Network


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

Hotspot


Dive into the research topics where Yael P. Mosse is active.

Publication


Featured researches published by Yael P. Mosse.


Nature | 2008

Identification of ALK as a major familial neuroblastoma predisposition gene

Yael P. Mosse; Marci Laudenslager; Luca Longo; Kristina A. Cole; Andrew K.W. Wood; Edward F. Attiyeh; Michael J. Laquaglia; Rachel Sennett; Jill Lynch; Patrizia Perri; Genevieve Laureys; Frank Speleman; Cecilia Kim; Cuiping Hou; Hakon Hakonarson; Ali Torkamani; Nicholas J. Schork; Garrett M. Brodeur; Gian Paolo Tonini; Eric Rappaport; Marcella Devoto; John M. Maris

Neuroblastoma is a childhood cancer that can be inherited, but the genetic aetiology is largely unknown. Here we show that germline mutations in the anaplastic lymphoma kinase (ALK) gene explain most hereditary neuroblastomas, and that activating mutations can also be somatically acquired. We first identified a significant linkage signal at chromosome bands 2p23–24 using a whole-genome scan in neuroblastoma pedigrees. Resequencing of regional candidate genes identified three separate germline missense mutations in the tyrosine kinase domain of ALK that segregated with the disease in eight separate families. Resequencing in 194 high-risk neuroblastoma samples showed somatically acquired mutations in the tyrosine kinase domain in 12.4% of samples. Nine of the ten mutations map to critical regions of the kinase domain and were predicted, with high probability, to be oncogenic drivers. Mutations resulted in constitutive phosphorylation, and targeted knockdown of ALK messenger RNA resulted in profound inhibition of growth in all cell lines harbouring mutant or amplified ALK, as well as in two out of six wild-type cell lines for ALK. Our results demonstrate that heritable mutations of ALK are the main cause of familial neuroblastoma, and that germline or acquired activation of this cell-surface kinase is a tractable therapeutic target for this lethal paediatric malignancy.


Nature Genetics | 2013

The genetic landscape of high-risk neuroblastoma

Trevor J. Pugh; Olena Morozova; Edward F. Attiyeh; Shahab Asgharzadeh; Jun S. Wei; Daniel Auclair; Scott L. Carter; Kristian Cibulskis; Megan Hanna; Adam Kiezun; Jaegil Kim; Michael S. Lawrence; Lee Lichenstein; Aaron McKenna; Chandra Sekhar Pedamallu; Alex H. Ramos; Erica Shefler; Andrey Sivachenko; Carrie Sougnez; Chip Stewart; Adrian Ally; Inanc Birol; Readman Chiu; Richard Corbett; Martin Hirst; Shaun D. Jackman; Baljit Kamoh; Alireza Hadj Khodabakshi; Martin Krzywinski; Allan Lo

Neuroblastoma is a malignancy of the developing sympathetic nervous system that often presents with widespread metastatic disease, resulting in survival rates of less than 50%. To determine the spectrum of somatic mutation in high-risk neuroblastoma, we studied 240 affected individuals (cases) using a combination of whole-exome, genome and transcriptome sequencing as part of the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) initiative. Here we report a low median exonic mutation frequency of 0.60 per Mb (0.48 nonsilent) and notably few recurrently mutated genes in these tumors. Genes with significant somatic mutation frequencies included ALK (9.2% of cases), PTPN11 (2.9%), ATRX (2.5%, and an additional 7.1% had focal deletions), MYCN (1.7%, causing a recurrent p.Pro44Leu alteration) and NRAS (0.83%). Rare, potentially pathogenic germline variants were significantly enriched in ALK, CHEK2, PINK1 and BARD1. The relative paucity of recurrent somatic mutations in neuroblastoma challenges current therapeutic strategies that rely on frequently altered oncogenic drivers.


Lancet Oncology | 2013

Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: a Children's Oncology Group phase 1 consortium study.

