic brain tumors, and is associated with rapid invasion into neighboring tissue. In other tumor types it is well established that such invasion involves a complex interaction between tumor cells and locally produced extracellular matrix. In GBMs, surprisingly little is known about the associated matrix components, in particular the fibrillar proteins such as collagens that are known to play a key role in the invasion of other tumor types. METHODOLOGY/PRINCIPAL FINDINGS: In this study we have used both the Masson's trichrome staining and a high resolution multiple immunofluorescence labeling method to demonstrate that intratumoral fibrillar collagens are an integral part of the extracellular matrix in a subset of GBMs. Correlated with this collagen deposition we observed high level expression of the collagen-binding receptor Endo180 (CD280) in the tumor cells. Further, interrogation of multiple expression array datasets identified Endo180 as one of the most highly upregulated transcripts in grade IV GBMs compared to grade III gliomas. Using promoter analysis studies we show that this increased expression is, in part, mediated via TGF-beta signaling. Functionally, we demonstrate that Endo180 serves as the major collagen internalization receptor in GBM cell lines and provide the first evidence that this activity is critical for the invasion of GBM cells through fibrillar collagen matrices. CONCLUSIONS/SIGNIFICANCE: This study demonstrates, for the first time, that fibrillar collagens are extensively deposited in GBMs and that the collagen internalization receptor Endo180 is both highly expressed in these tumors and that it serves to mediate the invasion of tumor cells through collagen-containing matrices. Together these data provide important insights into the mechanism of GBM invasion and identify Endo180 as a potential target to limit matrix turnover by glioma cells and thereby restrict tumor progression..
Drake, K.M.
Ruteshouser, E.C.
Natrajan, R.
Harbor, P.
Wegert, J.
Gessler, M.
Pritchard-Jones, K.
Grundy, P.
Dome, J.
Huff, V.
Jones, C.
Aldred, M.A.
(2009). Loss of heterozygosity at 2q37 in sporadic Wilms' tumor: putative role for miR-562. Clin cancer res,
Vol.15
(19),
pp. 5985-5992.
show abstract
PURPOSE: Wilms' tumor is a childhood cancer of the kidney with an incidence of approximately 1 in 10,000. Cooccurrence of Wilms' tumor with 2q37 deletion syndrome, an uncommon constitutional chromosome abnormality, has been reported previously in three children. Given these are independently rare clinical entities, we hypothesized that 2q37 harbors a tumor suppressor gene important in Wilms' tumor pathogenesis. EXPERIMENTAL DESIGN: To test this, we performed loss of heterozygosity analysis in a panel of 226 sporadic Wilms' tumor samples and mutation analysis of candidate genes. RESULTS: Loss of heterozygosity was present in at least 4% of cases. Two tumors harbored homozygous deletions at 2q37.1, supporting the presence of a tumor suppressor gene that follows a classic two-hit model. However, no other evidence of second mutations was found, suggesting that heterozygous deletion alone may be sufficient to promote tumorigenesis in concert with other genomic abnormalities. We show that miR-562, a microRNA within the candidate region, is expressed only in kidney and colon and regulates EYA1, a critical gene for renal development. miR-562 expression is reduced in Wilms' tumor and may contribute to tumorigenesis by deregulating EYA1. Two other candidate regions were localized at 2q37.3 and 2qter, but available data from patients with constitutional deletions suggest that these probably do not confer a high risk for Wilms' tumor. CONCLUSIONS: Our data support the presence of a tumor suppressor gene at 2q37.1 and suggest that, in individuals with constitutional 2q37 deletions, any increased risk for developing Wilms' tumor likely correlates with deletions encompassing 2q37.1..
Gaspar, N.
Sharp, S.Y.
Pacey, S.
Jones, C.
Walton, M.
Vassal, G.
Eccles, S.
Pearson, A.
Workman, P.
(2009). Acquired resistance to 17-allylamino-17-demethoxygeldanamycin (17-AAG, tanespimycin) in glioblastoma cells. Cancer res,
Vol.69
(5),
pp. 1966-1975.
show abstract
Heat shock protein 90 (HSP90) inhibitors, such as 17-allylamino-17-demethoxygeldanamycin (17-AAG, tanespimycin), which is currently in phase II/phase III clinical trials, are promising new anticancer agents. Here, we explored acquired resistance to HSP90 inhibitors in glioblastoma (GB), a primary brain tumor with poor prognosis. GB cells were exposed continuously to increased 17-AAG concentrations. Four 17-AAG-resistant GB cell lines were generated. High-resistance levels with resistance indices (RI = resistant line IC(50)/parental line IC(50)) of 20 to 137 were obtained rapidly (2-8 weeks). After cessation of 17-AAG exposure, RI decreased and then stabilized. Cross-resistance was found with other ansamycin benzoquinones but not with the structurally unrelated HSP90 inhibitors, radicicol, the purine BIIB021, and the resorcinylic pyrazole/isoxazole amide compounds VER-49009, VER-50589, and NVP-AUY922. An inverse correlation between NAD(P)H/quinone oxidoreductase 1 (NQO1) expression/activity and 17-AAG IC(50) was observed in the resistant lines. The NQO1 inhibitor ES936 abrogated the differential effects of 17-AAG sensitivity between the parental and resistant lines. NQO1 mRNA levels and NQO1 DNA polymorphism analysis indicated different underlying mechanisms: reduced expression and selection of the inactive NQO1*2 polymorphism. Decreased NQO1 expression was also observed in a melanoma line with acquired resistance to 17-AAG. No resistance was generated with VER-50589 and NVP-AUY922. In conclusion, low NQO1 activity is a likely mechanism of acquired resistance to 17-AAG in GB, melanoma, and, possibly, other tumor types. Such resistance can be overcome with novel HSP90 inhibitors..
Bielen, A.
Perryman, L.
Hoffman, F.
Pritchard-Jones, K.
Jones, C.
(2009). Inhibition of the insulin-like growth factor 1 receptor (IGF1R) induces cell cycle arrest and apoptosis in Wilms tumor cells. Cancer research,
Vol.69.
Messahel, B.
Williams, R.
Ridolfi, A.
A'hern, R.
Warren, W.
Tinworth, L.
Hobson, R.
Al-Saadi, R.
Whyman, G.
Brundler, M.-.
Kelsey, A.
Sebire, N.
Jones, C.
Vujanic, G.
Pritchard-Jones, K.
Children's Cancer and Leukaemia Group (CCLG),
(2009). Allele loss at 16q defines poorer prognosis Wilms tumour irrespective of treatment approach in the UKW1-3 clinical trials: a Children's Cancer and Leukaemia Group (CCLG) Study. Eur j cancer,
Vol.45
(5),
pp. 819-826.
show abstract
Survival from Wilms tumour is excellent. Hence, better markers are required to restrict treatments causing late sequelae to those at highest risk of relapse. We investigated the prognostic significance of loss of heterozygosity (LOH) on 1p and 16q in 426 favourable histology Wilms tumours treated with either immediate nephrectomy (63%) or preoperative chemotherapy (37%). Four years RFS and OS were 84.6% and 92.0%, respectively. 10.3% tumours had LOH 1p, 14.6% LOH 16q, with 2.6% at both loci. In multivariate analysis, LOH 16q was associated with an increased risk of relapse (hazard ratio (HR) 2.69, 95%CI: 1.47-4.92) and death (HR 2.67, 95%CI: 1.17-6.06). LOH 1p showed no significant associations. These results were not influenced by treatment approach. LOH 16q is an adverse risk factor in favourable histology Wilms tumour, regardless of initial approach to therapy. Its relationship with histological risk groups defined after neo-adjuvant chemotherapy requires analysis in a larger series, and is the subject of the current SIOP WT 2001 trial..
Dallosso, A.R.
Hancock, A.L.
Szemes, M.
Moorwood, K.
Chilukamarri, L.
Tsai, H.-.
Sarkar, A.
Barasch, J.
Vuononvirta, R.
Jones, C.
Pritchard-Jones, K.
Royer-Pokora, B.
Lee, S.B.
Owen, C.
Malik, S.
Feng, Y.
Frank, M.
Ward, A.
Brown, K.W.
Malik, K.
(2009). Frequent long-range epigenetic silencing of protocadherin gene clusters on chromosome 5q31 in Wilms' tumor. Plos genet,
Vol.5
(11),
p. e1000745.
show abstract
Wilms' tumour (WT) is a pediatric tumor of the kidney that arises via failure of the fetal developmental program. The absence of identifiable mutations in the majority of WTs suggests the frequent involvement of epigenetic aberrations in WT. We therefore conducted a genome-wide analysis of promoter hypermethylation in WTs and identified hypermethylation at chromosome 5q31 spanning 800 kilobases (kb) and more than 50 genes. The methylated genes all belong to alpha-, beta-, and gamma-protocadherin (PCDH) gene clusters (Human Genome Organization nomenclature PCDHA@, PCDHB@, and PCDHG@, respectively). This demonstrates that long-range epigenetic silencing (LRES) occurs in developmental tumors as well as in adult tumors. Bisulfite polymerase chain reaction analysis showed that PCDH hypermethylation is a frequent event found in all Wilms' tumor subtypes. Hypermethylation is concordant with reduced PCDH expression in tumors. WT precursor lesions showed no PCDH hypermethylation, suggesting that de novo PCDH hypermethylation occurs during malignant progression. Discrete boundaries of the PCDH domain are delimited by abrupt changes in histone modifications; unmethylated genes flanking the LRES are associated with permissive marks which are absent from methylated genes within the domain. Silenced genes are marked with non-permissive histone 3 lysine 9 dimethylation. Expression analysis of embryonic murine kidney and differentiating rat metanephric mesenchymal cells demonstrates that Pcdh expression is developmentally regulated and that Pcdhg@ genes are expressed in blastemal cells. Importantly, we show that PCDHs negatively regulate canonical Wnt signalling, as short-interfering RNA-induced reduction of PCDHG@ encoded proteins leads to elevated beta-catenin protein, increased beta-catenin/T-cell factor (TCF) reporter activity, and induction of Wnt target genes. Conversely, over-expression of PCDHs suppresses beta-catenin/TCF-reporter activity and also inhibits colony formation and growth of cancer cells in soft agar. Thus PCDHs are candidate tumor suppressors that modulate regulatory pathways critical in development and disease, such as canonical Wnt signaling..