Yael P. Mosse; Megan S. Lim; Stephan D. Voss; Keith D. Wilner; Katherine Ruffner; Julie Laliberte; Delphine Rolland; Frank M. Balis; John M. Maris; Brenda Weigel; Ashish M. Ingle; Charlotte H. Ahern; Peter C. Adamson; Susan M. Blaney

BACKGROUND Various human cancers have ALK gene translocations, amplifications, or oncogenic mutations, such as anaplastic large-cell lymphoma, inflammatory myofibroblastic tumours, non-small-cell lung cancer (NSCLC), and neuroblastoma. Therefore, ALK inhibition could be a useful therapeutic strategy in children. We aimed to determine the safety, recommended phase 2 dose, and antitumour activity of crizotinib in children with refractory solid tumours and anaplastic large-cell lymphoma. METHODS In this open-label, phase 1 dose-escalation trial, patients older than 12 months and younger than 22 years with measurable or evaluable solid or CNS tumours, or anaplastic large-cell lymphoma, refractory to therapy and for whom there was no known curative treatment were eligible. Crizotinib was given twice daily without interruption. Six dose levels (100, 130, 165, 215, 280, 365 mg/m(2) per dose) were assessed in the dose-finding phase of the study (part A1), which is now completed. The primary endpoint was to estimate the maximum tolerated dose, to define the toxic effects of crizotinib, and to characterise the pharmacokinetics of crizotinib in children with refractory cancer. Additionally, patients with confirmed ALK translocations, mutations, or amplification (part A2 of the study) or neuroblastoma (part A3) could enrol at one dose level lower than was currently given in part A1. We assessed ALK genomic status in tumour tissue and used quantitative RT-PCR to measure NPM-ALK fusion transcript in bone marrow and blood samples of patients with anaplastic large-cell lymphoma. All patients who received at least one dose of crizotinib were evaluable for response; patients completing at least one cycle of therapy or experiencing dose limiting toxicity before that were considered fully evaluable for toxicity. This study is registered with ClinicalTrials.gov, NCT00939770. FINDINGS 79 patients were enrolled in the study from Oct 2, 2009, to May 31, 2012. The median age was 10.1 years (range 1.1-21.4); 43 patients were included in the dose escalation phase (A1), 25 patients in part A2, and 11 patients in part A3. Crizotinib was well tolerated with a recommended phase 2 dose of 280 mg/m(2) twice daily. Grade 4 adverse events in cycle 1 were neutropenia (two) and liver enzyme elevation (one). Grade 3 adverse events that occurred in more than one patient in cycle 1 were lymphopenia (two), and neutropenia (eight). The mean steady state peak concentration of crizotinib was 630 ng/mL and the time to reach this peak was 4 h (range 1-6). Objective tumour responses were documented in 14 of 79 patients (nine complete responses, five partial responses); and the anti-tumour activity was enriched in patients with known activating ALK aberrations (eight of nine with anaplastic large-cell lymphoma, one of 11 with neuroblastoma, three of seven with inflammatory myofibroblastic tumour, and one of two with NSCLC). INTERPRETATION The findings suggest that a targeted inhibitor of ALK has antitumour activity in childhood malignancies harbouring ALK translocations, particularly anaplastic large-cell lymphoma and inflammatory myofibroblastic tumours, and that further investigation in the subset of neuroblastoma harbouring known ALK oncogenic mutations is warranted. FUNDING Pfizer and National Cancer Institute grant to the Childrens Oncology Group.


Nature | 2009

Copy number variation at 1q21.1 associated with neuroblastoma

Sharon J. Diskin; Cuiping Hou; Joseph T. Glessner; Edward F. Attiyeh; Marci Laudenslager; Kristopher R. Bosse; Kristina A. Cole; Yael P. Mosse; Andrew C. Wood; Jill Lynch; Katlyn Pecor; Maura Diamond; Cynthia Winter; Kai Wang; Cecilia Kim; Elizabeth A. Geiger; Patrick McGrady; Alexandra I. F. Blakemore; Wendy B. London; Tamim H. Shaikh; Jonathan P. Bradfield; Struan F. A. Grant; Hongzhe Li; Marcella Devoto; Eric R. Rappaport; Hakon Hakonarson; John M. Maris

Common copy number variations (CNVs) represent a significant source of genetic diversity, yet their influence on phenotypic variability, including disease susceptibility, remains poorly understood. To address this problem in human cancer, we performed a genome-wide association study of CNVs in the childhood cancer neuroblastoma, a disease in which single nucleotide polymorphism variations are known to influence susceptibility. We first genotyped 846 Caucasian neuroblastoma patients and 803 healthy Caucasian controls at ∼550,000 single nucleotide polymorphisms, and performed a CNV-based test for association. We then replicated significant observations in two independent sample sets comprised of a total of 595 cases and 3,357 controls. Here we describe the identification of a common CNV at chromosome 1q21.1 associated with neuroblastoma in the discovery set, which was confirmed in both replication sets. This CNV was validated by quantitative polymerase chain reaction, fluorescent in situ hybridization and analysis of matched tumour specimens, and was shown to be heritable in an independent set of 713 cancer-free parent–offspring trios. We identified a previously unknown transcript within the CNV that showed high sequence similarity to several neuroblastoma breakpoint family (NBPF) genes and represents a new member of this gene family (NBPF23). This transcript was preferentially expressed in fetal brain and fetal sympathetic nervous tissues, and the expression level was strictly correlated with CNV state in neuroblastoma cells. These data demonstrate that inherited copy number variation at 1q21.1 is associated with neuroblastoma and implicate a previously unknown neuroblastoma breakpoint family gene in early tumorigenesis of this childhood cancer.


Molecular Cancer Research | 2008

A Functional Screen Identifies miR-34a as a Candidate Neuroblastoma Tumor Suppressor Gene

Kristina A. Cole; Edward F. Attiyeh; Yael P. Mosse; Michael J. Laquaglia; Sharon J. Diskin; Garrett M. Brodeur; John M. Maris

MicroRNAs are small noncoding RNAs that have critical roles in regulating a number of cellular functions through transcriptional silencing. They have been implicated as oncogenes and tumor suppressor genes (oncomirs) in several human neoplasms. We used an integrated genomics and functional screening strategy to identify potential oncomirs in the pediatric neoplasm neuroblastoma. We first identified microRNAs that map within chromosomal regions that we and others have defined as frequently deleted (1p36, 3p22, and 11q23-24) or gained (17q23) in high-risk neuroblastoma. We then transiently transfected microRNA precursor mimics or inhibitors into a panel of six neuroblastoma cell lines that we characterized for these genomic aberrations. The majority of transfections showed no phenotypic effect, but the miR-34a (1p36) and miR-34c (11q23) mimics showed dramatic growth inhibition in cell lines with 1p36 hemizygous deletion. In contrast, there was no growth inhibition by these mimics in cell lines without 1p36 deletions. Quantitative reverse transcription-PCR showed a perfect correlation of absent miR-34a expression in cell lines with a 1p36 aberration and phenotypic effect after mimetic add-back. Expression of miR-34a was also decreased in primary tumors (n = 54) with 1p36 deletion (P = 0.009), but no mutations were discovered in resequencing of the miR-34a locus in 30 neuroblastoma cell lines. Flow cytometric time series analyses showed that the likely mechanism of miR-34a growth inhibition is through cell cycle arrest followed by apoptosis. BCL2 and MYCN were identified as miR-34a targets and likely mediators of the tumor suppressor phenotypic effect. These data support miR-34a as a tumor suppressor gene in human neuroblastoma. (Mol Cancer Res 2008;6(5):735–42)


The New England Journal of Medicine | 2008

Chromosome 6p22 Locus Associated with Clinically Aggressive Neuroblastoma

John M. Maris; Yael P. Mosse; Jonathan P. Bradfield; Cuiping Hou; Stefano Monni; Richard H. Scott; Shahab Asgharzadeh; Edward F. Attiyeh; Sharon J. Diskin; Marci Laudenslager; Cynthia Winter; Kristina A. Cole; Joseph T. Glessner; Cecilia Kim; Edward C. Frackelton; Tracy Casalunovo; Andrew W. Eckert; Mario Capasso; Eric Rappaport; Carmel McConville; Wendy B. London; Robert C. Seeger; Nazneen Rahman; Marcella Devoto; Struan F. A. Grant; Hongzhe Li; Hakon Hakonarson