Vuononvirta, R.
Sebire, N.J.
Messahel, B.
Perusinghe, N.
Reis-Filho, J.S.
Pritchard-Jones, K.
Vujanic, G.M.
Jones, C.
(2009). Expression of hepatocyte growth factor and its receptor met in Wilms' tumors and nephrogenic rests reflects their roles in kidney development. Clin cancer res,
Vol.15
(8),
pp. 2723-2730.
show abstract
PURPOSE: Hepatocyte growth factor (HGF) and its receptor Met are known to play diverse roles in both organogenesis and cancer. Wilms' tumor (WT) is a prototype for the link between abrogated development and neoplasia, with dysregulation of growth factor/receptor pathways playing key roles. Despite this, an understanding of the HGF/Met axis in the process is lacking. EXPERIMENTAL DESIGN: Observing copy number alterations at the loci for these genes in WTs and their precursor lesions nephrogenic rests, we examined protein expression by immunohistochemistry and investigated the effects of HGF on an in vitro model of kidney development. RESULTS: HGF was preferentially expressed in the blastemal cells of nephrogenic rests but not WTs. Met expression was infrequent and restricted to well-differentiated epithelial cells and stroma in both lesions. In an independent cohort of favorable histology WTs on a tissue microarray, HGF was expressed in 15 of 193 (8%) cases and correlated with a predominance of epithelial cells, whereas Met expression was observed in 25 of 179 (14%) cases and was associated with stromal subtypes. In a mouse mesonephric cell line model, we observed Met expression in culture conditions reflecting both mesenchymal and epithelial differentiation, whereas HGF was up-regulated in association with acquisition of a more epithelial-like phenotype. This could be mimicked by exogenous exposure of mesenchymal-like cells to recombinant HGF. CONCLUSIONS: These data show that the relatively infrequent expression of HGF and Met in WT tumorigenesis reflects their roles in nephrogenesis, particularly the mesenchymal-to-epithelial transition, rather than a dependence on oncogenic signaling pathways..
Bax, D.A.
Gaspar, N.
Little, S.E.
Marshall, L.
Perryman, L.
Regairaz, M.
Viana-Pereira, M.
Vuononvirta, R.
Sharp, S.Y.
Reis-Filho, J.S.
Stávale, J.N.
Al-Sarraj, S.
Reis, R.M.
Vassal, G.
Pearson, A.D.
Hargrave, D.
Ellison, D.W.
Workman, P.
Jones, C.
(2009). EGFRvIII deletion mutations in pediatric high-grade glioma and response to targeted therapy in pediatric glioma cell lines. Clin cancer res,
Vol.15
(18),
pp. 5753-5761.
show abstract
PURPOSE: The epidermal growth factor receptor (EGFR) is amplified and overexpressed in adult glioblastoma, with response to targeted inhibition dependent on the underlying biology of the disease. EGFR has thus far been considered to play a less important role in pediatric glioma, although extensive data are lacking. We have sought to clarify the role of EGFR in pediatric high-grade glioma (HGG). EXPERIMENTAL DESIGN: We retrospectively studied a total of 90 archival pediatric HGG specimens for EGFR protein overexpression, gene amplification, and mutation and assessed the in vitro sensitivity of pediatric glioma cell line models to the small-molecule EGFR inhibitor erlotinib. RESULTS: Amplification was detected in 11% of cases, with corresponding overexpression of the receptor. No kinase or extracellular domain mutations were observed; however, 6 of 35 (17%) cases harbored the EGFRvIII deletion, including two anaplastic oligodendrogliomas and a gliosarcoma overexpressing EGFRvIII in the absence of gene amplification and coexpressing platelet-derived growth factor receptor alpha. Pediatric glioblastoma cells transduced with wild-type or deletion mutant EGFRvIII were not rendered more sensitive to erlotinib despite expressing wild-type PTEN. Phosphorylated receptor tyrosine kinase profiling showed a specific activation of platelet-derived growth factor receptor alpha/beta in EGFRvIII-transduced pediatric glioblastoma cells, and targeted coinhibition with erlotinib and imatinib leads to enhanced efficacy in this model. CONCLUSIONS: These data identify an elevated frequency of EGFR gene amplification and EGFRvIII mutation in pediatric HGG than previously recognized and show the likely necessity of targeting multiple genetic alterations in the tumors of these children..
Bax, D.A.
Little, S.E.
Gaspar, N.
Perryman, L.
Marshall, L.
Viana-Pereira, M.
Jones, T.A.
Williams, R.D.
Grigoriadis, A.
Vassal, G.
Workman, P.
Sheer, D.
Reis, R.M.
Pearson, A.D.
Hargrave, D.
Jones, C.
(2009). Molecular and phenotypic characterisation of paediatric glioma cell lines as models for preclinical drug development. Plos one,
Vol.4
(4),
p. e5209.
show abstract
BACKGROUND: Although paediatric high grade gliomas resemble their adult counterparts in many ways, there appear to be distinct clinical and biological differences. One important factor hampering the development of new targeted therapies is the relative lack of cell lines derived from childhood glioma patients, as it is unclear whether the well-established adult lines commonly used are representative of the underlying molecular genetics of childhood tumours. We have carried out a detailed molecular and phenotypic characterisation of a series of paediatric high grade glioma cell lines in comparison to routinely used adult lines. PRINCIPAL FINDINGS: All lines proliferate as adherent monolayers and express glial markers. Copy number profiling revealed complex genomes including amplification and deletions of genes known to be pivotal in core glioblastoma signalling pathways. Expression profiling identified 93 differentially expressed genes which were able to distinguish between the adult and paediatric high grade cell lines, including a number of kinases and co-ordinated sets of genes associated with DNA integrity and the immune response. SIGNIFICANCE: These data demonstrate that glioma cell lines derived from paediatric patients show key molecular differences to those from adults, some of which are well known, whilst others may provide novel targets for evaluation in primary tumours. We thus provide the rationale and demonstrate the practicability of using paediatric glioma cell lines for preclinical and mechanistic studies..
Vuononvirta, R.
Sebire, N.J.
Dallosso, A.R.
Reis-Filho, J.S.
Williams, R.D.
Mackay, A.
Fenwick, K.
Grigoriadis, A.
Ashworth, A.
Pritchard-Jones, K.
Brown, K.W.
Vujanic, G.M.
Jones, C.
(2008). Perilobar nephrogenic rests are nonobligate molecular genetic precursor lesions of insulin-like growth factor-II-associated Wilms tumors. Clin cancer res,
Vol.14
(23),
pp. 7635-7644.
show abstract
PURPOSE: Perilobar nephrogenic rests (PLNRs) are abnormally persistent foci of embryonal immature blastema that have been associated with dysregulation at the 11p15 locus by genetic/epigenetic means and are thought to be precursor lesions of Wilms tumor. The precise genomic events are, however, largely unknown. EXPERIMENTAL DESIGN: We used array comparative genomic hybridization to analyze a series of 50 PLNRs and 25 corresponding Wilms tumors characterized for 11p15 genetic/epigenetic alterations and insulin-like growth factor-II expression. RESULTS: The genomic profiles of PLNRs could be subdivided into three categories: those with no copy number changes (22 of 50, 44%); those with single, whole chromosome alterations (8 of 50, 16%); and those with multiple gains/losses (20 of 50, 40%). The most frequent aberrations included 1p- (7 of 50, 14%) +18 (6 of 50, 12%), +13 (5 of 50, 10%), and +12 (3 of 50, 6%). For the majority (19 of 25, 76%) of cases, the rest harbored a subset of the copy number changes in the associated Wilms tumor. We identified a temporal order of genomic changes, which occur during the insulin-like growth factor-II/PLNR pathway of Wilms tumorigenesis, with large-scale chromosomal alterations such as 1p-, +12, +13, and +18 regarded as "early" events. In some of the cases (24%), the PLNRs harbored large-scale copy number changes not observed in the concurrent Wilms tumor, including +10p, +14q, and +18. CONCLUSIONS: These data suggest that although the evidence for PLNRs as precursors is compelling, not all lesions must necessarily undergo malignant transformation..
Natrajan, R.
Warren, W.
Messahel, B.
Reis-Filho, J.S.
Brundler, M.-.
Dome, J.S.
Grundy, P.E.
Vujanic, G.
Pritchard-Jones, K.
Jones, C.