BACKGROUND Neuroblastoma is a malignant condition of the developing sympathetic nervous system that most commonly affects young children and is often lethal. Its cause is not known. METHODS We performed a genomewide association study by first genotyping blood DNA samples from 1032 patients with neuroblastoma and 2043 control subjects of European descent using the Illumina HumanHap550 BeadChip. Samples from three independent groups of patients with neuroblastoma (a total of 720 patients) and 2128 control subjects were then genotyped to replicate significant associations. RESULTS We observed a significant association between neuroblastoma and the common minor alleles of three consecutive single-nucleotide polymorphisms (SNPs) at chromosome band 6p22 and containing the predicted genes FLJ22536 and FLJ44180 (P=1.71x10(-9) to 7.01x10(-10); allelic odds ratio, 1.39 to 1.40). Homozygosity for the at-risk G allele of the most significantly associated SNP, rs6939340, resulted in an increased likelihood of the development of neuroblastoma (odds ratio, 1.97; 95% confidence interval, 1.58 to 2.45). Subsequent genotyping of the three 6p22 SNPs in three independent case series confirmed our observation of an association (P=9.33x10(-15) at rs6939340 for joint analysis). Patients with neuroblastoma who were homozygous for the risk alleles at 6p22 were more likely to have metastatic (stage 4) disease (P=0.02), amplification of the MYCN oncogene in the tumor cells (P=0.006), and disease relapse (P=0.01). CONCLUSIONS A common genetic variation at chromosome band 6p22 is associated with susceptibility to neuroblastoma.


Nature Genetics | 2009

Common variations in BARD1 influence susceptibility to high-risk neuroblastoma

Mario Capasso; Marcella Devoto; Cuiping Hou; Shahab Asgharzadeh; Joseph T. Glessner; Edward F. Attiyeh; Yael P. Mosse; Cecilia Kim; Sharon J. Diskin; Kristina A. Cole; Kristopher R. Bosse; Maura Diamond; Marci Laudenslager; Cynthia Winter; Jonathan P. Bradfield; Richard H. Scott; Jayanti Jagannathan; Maria Garris; Carmel McConville; Wendy B. London; Robert C. Seeger; Struan F. A. Grant; Hongzhe Li; Nazneen Rahman; Eric Rappaport; Hakon Hakonarson; John M. Maris

We conducted a SNP-based genome-wide association study (GWAS) focused on the high-risk subset of neuroblastoma. As our previous unbiased GWAS showed strong association of common 6p22 SNP alleles with aggressive neuroblastoma, we restricted our analysis here to 397 high-risk cases compared to 2,043 controls. We detected new significant association of six SNPs at 2q35 within the BARD1 locus (Pallelic = 2.35 × 10−9–2.25 × 10−8). We confirmed each SNP association in a second series of 189 high-risk cases and 1,178 controls (Pallelic = 7.90 × 10−7–2.77 × 10−4). We also tested the two most significant SNPs (rs6435862, rs3768716) in two additional independent high-risk neuroblastoma case series, yielding combined allelic odds ratios of 1.68 each (P = 8.65 × 10−18 and 2.74 × 10−16, respectively). We also found significant association with known BARD1 nonsynonymous SNPs. These data show that common variation in BARD1 contributes to the etiology of the aggressive and most clinically relevant subset of human neuroblastoma.