(2008). Complex patterns of chromosome 9 alterations including the p16INK4a locus in Wilms tumours. J clin pathol,
Vol.61
(1),
pp. 95-102.
show abstract
BACKGROUND: Previous data implicating genetic and epigenetic events on chromosome 9, including the CDKN2A/2B locus, as molecular predictors of Wilms tumour relapse, have been conflicting. AIMS: To clarify this using genome-wide and focused molecular genetic analysis. METHODS: Microarray-based comparative genomic hybridisation (aCGH) using genome-wide coverage was applied to 76 favourable histology Wilms tumours. Additional investigation of the 9p21 locus was carried out using loss of heterozygosity (LOH) and fluorescence in situ hybridisation (FISH), as well as immunohistochemistry for CDKN2A/p16(INK4a) on a paediatric renal tumour tissue microarray. RESULTS: Approximately half of the tumours were found to show chromosome 9 copy number changes. Those cases which harboured alterations comprised at least four distinct patterns: gain of the entire chromosome, loss of 9p, gain of 9q34, or a more complex combination of gains/losses. None of these tumour groups showed any statistically significant correlation with clinicopathological variables. Deletion mapping of 9p by LOH revealed several regions of overlap, including the CDKN2A/2B locus in 4/34 (11.8%) tumours, which was confirmed to represent hemizygous deletions by FISH. CDKN2A/p16(INK4a) protein expression was predominantly negative in Wilms tumours as assessed by immunohistochemistry on a tissue array, reflecting the expression pattern in normal kidney. However, 38/236 (16.1%) non-anaplastic Wilms tumours, 4/9 (44.4%) anaplastic Wilms tumours, 5/7 (71.4%) rhabdoid tumours of the kidney, and 4/10 (40%) clear cell sarcomas of the kidney showed nuclear CDKN2A/p16(INK4a )immunoreactivity. CONCLUSIONS: These data reveal the complex nature of genetic alterations on chromosome 9 in Wilms tumours, but do not provide evidence for their involvement in or association with treatment failure..
Simonavicius, N.
Robertson, D.
Bax, D.A.
Jones, C.
Huijbers, I.J.
Isacke, C.M.
(2008). Endosialin (CD248) is a marker of tumor-associated pericytes in high-grade glioma. Mod pathol,
Vol.21
(3),
pp. 308-315.
show abstract
Gliomas are the most frequent primary tumors of the central nervous system in adults. The most prevalent and aggressive subclass of these is glioblastoma multiforme, which is characterized by massive neovascularization. Endosialin (CD248) has generated interest as a target for antiangiogenic therapy following reports that its expression is upregulated on angiogenic endothelial cells. We demonstrate here that endosialin is not expressed in normal human adult brain but is strongly upregulated in the angiogenic vasculature of all high-grade glioma specimens examined. However, by taking advantage of a technique which allows for multiple fluorescent labeling of formalin-fixed paraffin-embedded archival sections, we demonstrate unambiguously that endosialin is not expressed by the glioma endothelial cells but on closely associated perivascular cells. With increasing awareness that targeting pericytes is an attractive adjunct in antiangiogenic therapy, this finding has important implications for understanding the molecular mechanisms regulating angiogenesis in these highly vascularized tumors..
Arriola, E.
Marchio, C.
Tan, D.S.
Drury, S.C.
Lambros, M.B.
Natrajan, R.
Rodriguez-Pinilla, S.M.
Mackay, A.
Tamber, N.
Fenwick, K.
Jones, C.
Dowsett, M.
Ashworth, A.
Reis-Filho, J.S.
(2008). Genomic analysis of the HER2/TOP2A amplicon in breast cancer and breast cancer cell lines. Lab invest,
Vol.88
(5),
pp. 491-503.
show abstract
HER2 and TOP2A are targets for the therapeutic agents trastuzumab and anthracyclines and are frequently amplified in breast cancers. The aims of this study were to provide a detailed molecular genetic analysis of the 17q12-q21 amplicon in breast cancers harbouring HER2/TOP2A co-amplification and to investigate additional recurrent co-amplifications in HER2/TOP2A-co-amplified cancers. In total, 15 breast cancers with HER2 amplification, 10 of which also harboured TOP2A amplification, as defined by chromogenic in situ hybridisation, and 6 breast cancer cell lines known to be amplified for HER2 were subjected to high-resolution microarray-based comparative genomic hybridisation analysis. This revealed that the genomes of 12 cases were characterised by at least one localised region of clustered, relatively narrow peaks of amplification, with each cluster confined to a single chromosome arm (ie 'firestorm' pattern) and 3 cases displayed many narrow segments of duplication and deletion affecting the vast majority of chromosomes (ie 'sawtooth' pattern). The smallest region of amplification (SRA) on 17q12 in the whole series extended from 34.73 to 35.48 Mb, and encompassed HER2 but not TOP2A. In HER2/TOP2A-co-amplified samples, the SRA extended from 34.73 to 36.54 Mb, spanning a region of approximately 1.8 Mb. Apart from HER2 and TOP2A, this region encompassed four additional genes whose expression levels as defined by quantitative real-time PCR are significantly higher in HER2/TOP2A-co-amplified vs HER2-amplified breast cancers: CASC3, CDC6, RARA and SMARCE1. Of the cell lines studied, SKBR3 and UACC812 showed HER2/TOP2A co-amplification. In conclusion, this is the first detailed genome-wide characterisation of HER2/TOP2A-amplified breast cancers; cell lines were identified that can be used to model these cancers in vitro. The 17q12 amplicon is complex and harbours multiple genes that may be associated with breast cancer development and progression, and potentially exploitable as therapeutic targets..
Simpson, P.T.
Rels-Filho, J.S.
Lambros, M.B.
Jones, C.
Steele, D.
Mackay, A.
Iravani, M.
Fenwick, K.
Dexter, T.
Jones, A.
Reid, L.
Da Silva, L.
Shin, S.J.
Hardisson, D.
Ashworth, A.
Schmitt, F.C.
Palacios, J.
Lakhani, S.R.
(2008). Molecular profiling pleomorphic lobular carcinomas of the breast: evidence for a common molecular genetic pathway with classic lobular carcinomas. Journal of pathology,
Vol.215
(3),
pp. 231-14.
Rudenko, H.C.
Else, M.
Dearden, C.
Brito-Babapulle, V.
Jones, C.
Dexter, T.
Fenwick, K.
Mackay, A.
Ashworth, A.
Matutes, E.
Gonzalez, D.
Catovsky, D.
Morgan, G.J.
(2008). Characterising the TP53-deleted subgroup of chronic lymphocytic leukemia: an analysis of additional cytogenetic abnormalities detected by interphase fluorescence in situ hybridisation and array-based comparative genomic hybridisation. Leuk lymphoma,
Vol.49
(10),
pp. 1879-1886.
show abstract
Deletion of the TP53 gene on chromosome 17p13.1 is the prognostic factor associated with the shortest survival in CLL. We used array-based comparative genomic hybridisation (arrayCGH) to identify additional DNA copy number changes in peripheral blood samples from 74 LRF CLL4 trial patients, 37 with >or=5% and 37 without TP53-deleted cells. ArrayCGH reliably detected deletions on 17p, including the TP53 locus, in cases with >or=50%TP53-deleted cells detected by fluorescence in situ hybridisation, plus seven additional cases with deleted regions on 17p excluding TP53. Losses on chromosomal regions 18p and/or 20p were found exclusively in cases with >or=5%TP53-deleted cells (p<0.001), 38% having one or both losses. The incidence of additional cytogenetic abnormalities, reflecting an increased chromosomal instability, was higher in >or=5%TP53-deleted cases (p=0.02). In particular, amplification of 2p and deletion of 6q were both more frequent. Cases with >20%TP53-deleted cells had the worst prognosis in the LRF CLL4 trial..
Little, S.E.
Bax, D.A.
Rodriguez-Pinilla, M.
Natrajan, R.
Messahel, B.
Pritchard-Jones, K.
Vujanic, G.M.
Reis-Filho, J.S.
Jones, C.
(2007). Multifaceted dysregulation of the epidermal growth factor receptor pathway in clear cell sarcoma of the kidney. Clin cancer res,
Vol.13
(15 Pt 1),
pp. 4360-4364.
show abstract
PURPOSE: Epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase overexpressed in a variety of human malignancies, against which targeted therapies have shown efficacy in lung and brain tumors. Clinical responses to EGFR inhibitors have been found to be highly dependent on the presence of activating mutations, whereas gene amplification, downstream activation of Akt, and abnormalities in PTEN are also reported predictive factors. We sought to evaluate these variables in pediatric renal tumors. EXPERIMENTAL DESIGN: We screened a series of 307 pediatric renal tumors for EGFR expression by immunohistochemistry and gene amplification by chromogenic in situ hybridization. In identifying a striking predilection for certain tumor types, we further analyzed the clear cell sarcomas of the kidney (CCSK) for mutations in EGFR and PTEN. RESULTS: Although only 23 of 177 (13.0%) nonanaplastic Wilms' tumors were EGFR positive, 4 of 11 (36.4%) anaplastic tumors showed receptor overexpression. In addition, 5 of 9 (55.6%) mesoblastic nephromas and 12 of 12 (100%) CCSKs were strongly immunoreactive for EGFR. In studying the CCSKs in more detail, we identified gene amplification in 1 of 12 (8.3%) cases and a somatic T790M EGFR mutation in a further case. These two samples additionally harbored mutations in PTEN. Downstream pathway activation, as assayed by phosphorylated Akt expression, was observed in 8 of 12 (66.7%) cases. CONCLUSIONS: Together, these data show dysregulation of the EGFR pathway at multiple levels in CCSKs. Identification of factors predictive of poor response to targeted therapy, including the drug resistance T790M mutation, may provide a rationale for upfront trials with irreversible inhibitors of EGFR in children with these tumors..
Natrajan, R.
Little, S.E.
Sodha, N.
Reis-Filho, J.S.
Mackay, A.
Fenwick, K.
Ashworth, A.
Perlman, E.J.
Dome, J.S.
Grundy, P.E.
Pritchard-Jones, K.
Jones, C.
(2007). Analysis by array CGH of genomic changes associated with the progression or relapse of Wilms' tumour. J pathol,
Vol.211
(1),
pp. 52-59.
show abstract
Despite aggressive salvage regimens, approximately half of all children who suffer a Wilms' tumour recurrence will die of their disease. Although there are increasing data on molecular genetic prognostic factors present in the tumour at diagnosis, there is little information regarding the molecular events that occur with Wilms' tumour progression and relapse. In the present study, microarray-based comparative genomic hybridization (aCGH) analysis has been carried out on 58 Wilms' tumour samples, which included 38 untreated primary and 20 recurrent tumours. A higher degree of copy number changes was observed in the recurrent tumours (33.0% genomic clones) than in the primary tumour (21.2%). Paired analysis highlighted the acquisition of 15q gain with high levels of IGF1R expression in the tumour recurrence in two cases. The most statistically significant abnormality acquired between diagnosis and relapse was loss of 17p. One case that experienced 17p loss was classified as favourable histology at diagnosis, but exhibited diffuse anaplasia at recurrence and had a homozygous TP53 deletion. Another instructive case with a constitutional 11p13 deletion presented with bilateral tumours and suffered two subsequent recurrences in the left kidney. A somatic WT1 mutation was found only in the right kidney tumour, while the constitutional 11p13 deletion was the only abnormality detected in the initial left kidney tumour by aCGH. The two subsequent relapses in the left kidney contained an accumulation of additional genetic alterations, including an independent WT1 mutation..
Arriola, E.
Lambros, M.B.
Jones, C.
Dexter, T.
Mackay, A.
Tan, D.S.
Tamber, N.
Fenwick, K.
Ashworth, A.
Dowsett, M.
Reis-Filho, J.S.
(2007). Evaluation of Phi29-based whole-genome amplification for microarray-based comparative genomic hybridisation. Lab invest,
Vol.87
(1),
pp. 75-83.
show abstract
For the optimal performance of high throughput genomic technologies sufficient yields of high-quality DNA are crucial. Following microdissection, most samples fail to produce sufficient quantities of DNA for genome-wide experiments. Various PCR-based amplification methods have been used, but these usually produce nonuniform representations of the genome. Bacteriophage Phi29 DNA polymerase random-primed DNA amplification is based on isothermal multiple displacement amplification. We sought to define the genome representation of this method in a bacterial artificial chromosome microarray comparative genomic hybridisation (aCGH) platform. Test genomic female DNA was amplified using Phi29 amplification at four different starting concentrations (0.5, 5, 10 and 50 ng). These products were combined with unamplified and amplified genomic female DNA as reference. In addition, 50 ng of DNA from five microdissected breast cancer frozen samples, were amplified using the same method. Three combinations were performed: unamplified test with unamplified reference, amplified test with unamplified reference and both amplified tumour and reference DNA. aCGH was performed with an in-house 16 K BAC platform (a resolution of approximately 100 Kb). Pearson's correlation tests and hierarchical clustering were performed to compare the profiles obtained. aCGH profiles obtained with amplified test and unamplified reference female genomic DNA showed copy number biases throughout the genome. These biases were more conspicuous with smaller amounts of starting material and mapped to regions of known copy number polymorphisms. When similar concentrations of test and reference DNA were amplified, the biases were significantly reduced, rendering accurate profiles. For the tumours, representative profiles were obtained when both test and reference DNA were amplified. Phi29 amplification induces copy number biases and unamplified material remains the gold standard for copy number analysis. For accurate results using Phi29 amplification, samples subjected to aCGH analysis should be combined with reference DNA amplified with the same method, using similar amounts of starting template..
Savage, K.
Lambros, M.B.
Robertson, D.
Jones, R.L.
Jones, C.
Mackay, A.
James, M.
Hornick, J.L.
Pereira, E.M.
Milanezi, F.
Fletcher, C.D.
Schmitt, F.C.
Ashworth, A.
Reis-Filho, J.S.
(2007). Caveolin 1 is overexpressed and amplified in a subset of basal-like and metaplastic breast carcinomas:: A morphologic, ultrastructural, immunohistochemical, and in situ hybridization analysis. Clinical cancer research,
Vol.13
(1),
pp. 90-12.
Di Palma, S.
Lambros, M.B.
Savage, K.
Jones, C.
Mackay, A.
Dexter, T.
Iravani, M.
Fenwick, K.
Ashworth, A.
Reis-Filho, J.S.
(2007). Oncocytic change in pleomorphic adenoma: molecular evidence in support of an origin in neoplastic cells. Journal of clinical pathology,
Vol.60
(5),
pp. 492-8.
full text
Natrajan, R.
Williams, R.D.
Grigoriadis, A.
Mackay, A.
Fenwick, K.
Ashworth, A.
Dome, J.S.
Grundy, P.E.
Pritchard-Jones, K.
Jones, C.
(2007). Delineation of a 1Mb breakpoint region at 1p13 in Wilms tumors by fine-tiling oligonucleotide array CGH. Genes chromosomes cancer,
Vol.46
(6),
pp. 607-615.
show abstract
Wilms tumor karyotypes frequently exhibit recurrent, large-scale chromosomal imbalances, among the most common of which are concurrent loss of 1p and gain of 1q. We have previously identified a novel breakpoint at 1p13 by 1 Mb-spaced array CGH, and have now undertaken a fine-tiling oligonucleotide array approach to map the region accurately in four tumors exhibiting rearrangements at this locus. The use of a 10 bp-spaced platform revealed that all four tumors in fact harbored different breakpoints, which targeted intragenic sequences in PHTF1, DCLRE1B, and NRAS, and an intergenic region immediately downstream of TRIM33. All four genes and breakpoints were within the 1.78 Mb intervals identified by the genome-wide BAC arrays. The precise breakpoint interval was in each case mapped to a 200-1,200 bp region and was confirmed for one case to lie within intron 3 of DCLRE1B by quantitative PCR. Analysis of local genome architecture revealed no convincing conservation of repetitive sequences or specific translocation/recombination-associated elements within the breakpoint regions. This study highlights the power of fine-tiling oligonucleotide arrays to delineate breakpoint regions identified by genome-wide screens..
Jones, C.
Rodriguez-Pinilla, M.
Lambros, M.
Bax, D.
Messahel, B.
Vujanic, G.M.
Reis-Filho, J.S.
Pritchard-Jones, K.
(2007). c-KIT overexpression, without gene amplification and mutation, in paediatric renal tumours. J clin pathol,
Vol.60
(11),
pp. 1226-1231.
show abstract
full text
AIMS: To investigate the presence and prognostic relevance of KIT expression in paediatric renal tumours, and to determine whether receptor overexpression is associated with gene amplification and/or mutation. METHODS: Immunohistochemistry without antigen retrieval for CD117 was carried out on tissue microarrays consisting of 274 Wilms' tumours, 13 clear cell sarcomas of the kidney (CCSK), 10 mesoblastic nephromas (MN), and 7 rhabdoid tumours of the kidney (RTK). In addition, gene copy number was investigated by chromogenic in situ hybridisation (CISH), and overexpressing tumours were sequenced for KIT mutations in exons 9, 11, 13 and 17. RESULTS: Only 8/200 (4.0%) Wilms' tumours exhibited any degree of moderate-strong KIT staining in any of their assessable cell types. This small group of KIT-positive tumours had a shorter time to relapse (p = 0.0044, log-rank test). There were no positive MNs or RTKs; however 3/11 (27.3%) CCSKs were strongly positive, with an additional two cases weakly reactive. No cases exhibited gene amplification or mutation. CONCLUSIONS: KIT overexpression in rare in Wilms' tumours, although does appear to confer a worse prognosis, in particular for patients primarily treated with preoperative chemotherapy. CCSKs are associated with an increased expression of KIT, however, in the absence of gene amplification and/or activating mutation. The potential of anti-KIT therapeutic strategies in the treatment of paediatric renal tumours appears to be limited..
Pierga, J.-.
Reis-Filho, J.S.
Cleator, S.J.
Dexter, T.
Mackay, A.
Simpson, P.
Fenwick, K.
Iravani, M.
Salter, J.
Hills, M.
Jones, C.
Ashworth, A.
Smith, I.E.
Powles, T.
Dowsett, M.
(2007). Microarray-based comparative genomic hybridisation of breast cancer patients receiving neoadjuvant chemotherapy. Br j cancer,
Vol.96
(2),
pp. 341-351.
show abstract
We analysed the molecular genetic profiles of breast cancer samples before and after neoadjuvant chemotherapy with combination doxorubicin and cyclophosphamide (AC). DNA was obtained from microdissected frozen breast core biopsies from 44 patients before chemotherapy. Additional samples were obtained before the second course of chemotherapy (D21) and after the completion of the treatment (surgical specimens) in 17 and 21 patients, respectively. Microarray-based comparative genome hybridisation was performed using a platform containing approximately 5800 bacterial artificial chromosome clones (genome-wide resolution: 0.9 Mb). Analysis of the 44 pretreatment biopsies revealed that losses of 4p, 4q, 5q, 12q13.11-12q13.12, 17p11.2 and 17q11.2; and gains of 1p, 2p, 7q, 9p, 11q, 19p and 19q were significantly associated with oestrogen receptor negativity. 16q21-q22.1 losses were associated with lobular and 8q24 gains with ductal types. Losses of 5q33.3-q4 and 18p11.31 and gains of 6p25.1-p25.2 and Xp11.4 were associated with HER2 amplification. No correlations between DNA copy number changes and clinical response to AC were found. Microarray-based comparative genome hybridisation analysis of matched pretreatment and D21 biopsies failed to identify statistically significant differences, whereas a comparison between matched pretreatment and surgical samples revealed a statistically significant acquired copy number gain on 11p15.2-11p15.5. The modest chemotherapy-driven genomic changes, despite profound loss of cell numbers, suggest that there is little therapeutic selection of resistant non-modal cell lineages..