Nature | 2011

Integrative genomics identifies LMO1 as a neuroblastoma oncogene

Kai Wang; Sharon J. Diskin; Haitao Zhang; Edward F. Attiyeh; Cynthia Winter; Cuiping Hou; Robert W. Schnepp; Maura Diamond; Kristopher R. Bosse; Patrick A. Mayes; Joseph T. Glessner; Cecilia Kim; Edward C. Frackelton; Maria Garris; Qun Wang; Wendy Glaberson; Rosetta M. Chiavacci; Le Nguyen; Jayanti Jagannathan; Norihisa Saeki; Hiroki Sasaki; Struan F. A. Grant; Achille Iolascon; Yael P. Mosse; Kristina A. Cole; Hongzhe Li; Marcella Devoto; Patrick McGrady; Wendy B. London; Mario Capasso

Neuroblastoma is a childhood cancer of the sympathetic nervous system that accounts for approximately 10% of all paediatric oncology deaths. To identify genetic risk factors for neuroblastoma, we performed a genome-wide association study (GWAS) on 2,251 patients and 6,097 control subjects of European ancestry from four case series. Here we report a significant association within LIM domain only 1 (LMO1) at 11p15.4 (rs110419, combined P = 5.2 × 10−16, odds ratio of risk allele = 1.34 (95% confidence interval 1.25–1.44)). The signal was enriched in the subset of patients with the most aggressive form of the disease. LMO1 encodes a cysteine-rich transcriptional regulator, and its paralogues (LMO2, LMO3 and LMO4) have each been previously implicated in cancer. In parallel, we analysed genome-wide DNA copy number alterations in 701 primary tumours. We found that the LMO1 locus was aberrant in 12.4% through a duplication event, and that this event was associated with more advanced disease (P < 0.0001) and survival (P = 0.041). The germline single nucleotide polymorphism (SNP) risk alleles and somatic copy number gains were associated with increased LMO1 expression in neuroblastoma cell lines and primary tumours, consistent with a gain-of-function role in tumorigenesis. Short hairpin RNA (shRNA)-mediated depletion of LMO1 inhibited growth of neuroblastoma cells with high LMO1 expression, whereas forced expression of LMO1 in neuroblastoma cells with low LMO1 expression enhanced proliferation. These data show that common polymorphisms at the LMO1 locus are strongly associated with susceptibility to developing neuroblastoma, but also may influence the likelihood of further somatic alterations at this locus, leading to malignant progression.


Proceedings of the National Academy of Sciences of the United States of America | 2011

RNAi screen of the protein kinome identifies checkpoint kinase 1 (CHK1) as a therapeutic target in neuroblastoma

Kristina A. Cole; Jonathan Huggins; Michael P. LaQuaglia; Chase Hulderman; Mike R. Russell; Kristopher R. Bosse; Sharon J. Diskin; Edward F. Attiyeh; Rachel Sennett; Geoffrey Norris; Marci Laudenslager; Andrew C. Wood; Patrick A. Mayes; Jayanti Jagannathan; Cynthia Winter; Yael P. Mosse; John M. Maris

Neuroblastoma is a childhood cancer that is often fatal despite intense multimodality therapy. In an effort to identify therapeutic targets for this disease, we performed a comprehensive loss-of-function screen of the protein kinome. Thirty kinases showed significant cellular cytotoxicity when depleted, with loss of the cell cycle checkpoint kinase 1 (CHK1/CHEK1) being the most potent. CHK1 mRNA expression was higher in MYC–Neuroblastoma-related (MYCN)–amplified (P < 0.0001) and high-risk (P = 0.03) tumors. Western blotting revealed that CHK1 was constitutively phosphorylated at the ataxia telangiectasia response kinase target site Ser345 and the autophosphorylation site Ser296 in neuroblastoma cell lines. This pattern was also seen in six of eight high-risk primary tumors but not in control nonneuroblastoma cell lines or in seven of eight low-risk primary tumors. Neuroblastoma cells were sensitive to the two CHK1 inhibitors SB21807 and TCS2312, with median IC50 values of 564 nM and 548 nM, respectively. In contrast, the control lines had high micromolar IC50 values, indicating a strong correlation between CHK1 phosphorylation and CHK1 inhibitor sensitivity (P = 0.0004). Furthermore, cell cycle analysis revealed that CHK1 inhibition in neuroblastoma cells caused apoptosis during S-phase, consistent with its role in replication fork progression. CHK1 inhibitor sensitivity correlated with total MYC(N) protein levels, and inducing MYCN in retinal pigmented epithelial cells resulted in CHK1 phosphorylation, which caused growth inhibition when inhibited. These data show the power of a functional RNAi screen to identify tractable therapeutical targets in neuroblastoma and support CHK1 inhibition strategies in this disease.