Fulford, L.G.
Reis-Filho, J.S.
Ryder, K.
Jones, C.
Gillett, C.E.
Hanby, A.
Easton, D.
Lakhani, S.R.
(2007). Basal-like grade III invasive ductal carcinoma of the breast: patterns of metastasis and long-term survival. Breast cancer res,
Vol.9
(1),
p. R4.
show abstract
INTRODUCTION: Cytokeratin (CK) 14, one of several markers expressed in normal myoepithelial/basal cells, is also expressed in a proportion of breast carcinomas. Previous studies have suggested that expression of such 'basal' markers predicts different biological behaviour, with more frequent lung and brain metastases and poorer prognosis than other carcinomas. METHODS: We performed CK14 immunohistochemistry on 443 grade III invasive ductal carcinomas with extended clinical follow-up (mean 116 months), and we correlated CK14 immunopositivity (basal-like phenotype) with clinicopathological criteria. RESULTS: Eighty-eight of 443 (20%) tumours showed CK14 expression. CK14-positive tumours were more likely to be oestrogen receptor-negative (p < 0.0001) and axillary node-negative (p = 0.001) than were CK14-negative cases. CK14-positive cases developed less bone and liver metastases (hazard ratio [HR] 0.49, p = 0.01, and HR 0.53, p = 0.035, respectively) but more frequent brain metastases (HR 1.92, p = 0.051). In patients without metastatic disease, disease-free survival in CK14-positive cases was significantly better than in CK14-negative cases (HR 0.65, p = 0.005). In patients with metastatic disease, however, CK14 positivity was associated with a poorer prognosis (HR 1.84, p = 0.001). The overall survival in CK14-positive and -negative patients was similar at 5 years (60% and 59%, respectively), but the long-term survival was better in CK14-positive patients (HR 0.69, p = 0.02). CONCLUSION: These results demonstrate that basal-like tumours differ in their biological behaviour from other tumours, with a distinct pattern of metastatic spread. Compared to other grade III tumours, basal-like tumours appear to have a relatively good long-term survival but survival after metastases is poor..
Natrajan, R.
Reis-Filho, J.S.
Little, S.E.
Messahel, B.
Brundler, M.-.
Dome, J.S.
Grundy, P.E.
Vujanic, G.M.
Pritchard-Jones, K.
Jones, C.
(2006). Blastemal expression of type I insulin-like growth factor receptor in Wilms' tumors is driven by increased copy number and correlates with relapse. Cancer res,
Vol.66
(23),
pp. 11148-11155.
show abstract
Most Wilms' tumors are of low stage, favorable histology, and have a high likelihood of cure with current multimodal therapy. Despite this, there remains a group of patients whose tumors recur for whom intensive salvage regimens result in survival of only 50%. Fitting a Cox proportional hazards model to microarray-based comparative genomic hybridization (aCGH) data on 68 Wilms' tumor samples, we identified a significant correlation between increased copy number at chromosome 15q26.3 insulin-like growth factor I receptor (IGFIR) and tumor relapse (adjusted P = 0.014). Wilms' tumors (13%) exhibited a low-level gain corresponding to three to four copies of the gene by aCGH analysis, 9 of 10 of which exhibited high IGFIR mRNA levels. Although IGFIR protein expression was restricted to the epithelial cells of fetal kidney and Wilms' tumors in most cases, 12% of tumors were also found to express IGFIR in the blastemal compartment. Blastemal IGFIR protein expression was associated with an increased copy number and a shorter relapse-free survival time (P = 0.027, log-rank test). In addition to the membrane localization, IGFIR was localized to the perinuclear region of the blastemal cells in 6% of Wilms' tumors. These data provide evidence that an increase in IGFIR gene copy number results in aberrant expression in the blastemal compartment of some Wilms' tumors and is associated with an adverse outcome in these patients. These findings suggest the possibility of use of targeted agents in the therapy of these children..
Little, S.E.
Vuononvirta, R.
Reis-Filho, J.S.
Natrajan, R.
Iravani, M.
Fenwick, K.
Mackay, A.
Ashworth, A.
Pritchard-Jones, K.
Jones, C.
(2006). Array CGH using whole genome amplification of fresh-frozen and formalin-fixed, paraffin-embedded tumor DNA. Genomics,
Vol.87
(2),
pp. 298-306.
show abstract
The ability to utilize formalin-fixed, paraffin-embedded (FFPE) archival specimens reliably for high-resolution molecular genetic analysis would be of immense practical application in the study of human disease. We have evaluated the ability of the GenomePlex whole genome amplification (WGA) kit to amplify frozen and FFPE tissue for use in array CGH (aCGH). GenomePlex gave highly representative data compared with unamplified controls both from frozen material (Pearson's R(2) = 0.898) and from FFPE (R(2) = 0.883). Artifactual amplification observed using DOP-PCR at chromosomes 1p, 3, 13q, and 16p was not seen with GenomePlex. Highly reproducible aCGH profiles were obtained using as little as 5 ng starting material from FFPE (R(2) = 0.918). This WGA method should readily lend itself to the determination of DNA copy number alterations from small fresh-frozen and FFPE clinical tumor specimens, although some care must be taken to optimize the DNA extraction procedure..
Lambros, M.B.
Simpson, P.T.
Jones, C.
Natrajan, R.
Westbury, C.
Steele, D.
Savage, K.
Mackay, A.
Schmitt, F.C.
Ashworth, A.
Reis, J.S.
(2006). Unlocking pathology archives for molecular genetic studies:: a reliable method to generate probes for chromogenic and fluorescent in situ hybridization. Laboratory investigation,
Vol.86
(4),
pp. 398-11.
Reis, J.S.
Pinheiro, C.
Lambros, M.B.
Milanezi, F.
Carvalho, S.
Savage, K.
Simpson, P.T.
Jones, C.
Swift, S.
Mackay, A.
Reis, R.M.
Hornick, J.L.
Pereira, E.M.
Baltazar, F.
Fletcher, C.D.
Ashworth, A.
Lakhani, S.R.
Schmitt, F.C.
(2006). EGFR amplification and lack of activating mutations in metaplastic breast carcinomas. Journal of pathology,
Vol.209
(4),
pp. 445-9.
Natrajan, R.
Williams, R.D.
Hing, S.N.
Mackay, A.
Reis-Filho, J.S.
Fenwick, K.
Iravani, M.
Valgeirsson, H.
Grigoriadis, A.
Langford, C.F.
Dovey, O.
Gregory, S.G.
Weber, B.L.
Ashworth, A.
Grundy, P.E.
Pritchard-Jones, K.
Jones, C.
(2006). Array CGH profiling of favourable histology Wilms tumours reveals novel gains and losses associated with relapse. J pathol,
Vol.210
(1),
pp. 49-58.
show abstract
Despite the excellent survival of Wilms tumour patients treated with multimodality therapy, approximately 15% will suffer from tumour relapse, where response rates are markedly reduced. We have carried out microarray-based comparative genomic hybridisation on a series of 76 Wilms tumour samples, enriched for cases which recurred, to identify changes in DNA copy number associated with clinical outcome. Using 1Mb-spaced genome-wide BAC arrays, the most significantly different genomic changes between favourable histology tumours that did (n = 37), and did not (n = 39), subsequently relapse were gains on 1q, and novel deletions at 12q24 and 18q21. Further relapse-associated loci included losses at 1q32.1, 2q36.3-2q37.1, and gain at 13q31. 1q gains correlated strongly with loss of 1p and/or 16q. In 3 of 11 cases with concurrent 1p(-)/1q(+), a breakpoint was identified at 1p13. Multiple low-level sub-megabase gains along the length of 1q were identified using chromosome 1 tiling-path arrays. One such recurrent region at 1q22-q23.1 included candidate genes RAB25, NES, CRABP2, HDGF and NTRK1, which were screened for mRNA expression using quantitative RT-PCR. These data provide a high-resolution catalogue of genomic copy number changes in relapsing favourable histology Wilms tumours..
Natrajan, R.
Little, S.E.
Reis-Filho, J.S.
Hing, L.
Messahel, B.
Grundy, P.E.
Dome, J.S.
Schneider, T.
Vujanic, G.M.
Pritchard-Jones, K.
Jones, C.
(2006). Amplification and overexpression of CACNA1E correlates with relapse in favorable histology Wilms' tumors. Clin cancer res,
Vol.12
(24),
pp. 7284-7293.
show abstract
PURPOSE: The most well established molecular markers of poor outcome in Wilms' tumor are loss of heterozygosity at chromosomes 1p and/or 16q, although to date no specific genes at these loci have been identified. We have previously shown a link between genomic gain of chromosome 1q and tumor relapse and sought to further elucidate the role of genes on 1q in treatment failure. EXPERIMENTAL DESIGN: Microarray-based comparative genomic hybridization identified a microamplification harboring a single gene (CACNA1E) at 1q25.3 in 6 of 76 (7.9%) Wilms' tumors, correlating with a shorter relapse-free survival (P = 0.0044, log-rank test). Further characterization of this gene was carried out by measuring mRNA and protein expression as well as stable transfection of HEK293 cells. RESULTS: Overexpression of the CACNA1E transcript was associated with DNA copy number (P = 0.0204, ANOVA) and tumor relapse (P = 0.0851, log-rank test). Immunohistochemistry against the protein product Ca(V)2.3 revealed expression localized to the apical membrane in the distal tubules of normal kidney but not to the metanephric blastemal cells of fetal kidney from which Wilms' tumors arise. Nuclear localization in 99 of 160 (61.9%) Wilms' tumor cases correlated with a reduced relapse-free survival, particularly in cases treated with preoperative chemotherapy (P = 0.009, log-rank test). Expression profiling of stably transfected HEK293 cells revealed specific up-regulation of the immediate early response genes EGR1/EGR2/EGR3 and FOS/FOSB, mediated by activation of the MEK/ERK5/Nur77 pathway. CONCLUSIONS: These data identify a unique genetic aberration with direct clinical relevance in Wilms' tumor relapse and provide evidence for a potential novel mechanism of treatment resistance in these tumors..
Reis-Filho, J.S.
Simpson, P.T.
Turner, N.C.
Lambros, M.B.
Jones, C.
Mackay, A.
Grigoriadis, A.
Sarrio, D.
Savage, K.
Dexter, T.
Iravani, M.
Fenwick, K.
Weber, B.
Hardisson, D.
Schmitt, F.C.
Palacios, J.
Lakhani, S.R.
Ashworth, A.
(2006). FGFR1 emerges as a potential therapeutic target for lobular breast carcinomas. Clinical cancer research,
Vol.12
(22),
pp. 6652-11.
Simpson, P.
Jones, C.
Mackay, A.
Lakhani, S.R.
(2006). Gene expression analysis using filter cDNA microarrays. Methods mol med,
Vol.120,
pp. 415-424.
show abstract
The analysis of gene expression patterns by filter-based complementary (c)DNA microarray remains an important technique in the molecular biology laboratory, despite the development of large-scale cDNA microarray analysis (see Chapter 27). This chapter provides an overview of the methods necessary to carry out the production of membrane-based cDNA arrays and the subsequent synthesis and hybridization of radiolabeled cDNA probes..
Jones, C.
Simpson, P.
Mackay, A.
Lakhani, S.R.
(2006). Expression profiling using cDNA microarrays. Methods mol med,
Vol.120,
pp. 403-414.
show abstract
Microarray technology is revolutionizing the assessment of gene expression in human disease. Coupled with the publication of the human genome, it is becoming possible to measure the relevant levels of all 30,000-40,000 human genes in a single experiment. The applications of these methods to breast cancer research are helping us to both further our understanding of the biology of mammary tumorigenesis, as well as providing novel subclassifications of breast cancer of direct clinical relevance. Unbiased RNA amplification techniques are allow the study of small biopsy samples as well as archival pathology specimens, increasing our molecular understanding, and providing real hope for the clinical application of gene expression profiling..
Simpson, P.T.
Gale, T.
Reis-Filho, J.S.
Jones, C.
Parry, S.
Sloane, J.P.
Hanby, A.
Pinder, S.E.
Lee, A.H.
Humphreys, S.
Ellis, I.O.
Lakhani, S.R.
(2005). Columnar cell lesions of the breast: the missing link in breast cancer progression? A morphological and molecular analysis. Am j surg pathol,
Vol.29
(6),
pp. 734-746.
show abstract
Columnar cell lesions (CCLs) of the breast are a spectrum of lesions that have posed difficulties to pathologists for many years, prompting discussion concerning their biologic and clinical significance. We present a study of CCL in context with hyperplasia of usual type (HUT) and the more advanced lesions ductal carcinoma in situ (DCIS) and invasive ductal carcinoma. A total of 81 lesions from 18 patients were subjected to a comprehensive morphologic review based upon a modified version of Schnitt's classification system for CCL, immunophenotypic analysis (estrogen receptor [ER], progesterone receptor [PgR], Her2/neu, cytokeratin 5/6 [CK5/6], cytokeratin 14 [CK14], E-cadherin, p53) and for the first time, a whole genome molecular analysis by comparative genomic hybridization. Multiple CCLs from 3 patients were studied in particular detail, with topographic information and/or showing a morphologic spectrum of CCL within individual terminal duct lobular units. CCLs were ER and PgR positive, CK5/6 and CK14 negative, exhibit low numbers of genetic alterations and recurrent 16q loss, features that are similar to those of low grade in situ and invasive carcinoma. The molecular genetic profiles closely reflect the degree of proliferation and atypia in CCL, indicating some of these lesions represent both a morphologic and molecular continuum. In addition, overlapping chromosomal alterations between CCL and more advanced lesions within individual terminal duct lobular units suggest a commonality in molecular evolution. These data further support the hypothesis that CCLs are a nonobligate, intermediary step in the development of some forms of low grade in situ and invasive carcinoma..
Reis-Filho, J.S.
Simpson, P.T.
Jones, C.
Steele, D.
Mackay, A.
Iravani, M.
Fenwick, K.
Valgeirsson, H.
Lambros, M.
Ashworth, A.
Palacios, J.
Schmitt, F.
Lakhani, S.R.
(2005). Pleomorphic lobular carcinoma of the breast: role of comprehensive molecular pathology in characterization of an entity. J pathol,
Vol.207
(1),
pp. 1-13.
show abstract
Immunohistochemical analysis of E-cadherin has changed the way lobular neoplasia is perceived. It has helped to classify difficult cases of carcinoma in situ with indeterminate features and led to the identification of new variants of lobular carcinoma. Pleomorphic lobular carcinoma (PLC) and pleomorphic lobular carcinoma in situ (PLCIS), recently described variants of invasive and in situ classic lobular carcinoma, are reported to be associated with more aggressive clinical behaviour. Although PLC/PLCIS show morphological features of classic lobular neoplasia and lack E-cadherin expression, it is still unclear whether these lesions evolve through the same genetic pathway as lobular carcinomas or are high-grade ductal neoplasms that have lost E-cadherin. Here we have analysed a case of extensive PLCIS and invasive PLC associated with areas of E-cadherin-negative carcinoma in situ with indeterminate features, using immunohistochemistry, chromogenic in situ hybridization, high-resolution comparative genomic hybridization (CGH) and array-based CGH. We observed that all lesions lacked E-cadherin and beta-catenin and showed gain of 1q and loss of 16q, features that are typical of lobular carcinomas but are not seen in high-grade ductal lesions. In addition, amplifications of c-myc and HER2 were detected in the pleomorphic components, which may account for the high-grade features in this case and the reported aggressive clinical behaviour of these lesions. Taken together, these data suggest that at least some PLCs may evolve from the same precursor or through the same genetic pathway as classic lobular carcinomas..
Jones, C.
Mackay, A.
Grigoriadis, A.
Cossu, A.
Reis-Filho, J.S.
Fulford, L.
Dexter, T.
Davies, S.
Bulmer, K.
Ford, E.
Parry, S.
Budroni, M.
Palmieri, G.
Neville, A.M.
O'Hare, M.J.
Lakhani, S.R.
(2004). Expression profiling of purified normal human luminal and myoepithelial breast cells: identification of novel prognostic markers for breast cancer. Cancer res,
Vol.64
(9),
pp. 3037-3045.
show abstract
The normal duct-lobular system of the breast is lined by two epithelial cell types, inner luminal secretory cells and outer contractile myoepithelial cells. We have generated comprehensive expression profiles of the two normal cell types, using immunomagnetic cell separation and gene expression microarray analysis. The cell-type specificity was confirmed at the protein level by immunohistochemistry in normal breast tissue. New prognostic markers for survival were identified when the luminal- and myoepithelial-specific molecules were evaluated on breast tumor tissue microarrays. Nuclear expression of luminal epithelial marker galectin 3 correlated with a shorter overall survival in these patients, and the expression of SPARC (osteonectin), a myoepithelial marker, was an independent marker of poor prognosis in breast cancers as a whole. These data provide a framework for the interpretation of breast cancer molecular profiling experiments, the identification of potential new diagnostic markers, and development of novel indicators of prognosis..
Simpson, P.T.
Gale, T.
Reis, J.S.
Jones, C.
Parry, S.
Steele, D.
Cossu, A.
Budroni, M.
Palmieri, G.
Lakhani, S.R.
(2004). Distribution and significance of 14-3-3 sigma, a novel myoepithelial marker, in normal, benign, and malignant breast tissue. Journal of pathology,
Vol.202
(3),
pp. 274-12.
Angele, S.
Jones, C.
Reis, J.S.
Fulford, L.G.
Treilleux, I.
Lakhani, S.R.
Hall, J.
(2004). Expression of ATM, p53, and the MRE11-Rad50-NBS1 complex in myoepithelial cells from benign and malignant proliferations of the breast. Journal of clinical pathology,
Vol.57
(11),
pp. 1179-6.
Jones, C.
Pritchard-Jones, K.
(2004). MIB-1 and p27Kip1 expression in nephroblastoma. Clin cancer res,
Vol.10
(22),
pp. 7785-7786.
Little, S.E.
Hanks, S.P.
King-Underwood, L.
Jones, C.
Rapley, E.A.
Rahman, N.
Pritchard-Jones, K.
(2004). Frequency and heritability of WT1 mutations in nonsyndromic Wilms' tumor patients: a UK Children's Cancer Study Group Study. J clin oncol,
Vol.22
(20),
pp. 4140-4146.
show abstract
PURPOSE: Constitutional WT1 mutations in patients with Wilms' tumor (WT) have specifically been associated with genitourinary abnormalities, such as cryptorchidism and hypospadias. We sought to ascertain the frequency and heritability of constitutional WT1 mutations in nonsyndromic WT patients. PATIENTS AND METHODS: Constitutional DNA from 282 patients treated at seven United Kingdom Children's Cancer Study Group centers was screened for WT1 mutations using heteroduplex analysis. Bidirectional sequencing was used to confirm the mutation and to analyze the corresponding parental DNA samples. RESULTS: Five different constitutional WT1 mutations were identified in six children. Mutations in four patients were confirmed to be de novo, and all five mutations are predicted to produce truncated protein. The WT1 mutation group had a young median age at diagnosis of 13.8 months, compared with 34.9 months in the group in whom no WT1 mutations were found; four were female and two were male; and all tumors were of favorable histology. The three tumors with known histologic subtype were stromal-predominant. Contrary to expectation, four of six mutations occurred in children with unilateral tumors without any associated genitourinary abnormality. CONCLUSION: Constitutional WT1 mutations occur with a low frequency (2.1%; 95% CI, 0.8% to 4.6%) in nonsyndromic WT patients. Most mutations occurred in children with unilateral WT without associated genitourinary abnormalities, creating difficulties in identifying individuals with germline mutations on phenotype alone. Two factors that may indicate that an individual is carrying a germline WT1 mutation are an early age of onset and stromal-predominant histology of the WT..
Jones, C.
Ford, E.
Gillett, C.
Ryder, K.
Merrett, S.
Reis, J.S.
Fulford, L.G.
Hanby, A.
Lakhani, S.R.
(2004). Molecular cytogenetic identification of subgroups of grade III invasive ductal breast carcinomas with different clinical outcomes. Clinical cancer research,
Vol.10
(18),
pp. 5988-10.
Mackay, A.
Jones, C.
Dexter, T.
Silva, R.L.
Bulmer, K.
Jones, A.
Simpson, P.
Harris, R.A.
Jat, P.S.
Neville, A.M.
Reis, L.F.
Lakhani, S.R.
O'Hare, M.J.
(2003). cDNA microarray analysis of genes associated with ERBB2 (HER2/neu) overexpression in human mammary luminal epithelial cells. Oncogene,
Vol.22
(17),
pp. 2680-9.
Jones, C.
Merrett, S.
Thomas, V.A.
Barker, T.H.
Lakhani, S.R.
(2003). Comparative genomic hybridization analysis of bilateral hyperplasia of usual type of the breast. Journal of pathology,
Vol.199
(2),
pp. 152-5.
Jones, C.
Tooze, R.
Lakhani, S.R.
(2003). Malignant adenomyoepithelioma of the breast metastasizing to the liver. Virchows archiv,
Vol.442
(5),
pp. 504-3.
Brown, R.S.
Edwards, J.
Bartlett, J.W.
Jones, C.
Dogan, A.
(2002). Routine acid decalcification of bone marrow samples can preserve DNA for FISH and CGH studies in metastatic prostate cancer. J histochem cytochem,
Vol.50
(1),
pp. 113-115.
show abstract
Production of paraffin-section material from tissue samples that contain bone requires decalcification. Techniques such as acidic decalcification or EDTA chelation are suitable methods. Acid decalcification is generally quicker than EDTA chelation but studies have suggested that it may result in hydrolysis of DNA. Here we show that limited acid decalcification (less than 24 hr) in 5% formic acid can preserve DNA sufficient for fluorescent in situ hybridization (FISH) or comparative genomic hybridization (CGH) and that prolonged 10% formic acid decalcification results in failure of FISH and only limited retrieval of DNA for CGH studies..
Leonard, N.
Chaggar, R.
Jones, C.
Takahashi, M.
Nikitopoulou, A.
Lakhani, S.R.
(2001). Loss of heterozygosity at cylindromatosis gene locus, CYLD, in sporadic skin adnexal tumours. Journal of clinical pathology,
Vol.54
(9),
pp. 689-4.
full text
Jones, C.
Damiani, S.
Wells, D.
Chaggar, R.
Lakhani, S.R.
Eusebi, V.
(2001). Molecular cytogenetic comparison of apocrine hyperplasia and apocrine carcinoma of the breast. Am j pathol,
Vol.158
(1),
pp. 207-214.
show abstract
The relationship of apocrine metaplasia to invasive breast cancer is controversial. Different authors have reported that apocrine differentiation in proliferative lesions may be a risk factor, a precursor lesion, or have no association with malignancy. The aim of this study was to compare the genetic alterations in benign apocrine hyperplasia with apocrine ductal carcinoma in situ (DCIS) and invasive apocrine carcinomas of the breast using comparative genomic hybridization. The mean number of alterations in apocrine hyperplasia was 4.1 (n = 10) compared to 10.2 in apocrine DCIS (n = 10) and 14.8 (n = 4) in invasive carcinoma. The most common alterations in apocrine hyperplasia were gains of 2q, 13q, and 1p and losses of 1p, 17q, 22q, 2p, 10q, and 16q. Apocrine DCIS and invasive carcinomas showed gains of 1q, 2q, 1p, and losses of 1p, 22q, 17q, 12q, and 16q as their most common DNA copy number changes. Apocrine hyperplasia is considered to be a benign lesion and its relationship to invasive carcinoma remains unclear. Our data suggest that some apocrine hyperplasias may be clonal proliferations. The mean number of alterations are lower in apocrine hyperplasia, however the changes show considerable overlap with those identified in in situ and invasive apocrine carcinoma. These alterations are also commonly seen in nonapocrine breast cancer. The data are consistent with apocrine hyperplasia as a putative nonobligate precursor of apocrine carcinoma..
Jones, C.
Nonni, A.V.
Fulford, L.
Merrett, S.
Chaggar, R.
Eusebi, V.
Lakhani, S.R.
(2001). CGH analysis of ductal carcinoma of the breast with basaloid/myoepithelial cell differentiation. Br j cancer,
Vol.85
(3),
pp. 422-427.
show abstract
2-18% of ductal carcinoma-No Special Type (NST) are reported to express basal cell keratin 14 and such tumours may have a different metastatic pattern and prognosis. We performed immunohistochemistry for cytokeratins 19 (luminal) and 14 (basal) on 92 ductal carcinoma-NST. Those tumours showing CK14 expression were further characterized by immunohistochemistry for myoepithelial cell phenotype and analysed by comparative genomic hybridization. The 7 cases of ductal carcinoma-NST exhibiting a basal cell phenotype were all grade III tumours and showed a molecular cytogenetic profile similar to more conventional myoepithelial cell carcinomas. Therefore it appears that grade III invasive ductal carcinomas contain a subset of tumours with specific morphological and cytogenetic characteristics, and probably prognosis for the patient..
Leonard, N.
Chaggar, R.
Jones, C.
Takahashi, M.
Lakhani, S.R.
(2000). Loss of heterozygosity on chromosome 16q (cylindromatosis gene locus-Cyld1) in sporadic skin adenexal tumours. Journal of pathology,
Vol.190,
pp. 68A-1.
Jones, C.
Foschini, M.P.
Chaggar, R.
Lu, Y.J.
Wells, D.
Shipley, J.M.
Eusebi, V.
Lakhani, S.R.
(2000). Comparative genomic hybridization analysis of myoepithelial carcinoma of the breast. Laboratory investigation,
Vol.80
(6),
pp. 831-6.
Jones, C.
Foschini, M.P.
Chaggar, R.
Lu, Y.-.
Wells, D.
Shipley, J.M.
Eusebi, V.
Lakhani, S.R.
(2000). Comparative genomic hybridisation analysis of myoepithelial carcinoma of the breast. Breast cancer research : bcr,
Vol.2
(Suppl 1),
pp. P8.06-P8.06.
Bignell, G.R.
Warren, W.
Seal, S.
Takahashi, M.
Rapley, E.
Barfoot, R.
Green, H.
Brown, C.
Biggs, P.J.
Lakhani, S.R.
Jones, C.
Hansen, J.
Blair, E.
Hofmann, B.
Siebert, R.
Turner, G.
Evans, D.G.
Schrander-Stumpel, C.
Beemer, F.A.
van Den Ouweland, A.
Halley, D.
Delpech, B.
Cleveland, M.G.
Leigh, I.
Leisti, J.
Rasmussen, S.
(2000). Identification of the familial cylindromatosis tumour-suppressor gene. Nat genet,
Vol.25
(2),
pp. 160-165.
show abstract
Familial cylindromatosis is an autosomal dominant genetic predisposition to multiple tumours of the skin appendages. The susceptibility gene (CYLD) has previously been localized to chromosome 16q and has the genetic attributes of a tumour-suppressor gene (recessive oncogene). Here we have identified CYLD by detecting germline mutations in 21 cylindromatosis families and somatic mutations in 1 sporadic and 5 familial cylindromas. All mutations predict truncation or absence of the encoded protein. CYLD encodes three cytoskeletal-associated-protein-glycine-conserved (CAP-GLY) domains, which are found in proteins that coordinate the attachment of organelles to microtubules. CYLD also has sequence homology to the catalytic domain of ubiquitin carboxy-terminal hydrolases (UCH)..
Payne, J.
Jones, C.
Lakhani, S.R.
Kortenkamp, A.
(2000). Improving the reproducibility of the MCF-7 cell proliferation assay for the detection of xenoestrogens. Sci total environ,
Vol.248
(1),
pp. 51-62.
show abstract
The MCF-7 cell proliferation assay is potentially a simple and highly reproducible tool for the identification of estrogenic compounds. However, its widespread use has been complicated by the lack of a standardised protocol, resulting in considerable inter-laboratory variability. We have explored the sources of variability both in relation to cell lines and test regimens and report on optimised procedures for the identification of estrogenic agents. Two supposedly identical MCF-7 parent cell lines (designated UCL and SOP), and the BUS subline were cultured according to an existing protocol, and responses to 17-estradiol (E2) assessed. Despite yielding almost identical EC50 values, the proliferative response varied widely between cell lines from 0.98-fold over controls (UCL) to 8.9-fold (BUS) indicating major differences between them. The underlying causes may be genetic, and to assess this we used comparative genomic hybridisation (CGH), a technique which allows the detection of DNA sequence copy number changes on a genome-wide scale. Although numerous similarities existed between the different cell lines, the least oestrogen-responsive line (MCF-7/UCL) exhibited the greatest number of cytogenetic changes, many of which were not seen in MCF-7/SOP cells. We suggest that care must be taken, therefore, when choosing a cell line for MCF-7 cell-based experiments. Selecting the MCF-7/SOP line for further work, we carried out a thorough and systematic optimisation of the MCF-7 cell proliferation assay, finding that a 72-h period in oestrogen-free medium before treatment strongly influenced the cells response to E2. With 1 nM E2, proliferation increased from 1.5-fold to 6.5-fold relative to vehicle-treated controls, a response similar to that seen with MCF-7/BUS cells in the E-SCREEN protocol devised by Soto et al. With parent MCF-7 cells, other laboratories have reported only 4.5-fold increases as maximal. Here we present evidence that the choice of cell line and culture conditions are crucial in determining test outcomes, and once chosen and adhered to the assay yields reproducible results..
Jones, C.
Kortenkamp, A.
(2000). RAPD library fingerprinting of bacterial and human DNA: applications in mutation detection. Teratog carcinog mutagen,
Vol.20
(2),
pp. 49-63.
show abstract
Random amplified polymorphic DNA (RAPD) fingerprinting is a modification of the polymerase chain reaction (PCR), which utilises a single, arbitrarily-chosen primer to amplify a number of fragments from a given template DNA to generate a discrete "fingerprint" when resolved by gel electrophoresis. Alterations by as little as a single base in the primer sequence lead to marked alterations in the fingerprints generated with a given template under optimised conditions. By inference, single base alterations in the genomic template DNA may also lead to changes in the RAPD fingerprints. We have examined this potential application to detect mutations in bacteria and cultured human cells. We have utilised Escherichia coli and human lymphoblastoid cell lines exposed to UV radiation, selected for by cellular mutation assays, and compared RAPD fingerprints of mutant and non-mutant samples. Polymorphisms became evident as the presence and/or absence of DNA fragments between the two samples. A dose-dependent increase in the number of polymorphic bands was seen with UV irradiation of E. coli. To a lesser degree, polymorphisms were also evident for human lymphoblastoid DNA. The possible underlying mechanisms for these alterations in fingerprints as a result of mutation(s) in the primer binding site(s) are discussed. The ability of RAPD fingerprinting to detect a mutant in a population of non-mutants is evaluated, and whilst the lack of sensitivity inherent in the technique precludes its use as a mutation screening assay, its potential for generation of mutant and non-mutant DNA probes for other mutation detection techniques may prove to be of great merit. Teratogenesis Carcinog. Mutagen. 20:49-63, 2000..
Jones, C.
Payne, J.
Wells, D.
Delhanty, J.D.
Lakhani, S.R.
Kortenkamp, A.
(2000). Comparative genomic hybridization reveals extensive variation among different MCF-7 cell stocks. Cancer genet cytogenet,
Vol.117
(2),
pp. 153-158.
show abstract
Comparative genomic hybridization (CGH) allows the detection of DNA sequence copy number changes on a genome-wide scale in a single hybridization reaction. The ability of CGH to be applied to formalin-fixed, paraffin-embedded tumor samples has lead to its widespread application in the cytogenetic analysis of archival material. When setting up CGH in the laboratory, rigorous control experiments must be carried out to ensure that the losses and gains are scored correctly. Groups interested in breast cancer frequently use the MCF-7 cell line as a positive control in these experiments, comparing the results to previously described genetic alterations. Here we present the results of CGH carried out with three stocks of MCF-7 cells. The cells differ widely in their proliferative response to 17-beta estradiol and show extensive variation in copy number changes affecting specific chromosomal regions. We suggest that care must be taken, therefore, when choosing a cell line as a positive control for CGH experiments..
Lakhani, S.R.
Chaggar, R.
Davies, S.
Jones, C.
Collins, N.
Odel, C.
Stratton, M.R.
O'Hare, M.J.
(1999). Genetic alterations in 'normal' luminal and myoepithelial cells of the breast. J pathol,
Vol.189
(4),
pp. 496-503.
show abstract
Chromosomal loci exhibiting loss of heterozygosity (LOH) at high frequency in invasive breast cancer have been investigated in 'normal' breast tissue from patients with carcinoma and from reduction mammoplasty specimens. Duct-lobular units dissected from paraffin-embedded tissues and 485 'normal' luminal and myoepithelial cell clones were studied. Overall, LOH was found in normal cells in 5/10 breast cancer cases and 1/3 reduction mammoplasty specimens. LOH was identified in normal cells adjacent to and distant from the tumour. In one case, all luminal and myoepithelial samples exhibited loss of the same allele on chromosome 13q. One case in which the patient had a germline truncating mutation in the BRCA1 gene exhibited LOH on 17q in 3/33 normal clones. One of these clones showed loss of wild-type allele indicating gene inactivation. This sample also had LOH at markers on chromosomes 11p and 13q. One of 93 clones from three reduction mammoplasties showed allele loss at a locus on chromosome 13q. The identification of LOH in breast lobules suggests that they may be clonal. The demonstration of genetic alteration in luminal and myoepithelial cells provides evidence for the presence of a common stem cell for the two epithelial cell types. LOH has been demonstrated in normal tissues near and away from the carcinoma, suggesting that genetic alterations are likely to be more heterogeneous and widespread than is currently envisaged, and probably occur very early in breast development. Homozygous deletion of BRCA1 per se does not appear to provide clonal advantage..
Kortenkamp, A.
Jones, C.
Baker, J.
(1997). Genotypic selection of mutated DNA sequences using mismatch cleavage analysis, a possible basis for novel mutation assays. Mutagenesis,
Vol.12,
pp. 335-338.
O'Brien, P.
Wang, G.
Earney, J.
Jones, C.
Kortenkamp, A.
(1995). Reactive chromium species potentially generated by welding fumes. Toxicol. environ. chem.,
Vol.49,
pp. 149-155.
Jones, C.
Greiner, B.
Wachter, F.
Terlouw, G.D.
Bechter, R.
(1993). An immunohistochemical investigation into the presence and localization of cytochrome P-450 isozymes in organogenesis-stage rat conceptal tissue. Toxicology in vitro,
Vol.7
(6),
pp. 685-693.
show abstract
To investigate the presence and localization of a variety of xenobiotic biotransforming isozymes of the cytochrome P-450 superfamily in the organogenesis-stage rat conceptal tissue, pregnant female rats were dosed with one of two inducing agents, 3-methylcholanthrene [3MC, 40 mg/kg body weight, ip, day 7 post coitum (pc)] and phenobarbital (PB, 40 mg/kg, ip, days 5, 6, 7 and 8 pc), or with their vehicles (3MC, olive oil; PB, 0.9% NaCl) as controls. The conceptuses were allowed to grow either in vivo, or in vitro, using the whole embryo culture system, from days 9.5 to 11.5 pc. The embryos and isolated visceral yolk sacs were submitted to immunohistochemical investigation using light microscopy. The livers of the dams served as positive controls. Polyclonal and monoclonal antibodies raised against a variety of cytochrome P-450 isozymes were used in the alkaline phosphatase-anti-alkaline phosphatase enzyme immune complex method. All pre-induced dam livers showed positive staining with all polyclonal and monoclonal antibodies tested. The presence of P450IA1 was detected in the visceral yolk sac of both ex vivo and cultured conceptuses, preinduced in utero with 3MC, with the appropriate polyclonal antibodies but not with the monoclonal antibodies. P450IIB1/2 was detected in the visceral yolk sac of both ex vivo and cultured conceptuses, pre-induced in utero by phenobarbital, with the appropriate polyclonal antibodies, but not with the monoclonal antibodies. No staining was seen in any embryo proper, with any vehicle-treated conceptal tissue, or with antibodies raised against P-450s IIE1, IIIA or IVA. Our results support the hypothesis that the organogenesis-stage rat conceptus contains, in the visceral yolk sac, a 3MC-inducible P-450 isozyme similar, but not identical, to adult IA1. They also provide evidence that a PB-inducible isozyme similar, but not identical, to adult IIB1/2, is present in the visceral yolk sac at this stage of conceptus development..
Viana-Pereira, M.
Lopes, J.M.
Little, S.
Milanezi, F.
Basto, D.
Pardal, F.
Jones, C.
Reis, R.M.
Analysis of EGFR overexpression, EGFR gene amplification and the EGFRvIII mutation in Portuguese high-grade gliomas. Anticancer research,
Vol.28
(2A),
pp. 913-8.
Jamin, Y.
Tucker, E.R.
Poon, E.S.
Popov, S.
Vaughan, L.
Boult, J.K.
Webber, H.
Hallsworth, A.
Baker, L.C.
Jones, C.
Koh, D.-.
Pearson, A.D.
Chesler, L.
Robinson, S.P.
Evaluation of clinically translatable magnetic resonance imaging biomarkers of therapeutic response in the TH-MYCN transgenic mouse model of neuroblastoma. Radiology,
.
full text