Science Translational Medicine | 2011

Differential Inhibitor Sensitivity of Anaplastic Lymphoma Kinase Variants Found in Neuroblastoma

Scott C. Bresler; Andrew C. Wood; Elizabeth Haglund; Joshua Courtright; Lili T. Belcastro; Jefferson S. Plegaria; Kristina A. Cole; Yana Toporovskaya; Huaqing Zhao; Erica L. Carpenter; James G. Christensen; John M. Maris; Mark A. Lemmon; Yael P. Mosse

Neuroblastoma sensitivity to crizotinib depends on the ATP-binding affinity of ALK variants, suggesting that higher doses may overcome resistance. A Boost for Neuroblastoma Therapy Neuroblastoma, a malignancy of the autonomic nervous system, is the most common cancer in children under 1 year of age. Nearly 10% of spontaneous neuroblastoma patients house mutations in the gene that encodes anaplastic lymphoma kinase (ALK). The U.S. Food and Drug Administration recently approved crizotinib—a small-molecule inhibitor of ALK’s tyrosine kinase activity and thus its cell signaling function—for the treatment of non–small cell lung carcinomas, and the drug is in early clinical trials for neuroblastoma. However, tumors with certain ALK mutations do not appear to respond to crizotinib. Bresler et al. now dissect the molecular mechanisms behind the differential crizotinib sensitivities of individual ALK mutations. Crizotinib inhibits kinase activity by competing for binding with the enzyme’s adenosine triphosphate (ATP) substrate. The authors used human neuroblastoma cell lines and xenografts in mice to show that cancers with the two most common ALK mutations, F1174L and R1275Q, are unresponsive to and effectively inhibited by crizotinib therapy, respectively. This reduced sensitivity was caused by a heightened ATP-binding affinity in F1174L-mutated ALK. These observations suggest that either increasing the dose of crizotinib or engineering higher-affinity inhibitors should improve therapy for patients with this common ALK mutation. Although careful toxicity studies need to be performed to find the maximum tolerated dose in the pediatric population, this mechanistic study provides more than a baby step toward improving crizotinib therapy in the clinic. Activating mutations in the anaplastic lymphoma kinase (ALK) gene were recently discovered in neuroblastoma, a cancer of the developing autonomic nervous system that is the most commonly diagnosed malignancy in the first year of life. The most frequent ALK mutations in neuroblastoma cause amino acid substitutions (F1174L and R1275Q) in the intracellular tyrosine kinase domain of the intact ALK receptor. Identification of ALK as an oncogenic driver in neuroblastoma suggests that crizotinib (PF-02341066), a dual-specific inhibitor of the ALK and Met tyrosine kinases, will be useful in treating this malignancy. Here, we assessed the ability of crizotinib to inhibit proliferation of neuroblastoma cell lines and xenografts expressing mutated or wild-type ALK. Crizotinib inhibited proliferation of cell lines expressing either R1275Q-mutated ALK or amplified wild-type ALK. In contrast, cell lines harboring F1174L-mutated ALK were relatively resistant to crizotinib. Biochemical analyses revealed that this reduced susceptibility of F1174L-mutated ALK to crizotinib inhibition resulted from an increased adenosine triphosphate–binding affinity (as also seen in acquired resistance to epidermal growth factor receptor inhibitors). Thus, this effect should be surmountable with higher doses of crizotinib and/or with higher-affinity inhibitors.

Collaboration


Dive into the Yael P. Mosse's collaboration.

Top Co-Authors

Avatar

John M. Maris

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Edward F. Attiyeh

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kristina A. Cole

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Cynthia Winter

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hakon Hakonarson

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Andrew C. Wood

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Cuiping Hou

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Fox

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge