460 results on '"James R. Downing"'
Search Results
202. Targetable Kinase-Activating Lesions in Ph-like Acute Lymphoblastic Leukemia
- Author
-
Donald Yergeau, I-Ming Chen, Stephen P. Hunger, Natalia Santiago-Morales, Kelly McCastlain, F. Keller, Changxue Lu, Jinghui Zhang, Richard C. Harvey, Kristy Boggs, James R. Downing, Pankaj Gupta, Ilaria Iacobucci, Guangchun Song, Jing Ma, J. A. Whitlock, Richard K. Wilson, Ji Wen, Heather L. Mulder, Gang Wu, William E. Evans, Erin Hedlund, John Easton, Mary V. Relling, Nyla A. Heerema, Charles G. Mullighan, Elisabeth Paietta, J. M. Guidry Auvil, William L. Carroll, Daniela S. Gerhard, Mignon L. Loh, Andrew J. Carroll, Amy Smith, Wendy Stock, Ching-Hon Pui, Kathryn G. Roberts, Naomi J. Winick, Andrew S. Moore, Jessica Kohlschmidt, Julie M. Gastier-Foster, Timothy P. Hughes, Xiang Chen, Bhavin Vadodaria, S. C. Chen, Eric Larsen, Steven W. Paugh, Clara D. Bloomfield, Elizabeth A. Raetz, Marina Konopleva, Dehua Pei, J. Cheng, G. Grayson, Michael Rusch, Li Ding, Robert S. Fulton, Krzysztof Mrózek, Melissa Frei-Jones, Malcolm A. Smith, Yongjin Li, Sarah K. Tasian, Steven M. Kornblau, Debbie Payne-Turner, Jared Becksfort, Meenakshi Devidas, Elaine R. Mardis, Cheryl L. Willman, Guido Marcucci, S. Reshmi, Panduka Nagahawatte, Cheng Cheng, Yung-Li Yang, Deborah L. White, Sima Jeha, and Marcus B. Valentine
- Subjects
Adult ,Male ,Adolescent ,PDGFRB ,Philadelphia chromosome ,Polymorphism, Single Nucleotide ,Article ,Mice ,Young Adult ,hemic and lymphatic diseases ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Medicine ,Animals ,Humans ,Philadelphia Chromosome ,Receptors, Cytokine ,Child ,Protein Kinase Inhibitors ,Oligonucleotide Array Sequence Analysis ,ABL ,PTK2B ,Crizotinib ,Genome, Human ,business.industry ,Infant ,DNA, Neoplasm ,General Medicine ,Protein-Tyrosine Kinases ,medicine.disease ,Survival Analysis ,Dasatinib ,Tyrosine kinase 2 ,Child, Preschool ,Cancer research ,Heterografts ,Female ,business ,Tyrosine kinase ,Signal Transduction ,medicine.drug - Abstract
BACKGROUND Philadelphia chromosome–like acute lymphoblastic leukemia (Ph-like ALL) is characterized by a gene-expression profile similar to that of BCR–ABL1–positive ALL, alterations of lymphoid transcription factor genes, and a poor outcome. The frequency and spectrum of genetic alterations in Ph-like ALL and its responsiveness to tyrosine kinase inhibition are undefined, especially in adolescents and adults. METHODS We performed genomic profiling of 1725 patients with precursor B-cell ALL and detailed genomic analysis of 154 patients with Ph-like ALL. We examined the functional effects of fusion proteins and the efficacy of tyrosine kinase inhibitors in mouse pre-B cells and xenografts of human Ph-like ALL. RESULTS Ph-like ALL increased in frequency from 10% among children with standard-risk ALL to 27% among young adults with ALL and was associated with a poor outcome. Kinase-activating alterations were identified in 91% of patients with Ph-like ALL; rearrangements involving ABL1, ABL2, CRLF2, CSF1R, EPOR, JAK2, NTRK3, PDGFRB, PTK2B, TSLP, or TYK2 and sequence mutations involving FLT3, IL7R, or SH2B3 were most common. Expression of ABL1, ABL2, CSF1R, JAK2, and PDGFRB fusions resulted in cytokine-independent proliferation and activation of phosphorylated STAT5. Cell lines and human leukemic cells expressing ABL1, ABL2, CSF1R, and PDGFRB fusions were sensitive in vitro to dasatinib, EPOR and JAK2 rearrangements were sensitive to ruxolitinib, and the ETV6–NTRK3 fusion was sensitive to crizotinib. CONCLUSIONS Ph-like ALL was found to be characterized by a range of genomic alterations that activate a limited number of signaling pathways, all of which may be amenable to inhibition with approved tyrosine kinase inhibitors. Trials identifying Ph-like ALL are needed to assess whether adding tyrosine kinase inhibitors to current therapy will improve the survival of patients with this type of leukemia. (Funded by the American Lebanese Syrian Associated Charities and others.)
- Published
- 2014
203. CONSERTING: integrating copy-number analysis with structural-variation detection
- Author
-
David W. Ellison, Xiang Chen, Samir Patel, Gang Wu, Aman Patel, James Dalton, Sheila A. Shurtleff, Kathryn G. Roberts, Joy Nakitandwe, Matthew Parker, Jinghui Zhang, Suzanne J. Baker, Pankaj Gupta, Jing Ma, James R. Downing, Debbie Payne, Charles G. Mullighan, Jianmin Wang, Linda Holmfeldt, Stanley Pounds, Stephen Espy, Michael A. Dyer, John Easton, and Michael Rusch
- Subjects
Adult ,Genetic Markers ,DNA Copy Number Variations ,Copy number analysis ,Genomics ,Computational biology ,Biology ,Biochemistry ,Genome ,Article ,Structural variation ,Data sequences ,Dna genetics ,Neoplasms ,Humans ,Child ,Molecular Biology ,Genetics ,Breakpoint ,Computational Biology ,Cell Biology ,DNA ,Gene Expression Regulation, Neoplastic ,Genetic marker ,Algorithms ,Software ,Biotechnology - Abstract
We developed Copy Number Segmentation by Regression Tree in Next Generation Sequencing (CONSERTING), an algorithm for detecting somatic copy-number alteration (CNA) using whole-genome sequencing (WGS) data. CONSERTING performs iterative analysis of segmentation on the basis of changes in read depth and the detection of localized structural variations, with high accuracy and sensitivity. Analysis of 43 cancer genomes from both pediatric and adult patients revealed novel oncogenic CNAs, complex rearrangements and subclonal CNAs missed by alternative approaches.
- Published
- 2014
204. C11ORF95-RELA FUSIONS DRIVE ONCOGENIC NF-KB SIGNALING IN EPENDYMOMA
- Author
-
David W. Ellison, Donald Yergeau, Xiang Chen, Richard J. Gilbertson, Kirti Gupta, Douglas R. Green, Kerri Ochoa, James Dalton, David Finkelstein, Heather L. Mulder, David Zhao, Yuxin Li, Jing Ma, Thomas E. Merchant, Ryan P. Lee, Bo Tang, Timothy N. Phoenix, Yongjin Li, Amar Gajjar, Pankaj Gupta, Erin Hedlund, John Easton, Radhika Thiruvenkatam, Gang Wu, Kumarasamypet M. Mohankumar, Lucinda L. Fulton, Chandanamali Punchihewa, Panduka Nagahawhatta, Frederick A. Boop, Sheila A. Shurtleff, Elaine R. Mardis, Kristy Boggs, Ricardo Weinlich, Elsie White, Charles Lu, Robert Huether, Michael Rusch, Jinghui Zhang, Ruth G. Tatevossian, Amy A Smith, Wilda Orisme, Jared Becksford, Bhavin Vadodaria, Li Ding, Guangchun Song, Richard K. Wilson, James R. Downing, Robert S. Fulton, and Matthew Parker
- Subjects
Ependymoma ,Genetics ,Cancer Research ,Mutation ,Chromothripsis ,Effector ,Cancer ,Biology ,medicine.disease ,medicine.disease_cause ,Fusion protein ,abstracts ,Transcriptome ,Oncology ,medicine ,Cancer research ,Neurology (clinical) ,Gene - Abstract
BACKGROUND: The nuclear factor-kB (NF-kB) family of transcriptional regulators are central mediators of the cellular inflammatory response. Although constitutive NF-kB signaling is present in most human tumours, mutations in pathway members are rare, complicating efforts to understand and block aberrant NF-kB activity in cancer. METHODS: To identify additional genetic alterations that drive ependymoma, we sequenced the whole genomes (WGS) of 41 tumours and matched normal blood, and the transcriptomes (RNAseq) of 77 tumours. The transforming significance of alterations were tested in mouse NSCs that we showed previously to be cells of origin of ependymoma. RESULTS: Here, we show that more than two thirds of supratentorial ependymomas contain oncogenic fusions between RELA, the principal effector of canonical NF-kB signalling, and an uncharacterized gene, C11orf95. In each case, C11orf95-RELA fusions resulted from chromothripsis involving chromosome 11q13.1. C11orf95-RELA fusion proteins translocated spontaneously to the nucleus to activate NF-kB target genes, and rapidly transformed neural stem cells—the cell of origin of ependymoma—to form these tumours in mice. CONCLUSIONS: Our data identify the first highly recurrent genetic alteration of RELA in human cancer, and the C11orf95-RELA fusion protein as a potential therapeutic target in supratentorial ependymoma. SECONDARY CATEGORY: Neuropathology & Tumor Biomarkers.
- Published
- 2014
205. The landscape of somatic mutations in epigenetic regulators across 1000 pediatric cancer genomes
- Author
-
Bhavin Vadordaria, Lei Wei, Tanja A. Gruber, Jared Becksfort, Jing Ma, Matthew Parker, Richard W. Kriwacki, Sheila A. Shurtleff, Robert Huether, Kristy Boggs, Li Dong, Gerard P. Zambetti, Li Ding, James R. Downing, Gang Wu, Amanda Larson Gedman, Guangchun Song, Michael Walsh, Gordon Lemmon, Richard J. Gilbertson, Janet F. Partridge, Donald Yergeau, Jinghui Zhang, David W. Ellison, Charles G. Mullighan, Michael Rusch, Richard K. Wilson, Heather L. Mulder, Catherine Weber, Xiang Chen, Suzanne J. Baker, Jinjun Cheng, Zhongling Cai, Erin Hedlund, John Easton, Michael N. Edmonson, Elaine R. Mardis, Jinjun Dang, and Zachary J Faber
- Subjects
Retinal Neoplasms ,General Physics and Astronomy ,SMC1A ,Biology ,Gene mutation ,medicine.disease_cause ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,General Biochemistry, Genetics and Molecular Biology ,Article ,Epigenesis, Genetic ,Neoplasms ,Genes, Regulator ,medicine ,Humans ,Epigenetics ,Child ,Gene ,ATRX ,Genetics ,Mutation ,Multidisciplinary ,Brain Neoplasms ,EZH2 ,Retinoblastoma ,Cancer ,General Chemistry ,Glioma ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Medulloblastoma - Abstract
Studies of paediatric cancers have shown a high frequency of mutation across epigenetic regulators. Here we sequence 633 genes, encoding the majority of known epigenetic regulatory proteins, in over 1,000 paediatric tumours to define the landscape of somatic mutations in epigenetic regulators in paediatric cancer. Our results demonstrate a marked variation in the frequency of gene mutations across 21 different paediatric cancer subtypes, with the highest frequency of mutations detected in high-grade gliomas, T-lineage acute lymphoblastic leukaemia and medulloblastoma, and a paucity of mutations in low-grade glioma and retinoblastoma. The most frequently mutated genes are H3F3A, PHF6, ATRX, KDM6A, SMARCA4, ASXL2, CREBBP, EZH2, MLL2, USP7, ASXL1, NSD2, SETD2, SMC1A and ZMYM3. We identify novel loss-of-function mutations in the ubiquitin-specific processing protease 7 (USP7) in paediatric leukaemia, which result in decreased deubiquitination activity. Collectively, our results help to define the landscape of mutations in epigenetic regulatory genes in paediatric cancer and yield a valuable new database for investigating the role of epigenetic dysregulations in cancer.
- Published
- 2014
206. LIM domain only-2 (LMO2) induces T-cell leukemia by two distinct pathways
- Author
-
LiQi Li, Nancy A. Jenkins, Rati Tripathi, Susan M. Cleveland, Deborah A. Swing, Utpal P. Davé, Elizabeth Mathias, Lino Tessarollo, Natalina Elliott, Yajun Yi, Stephen B. Smith, Neal G. Copeland, Mary Ann Thompson, Charles G. Mullighan, Paul E. Love, J. Andrew Hardaway, Charnise Goodings, Xi Chen, James R. Downing, and Yang Du
- Subjects
LMO2 ,Transcription, Genetic ,Mouse ,Carcinogenesis ,T-cell leukemia ,Gene Expression ,lcsh:Medicine ,Penetrance ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,E-Box Elements ,Hematologic Cancers and Related Disorders ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,Molecular Cell Biology ,Basic Cancer Research ,Gene expression ,Basic Helix-Loop-Helix Transcription Factors ,Promoter Regions, Genetic ,lcsh:Science ,Regulation of gene expression ,0303 health sciences ,Multidisciplinary ,Gene Expression Regulation, Leukemic ,Animal Models ,Hematology ,LIM Domain Proteins ,Neoplasm Proteins ,Up-Regulation ,Oncology ,030220 oncology & carcinogenesis ,Medicine ,Genetic Engineering ,Protein Binding ,Signal Transduction ,Research Article ,Biotechnology ,Leukemia, T-Cell ,Transgene ,Molecular Sequence Data ,CD2 Antigens ,Mice, Transgenic ,Biology ,03 medical and health sciences ,Model Organisms ,LYL1 ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Leukemias ,Animals ,Humans ,Enhancer ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Homeodomain Proteins ,Base Sequence ,Oncogene ,lcsh:R ,Oncogenes ,Hematopoiesis ,Cancer research ,lcsh:Q ,Transcription Factors ,Transgenics - Abstract
The LMO2 oncogene is deregulated in the majority of human T-cell leukemia cases and in most gene therapy-induced T-cell leukemias. We made transgenic mice with enforced expression of Lmo2 in T-cells by the CD2 promoter/enhancer. These transgenic mice developed highly penetrant T-ALL by two distinct patterns of gene expression: one in which there was concordant activation of Lyl1, Hhex, and Mycn or alternatively, with Notch1 target gene activation. Most strikingly, this gene expression clustering was conserved in human Early T-cell Precursor ALL (ETP-ALL), where LMO2, HHEX, LYL1, and MYCN were most highly expressed. We discovered that HHEX is a direct transcriptional target of LMO2 consistent with its concordant gene expression. Furthermore, conditional inactivation of Hhex in CD2-Lmo2 transgenic mice markedly attenuated T-ALL development, demonstrating that Hhex is a crucial mediator of Lmo2's oncogenic function. The CD2-Lmo2 transgenic mice offer mechanistic insight into concordant oncogene expression and provide a model for the highly treatment-resistant ETP-ALL subtype.
- Published
- 2014
207. The Role of the AML1 Transcription Factor in Leukemogenesis
- Author
-
Robert B. Lorsbach and James R. Downing
- Subjects
Oncogene Proteins, Fusion ,Transcription factor complex ,Biology ,medicine.disease_cause ,Translocation, Genetic ,chemistry.chemical_compound ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,medicine ,Humans ,Transcription factor ,Leukemia ,Hematology ,Gene rearrangement ,Hematopoietic Stem Cells ,medicine.disease ,Hematopoiesis ,DNA-Binding Proteins ,Haematopoiesis ,RUNX1 ,chemistry ,Core Binding Factor Alpha 2 Subunit ,Cancer research ,Stem cell ,Carcinogenesis ,Transcription Factors - Abstract
Chromosomal translocations are one of the hallmarks of human leukemias. These structural abnormalities result in the generation of genetic mutations that play a direct role in the transformation of hematopoietic stem cells. Some of the most common targets of these chromosomal rearrangements are the genes that encode the AML1/CBFbeta transcription factor complex. The AML1/CBFbeta complex plays a critical role in normal hematopoiesis, controlling the initiation of a transcriptional cascade required for the formation of definitive hematopoietic stem cells. Understanding how alterations in the normal biologic activity of this transcription factor complex lead to the initiation of leukemia will provide critical insights in the molecular pathogenesis of this disease. These insights in turn are likely to lead to the development of more rational approaches to the treatment of acute leukemia. In this review, we will summarize our current understanding of the mechanisms by which alterations in the activity of AML1/CBFbeta contribute to the development of leukemia.
- Published
- 2001
208. ETO, a Target of t(8;21) in Acute Leukemia, Makes Distinct Contacts with Multiple Histone Deacetylases and Binds mSin3A through Its Oligomerization Domain
- Author
-
James R. Downing, Hironori Harada, Shari Meyers, Noel Lenny, John Nip, Bart Lutterbach, Scott W. Hiebert, David K. Strom, and Joseph M. Amann
- Subjects
Oncogene Proteins, Fusion ,Transcription, Genetic ,Molecular Sequence Data ,Biology ,Hydroxamic Acids ,DNA-binding protein ,Histone Deacetylases ,Leukemia, Myelomonocytic, Acute ,Translocation, Genetic ,Cell Line ,Mice ,RUNX1 Translocation Partner 1 Protein ,Proto-Oncogene Proteins ,medicine ,Animals ,Nuclear Receptor Co-Repressor 1 ,Amino Acid Sequence ,Enzyme Inhibitors ,Molecular Biology ,Transcription factor ,Nuclear receptor co-repressor 1 ,Transcriptional Regulation ,Binding Sites ,Models, Genetic ,Sequence Homology, Amino Acid ,Nuclear Proteins ,Cell Biology ,DNA-binding domain ,Molecular biology ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Histone Deacetylase Inhibitors ,Repressor Proteins ,Sin3 Histone Deacetylase and Corepressor Complex ,Trichostatin A ,Histone ,Core Binding Factor Alpha 2 Subunit ,biology.protein ,Corepressor ,Transcription Factors ,medicine.drug - Abstract
t(8;21) and t(16;21) create two fusion proteins, AML-1-ETO and AML-1-MTG16, respectively, which fuse the AML-1 DNA binding domain to putative transcriptional corepressors, ETO and MTG16. Here, we show that distinct domains of ETO contact the mSin3A and N-CoR corepressors and define two binding sites within ETO for each of these corepressors. In addition, of eight histone deacetylases (HDACs) tested, only the class I HDACs HDAC-1, HDAC-2, and HDAC-3 bind ETO. However, these HDACs bind ETO through different domains. We also show that the murine homologue of MTG16, ETO-2, is also a transcriptional corepressor that works through a similar but distinct mechanism. Like ETO, ETO-2 interacts with N-CoR, but ETO-2 fails to bind mSin3A. Furthermore, ETO-2 binds HDAC-1, HDAC-2, and HDAC-3 but also interacts with HDAC-6 and HDAC-8. In addition, we show that expression of AML-1-ETO causes disruption of the cell cycle in the G(1) phase. Disruption of the cell cycle required the ability of AML-1-ETO to repress transcription because a mutant of AML-1-ETO, Delta469, which removes the majority of the corepressor binding sites, had no phenotype. Moreover, treatment of AML-1-ETO-expressing cells with trichostatin A, an HDAC inhibitor, restored cell cycle control. Thus, AML-1-ETO makes distinct contacts with multiple HDACs and an HDAC inhibitor biologically inactivates this fusion protein.
- Published
- 2001
209. Resistant T-Cell Acute Lymphoblastic Leukemias That Emerge after In Vivo Treatment with Dexamethasone Frequently Down-Regulate Glucocorticoid Receptor Protein Expression
- Author
-
Alessandro Scacchetti, Jeffrey W. Craig, Kevin Shannon, Barry S. Taylor, Olga K. Weinberg, Benjamin J. Huang, Robert P. Hasserjian, Gabriela C. Monsalve, Anica M. Wandler, Scott C. Kogan, Qing Li, Keith R. Yamamoto, Monique Dail, Joy Nakitandwe, Deepak Sampath, Jinghui Zhang, James R. Downing, Hannah Yan, Jon C. Aster, and Jasmine C. Wong
- Subjects
0301 basic medicine ,business.industry ,Immunology ,Cell Biology ,Hematology ,Drug resistance ,medicine.disease ,medicine.disease_cause ,Biochemistry ,Transcriptome ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,Glucocorticoid receptor ,In vivo ,Acute lymphocytic leukemia ,medicine ,Cancer research ,KRAS ,business ,Dexamethasone ,medicine.drug - Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer, with T lineage ALL (T-ALL) accounting for approximately 20% of cases. Many T-ALL patients present with high risk clinical features, and receive aggressive multi-agent genotoxic therapies that pose a substantial risk of long-term adverse health effects. Glucocorticoids (GCs) are an integral part of current treatment strategies for T-ALL and other lymphoid cancers. Despite decades of clinical use, the molecular mechanisms underlying GC efficacy and resistance are incompletely understood. Limited initial response to GCs correlates with poor outcome, and secondary GC resistance is frequently observed in relapsed patients. Thus, identifying drugs that either enhance initial responses or prevent resistance to GCs could significantly improve the treatment of T-ALL and other lymphoid malignancies. Our laboratory performed retroviral insertional mutagenesis (RIM) in wild-type (KrasWT) and KrasG12D "knock-in" mutant mice to generate a panel of primary transplantable T-ALLs that recapitulate the genetic heterogeneity found in human cancers. We used these reagents to assess the efficacy of treatment with the GC dexamethasone (DEX) alone and in combination with the PI3 kinase (PI3K) inhibitor GDC-0941. We observed a robust and significant overall response to 15 mg/kg/day DEX in cohorts of recipient mice transplanted with 10 independent parental KrasWT (n=5) and KrasG12D (n=5) T-ALLs, which was modestly enhanced by combined treatment with 125 mg/kg/day GDC-0941. Prolonged in vivo treatment resulted in outgrowth of 65 independent relapsed T-ALLs, many of which harbor novel retroviral integrations. We verified intrinsic drug resistance in a number of these relapsed leukemias by transplanting them into secondary recipients and treating these mice with 15 mg/kg/day DEX. Intriguingly, 23 (35%) of the T-ALLs that relapsed after an initial response to treatment exhibited markedly reduced GC receptor (GR) protein expression, suggesting a novel and common mechanism for evading GC-induced cell death. Similarly, analysis of T-ALL patient samples showed that low GR expression is rare in diagnostic specimens, but enriched in relapsed/refractory cases. We have identified two likely mechanisms of GR down-regulation in relapsed mouse T-ALLs including a nonsense mutation in the gene encoding GR and a retroviral integration in a putative distal GR promoter region that greatly reduces mRNA expression. We also performed transcriptome (RNA-seq) analysis in one sensitive parental and corresponding resistant KrasWT T-ALL after short-term in vivo DEX treatment, and observed a dramatic reduction in the number of differentially expressed genes in the resistant versus parental leukemia. We recently generated transcriptomes from this panel of 10 primary RIM-induced T-ALLs with the goal of identifying a GC response signature, and also to assess the ability of resistant cells that have lost or retained GR expression to activate this program. Finally, in contrast to previous studies in this genetically accurate in vivo preclinical model (Dail et al. Nature 2014), very few relapsed T-ALLs showed reduced Notch intracellular domain (NICD) expression relative to the corresponding parental leukemia suggesting that loss of Notch1 activation is not a major mechanism contributing to DEX resistance. Taken together, these data reveal an unexpected and common putative mechanism of GC resistance in T-ALL that can inform the development of treatment strategies for relapsed/refractory patients. We are currently focusing on analyzing transcriptome data and performing whole exome sequencing in order to uncover additional mechanisms of resistance to DEX, and then using these data to identify and investigate targeted inhibitors that might overcome GC resistance in vivo. Disclosures Dail: Genentech, Inc.: Employment. Sampath:Genentech: Employment.
- Published
- 2016
210. Activating Mutations Are Potent Pro-Leukemic Mediators in Murine MLL-MLLT3 Leukemia That Cause Distinct Transcriptional Profiles
- Author
-
Tanja A. Gruber, Anna Andersson, James R. Downing, Helena Sturesson, Jing Ma, Jenny Hansson, Julhash U. Kazi, Axel Hyrenius-Wittsten, Michael P. Walsh, Pankaj Gupta, Mattias Pilheden, Stephanie Nance, Jinghui Zhang, and Guangchun Song
- Subjects
Mutation ,Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease_cause ,medicine.disease ,Biochemistry ,Chromatin ,Transplantation ,Leukemia ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,medicine ,Cancer research - Abstract
Acquired activating mutations in kinase/PI3K/RAS signaling pathways occur in about half of pediatric acute leukemia cases with Mixed Lineage Leukemiagene rearrangements (MLL-R; MLL, also known as KMT2A). Mutations resulting in activated signaling cooperate with the MLL-R in mouse leukemia models, however, detailed insight on the effect on the transcriptional and proteomic landscape is lacking. In infant MLL-R acute lymphoblastic leukemia (ALL), a subtype with a very poor prognosis, a majority of the activating mutations are subclonal, but the biological mechanisms by which subclonal mutations affect leukemogenesis remain unclear. Here we show that NRASG12D, FLT3internal tandem duplication(ITD)and FLT3N676K,cooperates with MLL-MLLT3 in myeloid leukemogenesis using a competitive murine retroviral bone marrow transplantation model. The addition of an activating mutation remodels the gene expression patterns of the MLL-R leukemia with distinct profiles for each mutation as determined by RNA-sequencing. Gene set enrichment analysis revealed enrichment of genes involved in chromatin assembly, transcription and stemness in leukemia induced by MLL-MLLT3 and NRASG12D, FLT3ITD or FLT3N676K. Leukemia induced by only MLL-MLLT3 displayed upregulation of genes involved in signal transduction suggesting activation of such pathways by alternative mechanisms. Upon secondary transplantation, mice receiving MLL-MLLT3-only leukemic cells succumbed to disease at a similar latency to those receiving MLL-MLLT3 and an activating mutation, supporting that fully transformed MLL-MLLT3 leukemias have sustained active signaling via high expression of genes involved in signal transduction. Using the same model as above but with significantly reduced numbers of transplanted cells containing an activating mutation, we could further show that a subclonal mutation, exemplified by FLT3N676K, causes significantly reduced disease latency as compared to mice receiving MLL-MLLT3 only (34 vs 50 days). The size of the double positive (MLL-MLLT3+FLT3N676K) and single positive (MLL-MLLT3) clones within each mouse was determined by flow cytometry revealing that 8/24 mice had a double positive subclone (≤50%). The clonal evolution of the double positive cells from 22/24 mice was assessed in secondary recipients showing three distinct patterns 1) increase in size, 19/22 mice 2) maintained, 2/22 mice or 3) decreased in size, 1/22 mice. Targeted gene re-sequencing of the latter leukemia that lost its MLLT3+FLT3N676K subclone, identified a de-novo CblA308T in the SH2-like domain, in the MLL-MLLT3 onlycells that had gained clonal dominance in the secondary recipient. The decreased disease latency in mice with subclonal activating mutations raises the possibility that cells with an activating mutation support the growth of other leukemic cells by direct cell-cell contact or through secreted factors. Transcriptome and proteomic analyses identified a high expression of the macrophage inhibitory factor (MIF), a pro-inflammatory cytokine, in mice with MLL-MLLT3 and NRASG12D, FLT3ITD or FLT3N676K. Addition of rMIF increased the survival of MLL-MLLT3 murine leukemic cells in vitro. Although additional factors likely mediate pro-leukemic effects in vivo, our data suggest that MIF could be one of these factors. In summary, our data demonstrate that activating mutations cooperate with the MLL-R in murine leukemogenesis and cause widespread changes in the transcriptional landscape. In addition, our results suggest that cells containing an activating mutation in addition to an MLL-fusion positively influence the survival and likely also the growth of other leukemic cells, suggesting a pro-leukemic effect mediated by interclonal cooperation between clones carrying distinct mutational set-ups in leukemogenesis. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
211. Lack ofBCL10 mutations in multiple myeloma and plasma cell leukemia
- Author
-
James R. Downing, Stephan W. Morris, Jen-Fen Fu, Lee-Yung Shih, and Sheila A. Shurtleff
- Subjects
Plasma cell leukemia ,Cancer Research ,Plasma cell dyscrasia ,Plasma cell ,Biology ,medicine.disease ,medicine.disease_cause ,Molecular biology ,Leukemia ,Exon ,medicine.anatomical_structure ,Genetics ,medicine ,Carcinogenesis ,Gene ,Multiple myeloma - Abstract
To determine whether the BCL10 mutation plays a role in the oncogenesis of plasma cell dyscrasias, we used polymerase chain reaction-single-strand conformation polymorphism (PCR-SSCP) and direct sequencing analysis and examined the genomic BCL10 mutations in 57 patients with multiple myeloma or plasma cell leukemia and 52 normal bone marrow samples. We found three polymorphic sequence variants, either alone or in combination, at codons 5 and 8, and in intron 1 at base 58 of the BCL10 gene in 37 patients with plasma cell dyscrasia. Identical aberrant band shifts were also observed in 34 normal marrow samples. No polymorphic variants were identified in exon 2 or 3 in either patient or control samples, and no pathogenic mutations were detected. Patients with plasma cell dyscrasias in Taiwan appeared to have a higher frequency of polymorphisms at codon 5 and intron 1 at base 58, and a lower frequency at codons 8 and 213. Our results suggest that BCL10 is not involved in the oncogenesis of plasma cell dyscrasias.
- Published
- 2001
212. Acute Lymphoblastic Leukemia in a Developing Country: Preliminary Results of a Nonrandomized Clinical Trial in El Salvador
- Author
-
James R. Downing, William M. Crist, Ovidio Pena, Frederick G. Behm, Federico Antillon, Raul C. Ribeiro, Gladis deReyes, Sheila A. Shurtleff, Nelson Moreno, Miguel Bonilla, Patricia L. Harrison, and Neyessa Marina
- Subjects
Male ,medicine.medical_specialty ,Vincristine ,Time Factors ,Adolescent ,International Cooperation ,medicine.medical_treatment ,Disease-Free Survival ,Prednisone ,Acute lymphocytic leukemia ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,El Salvador ,Asparaginase ,Humans ,Medicine ,Child ,Developing Countries ,Childhood Acute Lymphoblastic Leukemia ,Survival analysis ,Neoplasm Staging ,Teniposide ,Chemotherapy ,business.industry ,Cytarabine ,Infant, Newborn ,Infant ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Survival Analysis ,United States ,Surgery ,Methotrexate ,El Niño ,Child, Preschool ,Female ,business ,medicine.drug - Abstract
Purpose: To improve outcome and study biology of childhood acute lymphoblastic leukemia (ALL) in El Salvador. Patients and Methods: Between January 1994 and December 1996, 153 children of El Salvador had newly diagnosed ALL treated in a collaborative program between Hospital Benjamin Bloom and St. Jude Children's Research Hospital (SJCRH). Therapy was based on a modified SJCRH protocol, with uniform remission induction (prednisone, vincristine, L-asparaginase) followed-up by consolidation with teniposide/ cytarabine and/or high-dose methotrexate. Continuation treatment was risk-stratified: 123 patients assigned to the high-risk group received weekly rotational drug pairs, and 16 assigned to the standard-risk group received daily 6-mercaptopurine, weekly methotrexate, and monthly pulses of vincristine plus dexamethasone. High risk was defined as: DNA index
- Published
- 2000
213. Alterations of the AML1 transcription factor in human leukemia
- Author
-
Masakazu Higuchi, Allen Eng Juh Yeoh, James R. Downing, and Noel Lenny
- Subjects
Human leukemia ,Oncogene Proteins, Fusion ,Regulator ,Chromosomal translocation ,Disease ,Biology ,Translocation, Genetic ,Pathogenesis ,RUNX1 Translocation Partner 1 Protein ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Animals ,Humans ,Point Mutation ,Gene ,Transcription factor ,Genetics ,Leukemia ,Gene Expression Regulation, Leukemic ,HDAC8 ,Cell Biology ,Hematopoietic Stem Cells ,Hematopoiesis ,DNA-Binding Proteins ,Transcription Factor AP-2 ,Leukemia, Myeloid ,Acute Disease ,Core Binding Factor Alpha 2 Subunit ,Transcription Factors ,Developmental Biology - Abstract
The identification of clonal chromosomal translocations in human leukemias provided one of the first insights into the underlying pathogenesis of this clinically heterogeneous disease. Over the last decade a large number of these chromosomal rearrangements have been molecularly cloned and the involved genes identified. A surprising finding that has emerged from this work is that many of these chromosomal alterations target the genes encoding the AML1/CBFβtranscription factor complex, a critical regulator of normal hematopoiesis. In this review, we summarize our present understanding of the mechanisms through which alterations of AML1/CBF βcontribute to leukemogenesis.
- Published
- 2000
214. Biological Characteristics of the Leukemia-Associated Transcriptional Factor AML1 Disclosed by Hematopoietic Rescue of AML1-Deficient Embryonic Stem Cells by Using a Knock-in Strategy
- Author
-
Makie Yoshida, Motohiro Nishimura, Kiyoshi Takeda, Shizuo Akira, Shigeki Yagyu, Tsukasa Okuda, James R. Downing, Tatsuo Abe, and Yasuko Fujita
- Subjects
Cellular differentiation ,Biology ,Mice ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Gene knockin ,Enhancer binding ,Animals ,Humans ,Enhancer ,Cell Growth and Development ,neoplasms ,Molecular Biology ,Transcription factor ,Leukemia ,Stem Cells ,Gene Expression Regulation, Developmental ,Gene targeting ,Cell Differentiation ,Cell Biology ,Hematopoietic Stem Cells ,Embryonic stem cell ,Molecular biology ,Hematopoiesis ,DNA-Binding Proteins ,COS Cells ,Core Binding Factor Alpha 2 Subunit ,Gene Targeting ,Mutation ,Stem cell ,Transcription Factors - Abstract
AML1 is one of the most frequently mutated genes associated with human acute leukemia and encodes the DNA-binding subunit of the heterodimering transcriptional factor complex, core-binding factor (CBF) (or polyoma enhancer binding protein 2 [PEBP2]). A null mutation in either AML1 or its dimerizing partner, CBFbeta, results in embryonic lethality secondary to a complete block in fetal liver hematopoiesis, indicating an essential role of this transcription complex in the development of definitive hematopoiesis. The hematopoietic phenotype that results from the loss of AML1 can be replicated in vitro with a two-step culture system of murine embryonic stem (ES) cells. Using this experimental system, we now demonstrate that this hematopoietic defect can be rescued by expressing the PEBP2alphaB1 (AML1b) isoform under the endogenous AML1-regulatory sequences through a knock-in (targeted insertion) approach. Moreover, we demonstrate that the rescued AML1(-/-) ES cell clones contribute to lymphohematopoiesis within the context of chimeric animals. Rescue requires the transcription activation domain of AML1 but does not require the C-terminal VWRPY motif, which is conserved in all AML1 family members and has been shown to interact with the transcriptional corepressor, Groucho/transducin-like Enhancer of split. Taken together, these data provide compelling evidence that the phenotype seen in AML1-deficient mice is due solely to the loss of transcriptionally active AML1.
- Published
- 2000
215. Molecular Classification of Cancer: Class Discovery and Class Prediction by Gene Expression Monitoring
- Author
-
Mignon L. Loh, Christine Huard, Michelle Gaasenbeek, Jill P. Mesirov, Todd R. Golub, Donna K. Slonim, Michael A. Caligiuri, Hilary A. Coller, Clara D. Bloomfield, Pablo Tamayo, James R. Downing, and Eric S. Lander
- Subjects
Biology ,Bioinformatics ,Predictive Value of Tests ,Neoplasms ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,Antineoplastic Combined Chemotherapy Protocols ,Cell Adhesion ,medicine ,Humans ,Microarray databases ,Oligonucleotide Array Sequence Analysis ,Homeodomain Proteins ,Multidisciplinary ,Gene Expression Profiling ,Cell Cycle ,Reproducibility of Results ,Myeloid leukemia ,Cancer ,Oncogenes ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Class (biology) ,Neoplasm Proteins ,Gene expression profiling ,Treatment Outcome ,Leukemia, Myeloid ,Acute Disease ,Significance analysis of microarrays ,DNA microarray - Abstract
Although cancer classification has improved over the past 30 years, there has been no general approach for identifying new cancer classes (class discovery) or for assigning tumors to known classes (class prediction). Here, a generic approach to cancer classification based on gene expression monitoring by DNA microarrays is described and applied to human acute leukemias as a test case. A class discovery procedure automatically discovered the distinction between acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) without previous knowledge of these classes. An automatically derived class predictor was able to determine the class of new leukemia cases. The results demonstrate the feasibility of cancer classification based solely on gene expression monitoring and suggest a general strategy for discovering and predicting cancer classes for other types of cancer, independent of previous biological knowledge.
- Published
- 1999
216. Oncogenic homeodomain transcription factor E2A-Pbx1 activates a novel WNT gene in pre-B acute lymphoblastoid leukemia
- Author
-
Cornelis Murre, Brett P. Monia, Saskia T. C. Neuteboom, Edward Wancewicz, James R. Downing, and John Mcwhirter
- Subjects
Oncogene Proteins, Fusion ,Molecular Sequence Data ,Chromosomal translocation ,Biology ,Transactivation ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,medicine ,Humans ,Amino Acid Sequence ,Autocrine signalling ,Transcription factor ,Homeodomain Proteins ,Regulation of gene expression ,Multidisciplinary ,Wnt signaling pathway ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Zebrafish Proteins ,Biological Sciences ,medicine.disease ,Molecular biology ,Fusion protein ,Wnt Proteins ,Leukemia ,Gene Expression Regulation ,Transcription Factors - Abstract
A large fraction of pediatric pre-B acute lymphoblastoid leukemias (ALL) consistently contain a t(1;19) chromosomal translocation. The t(1;19) translocation results in the production of a chimeric transcription factor containing the N-terminal transactivation domain of E2A fused to the C-terminal DNA-binding homeodomain of Pbx1. Here, we show that the E2A-Pbx1 fusion protein activates the expression of a novel WNT gene, WNT-16. WNT-16 normally is expressed in peripheral lymphoid organs such as spleen, appendix, and lymph nodes, but not in bone marrow. In contrast, high levels of WNT-16 transcripts are present in bone marrow and cell lines derived from pre-B ALL patients carrying the E2A-Pbx1 hybrid gene. Inhibition of E2A-Pbx1 expression leads to a significant decrease in WNT-16 mRNA levels, suggesting that WNT-16 is a downstream target of E2A-Pbx1. Three putative WNT receptors, FZ-2, FZ-3, and FZ-5, are expressed in cells of the B lineage, including pre-B ALL cells aberrantly expressing WNT-16. We propose that a WNT-16-mediated autocrine growth mechanism contributes to the development of t(1;19) pre-B ALL.
- Published
- 1999
217. THE AML1-ETO CHIMAERIC TRANSCRIPTION FACTOR IN ACUTE MYELOID LEUKAEMIA: BIOLOGY AND CLINICAL SIGNIFICANCE
- Author
-
James R. Downing
- Subjects
Chimera ,Chromosomes, Human, Pair 21 ,Chromosomal translocation ,Hematology ,Biology ,medicine.disease ,Fusion protein ,Translocation, Genetic ,Aml1 eto ,Leukemia ,Chimera (genetics) ,Haematopoiesis ,Enhancer Elements, Genetic ,Leukemia, Myeloid ,Acute Disease ,Immunology ,Cancer research ,medicine ,Humans ,Clinical significance ,Transcription factor ,Chromosomes, Human, Pair 8 ,Transcription Factors - Published
- 1999
218. HERF1, a Novel Hematopoiesis-Specific RING Finger Protein, Is Required for Terminal Differentiation of Erythroid Cells
- Author
-
Clayton W. Naeve, James R. Downing, Yuka Harada, Darin P. O’Brien, Hironori Harada, and Dennis S. Rice
- Subjects
Cellular differentiation ,Molecular Sequence Data ,Biology ,Core binding factor ,DNA-binding protein ,Protein Structure, Secondary ,Cell Line ,Tripartite Motif Proteins ,Mice ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Ring finger ,medicine ,Animals ,Dimethyl Sulfoxide ,Erythropoiesis ,RNA, Messenger ,Cloning, Molecular ,Cell Growth and Development ,Molecular Biology ,Transcription factor ,In Situ Hybridization ,Zinc finger ,Intracellular Signaling Peptides and Proteins ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Zinc Fingers ,Cell Biology ,Tetracycline ,Molecular biology ,DNA-Binding Proteins ,Haematopoiesis ,medicine.anatomical_structure ,Core Binding Factor Alpha 2 Subunit ,Carrier Proteins ,Transcription Factors - Abstract
The AML1/core binding factor beta (CBFbeta) transcription factor is essential for definitive hematopoiesis; however, the downstream pathways through which it functions remain incompletely defined. Using a differential cloning approach to define components of this pathway, we have identified a novel gene designated HERF1 (for hematopoietic RING finger 1), whose expression during development is dependent on the presence of functional AML1/CBFbeta. HERF1 contains a tripartite RING finger-B box-alpha-helical coiled-coil domain and a C-terminal region homologous to the ret proto-oncogene-encoded finger protein. Expression of HERF1 during embryogenesis coincides with the appearance of definitive erythropoiesis and in adult mice is restricted to erythroid cells, increasing 30-fold during terminal differentiation. Importantly, inhibition of HERF1 expression blocked terminal erythroid differentiation of the murine erythroleukemia cell line MEL, whereas its overexpression induced erythroid maturation. These results suggest an important role for this protein in erythropoiesis.
- Published
- 1999
219. Three distinct domains in TEL-AML1 are required for transcriptional repression of the IL-3 promoter
- Author
-
Hideo Uchida, Yasushi Miyazaki, Stephen D. Nimer, Jin Zhang, James R. Downing, and Richard C. Frank
- Subjects
Transcriptional Activation ,Gene isoform ,Cancer Research ,Oncogene Proteins, Fusion ,Mutant ,Repressor ,Biology ,Translocation, Genetic ,Transcription (biology) ,hemic and lymphatic diseases ,Gene expression ,Genetics ,Humans ,Child ,neoplasms ,Molecular Biology ,Psychological repression ,Gene ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Molecular biology ,Fusion protein ,Neoplasm Proteins ,Repressor Proteins ,Core Binding Factor Alpha 2 Subunit ,Interleukin-3 - Abstract
A cytogenetically cryptic (12;21) translocation is the most common molecular abnormality identified in childhood acute lymphoblastic leukemia (ALL), and it generates a chimeric TEL-AML1 protein. Fusion of the Helix-Loop-Helix (HLH) (also called the pointed) domain of TEL to AML1 has been suggested to convert AML1 from a transcriptional activator to a repressor. To define the structural features of this chimeric protein required for repression, we analysed the transcriptional activity of a series of TEL-AML1 mutants on the AML1-responsive interleukin-3 (IL-3) promoter, a potentially relevant gene target. Our results demonstrate that TEL-AML1 represses basal IL-3 promoter activity in lymphoid cells, and deletion mutant analysis identified three distinct domains of TEL-AML1 that are required for repression; the HLH (pointed) motif contained in the TEL portion of TEL-AML1, and both the runt homology domain (Rhd) and the 74 amino acids downstream of the Rhd that are present in the AML1 portion of the fusion protein. Although AML1B (and a shorter AML1 isoform, AML1A) have transcriptional activating activity on the IL-3 promoter, fusion of the AML1 gene to the TEL gene generates a repressor of IL-3 expression. Consistent with this activity, freshly isolated human ALL cells that contain TEL-AML1 do not express IL-3.
- Published
- 1999
220. Reappraisal of the clinical and biologic significance of myeloid-associated antigen expression in childhood acute lymphoblastic leukemia
- Author
-
Jeffrey E. Rubnitz, James R. Downing, David R. Head, Michael L. Hancock, Frederick G. Behm, Raul C. Ribeiro, Mary V. Relling, Susana C. Raimondi, William E. Evans, Gaston K. Rivera, John T. Sandlund, and Ching-Hon Pui
- Subjects
Male ,Cancer Research ,Myeloid ,Adolescent ,CD33 ,Bone Marrow Cells ,CD15 ,Disease-Free Survival ,Immunophenotyping ,Antigen ,Antigens, CD ,Antigens, Neoplasm ,Predictive Value of Tests ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,medicine ,Humans ,Prospective Studies ,Child ,Childhood Acute Lymphoblastic Leukemia ,business.industry ,Lymphoblast ,Infant ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Blotting, Southern ,ETV6 ,medicine.anatomical_structure ,Oncology ,Child, Preschool ,Immunology ,Cancer research ,Female ,business - Abstract
PURPOSE To reassess the clinical and biologic significance of myeloid-associated antigen expression in childhood acute lymphoblastic leukemia (ALL). PATIENTS AND METHODS We prospectively studied 334 newly diagnosed cases of this disease, using a comprehensive panel of antibodies that represented five myeloid cluster groups (CD13, CD14, CD15, CD33, and CD65). Blast cells were tested for ETV6 and MLL rearrangement using Southern blot analysis. RESULTS CD13 was expressed in 13.7% of cases, CD14 in 1%, CD15 in 6.6%, CD33 in 16%, and CD65 in 9.7%. Approximately one third of cases (31.4%) expressed one or more of these antigens (B-cell precursor, 31.9%; T-cell, 28.8%), while 10.5% expressed two or more (B-cell precursor, 11.3%; T-cell, 6.1%). Among the B-cell precursor leukemias, myeloid-associated antigen expression was significantly associated with a lack of hyperdiploidy and rearrangements of ETV6 or MLL gene. Most of the cases with MLL rearrangements (82%) expressed CD65, CD15, and CD33, either alone or in combination, whereas 48% of those with a rearranged ETV6 gene expressed CD13, CD33, or both. Myeloid-associated antigen expression did not correlate with event-free survival, whether the analysis was based on any of the five antigens in our panel or on the three more commonly tested antigens (CD13, CD33, and CD65). Importantly, this finding was not affected by exclusion of patients with ETV6 or MLL gene rearrangements. CONCLUSION Even though blast cell expression of myeloid-associated antigen expression shows significant associations with specific genetic abnormalities, it lacks prognostic value in childhood ALL.
- Published
- 1998
221. Characterization of new cryptic rearrangements of the erythropoietin receptor in Ph-like acute lymphoblastic leukemia
- Author
-
Julie M. Gastier-Foster, Jinghui Zhang, Richard C. Harvey, Marina Konopleva, I-Ming L. Chen, Elisabeth Paietta, Debbie Payne-Turner, Ching-Hon Pui, Kathryn G. Roberts, Kelly McCastlain, Michael Rusch, Mignon L. Loh, John Easton, Charles G. Mullighan, Cheryl L. Willman, Stephen P. Hunger, Ilaria Iacobucci, Steven M. Kornblau, Marcus B. Valentine, Jacob M. Rowe, Yongjin Li, and James R. Downing
- Subjects
Cancer Research ,Oncology ,business.industry ,Cancer research ,Medicine ,Hematology ,business ,Ph-Like Acute Lymphoblastic Leukemia ,Erythropoietin receptor - Published
- 2015
222. 12p Abnormalities and the TEL Gene (ETV6) in Childhood Acute Lymphoblastic Leukemia
- Author
-
James R. Downing, Frederick G. Behm, S. A. Shurtleff, Jeffrey E. Rubnitz, Gerard Grosveld, Susan Mathew, C H Pui, Gaston K. Rivera, Michael L. Hancock, and Susana C. Raimondi
- Subjects
Pathology ,medicine.medical_specialty ,education.field_of_study ,Immunology ,Population ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,ETV6 ,Immunophenotyping ,Acute lymphocytic leukemia ,Chromosome abnormality ,medicine ,Hyperdiploidy ,education ,Childhood Acute Lymphoblastic Leukemia ,Chromosome 12 - Abstract
Although abnormalities involving the short arm of chromosome 12 (12p) are one of the most frequently observed rearrangements in childhood acute lymphoblastic leukemia (ALL), little is known about the frequency of different structural abnormalities and their relationship to the status of the ETV6 (also named TEL) gene in this region. Of 815 children with newly diagnosed ALL, 94 (11.5%) had a total of 104 cytogenetic 12p abnormalities. Loss of genetic material was observed in 67 (64%) of these abnormalities. Cases with 12p alterations had a much lower frequency of hyperdiploidy greater than 50 (7%) than did the ALL population in general, but these cases had a similar distribution of immunophenotype and similar 5-year event-free survival (70% ± 5% SE v 64% ± 2%, P = .64). Rearrangement of the ETV6 gene was identified in 36 (56%) of 64 cases evaluated. The ETV6-CBFA2 (TEL-AML1) fusion transcript was found in 25 (66%) of 38 cases evaluated, and all but one of these showed ETV6 rearrangement. Importantly, ETV6 rearrangement was associated with a favorable prognosis (5-year event-free survival: 89% ± 6% v 60% ± 1%, P < .01). We conclude that most but not all 12p cytogenetic abnormalities in childhood ALL involve ETV6, and that rearrangement of ETV6 is associated with a favorable treatment outcome.
- Published
- 1997
223. Genetic studies of childhood acute lymphoblastic leukemia with emphasis on p16, MLL, and ETV6 gene abnormalities: results of St Jude Total Therapy Study XII
- Author
-
Susana C. Raimondi, John T. Sandlund, Patricia L. Harrison, Jeffrey E. Rubnitz, Mary V. Relling, Gerard Grosveld, William E. Evans, James R. Downing, Frederick G. Behm, Raul C. Ribeiro, and C H Pui
- Subjects
Oncology ,Heterozygote ,Cancer Research ,medicine.medical_specialty ,Chromosomes, Human, Pair 22 ,Chromosome Disorders ,Chromosomal translocation ,Biology ,Disease-Free Survival ,Translocation, Genetic ,Risk Factors ,Acute lymphocytic leukemia ,Internal medicine ,Proto-Oncogenes ,medicine ,Humans ,Child ,Childhood Acute Lymphoblastic Leukemia ,Cyclin-Dependent Kinase Inhibitor p16 ,Survival analysis ,Chromosome Aberrations ,Proto-Oncogene Proteins c-ets ,DNA, Neoplasm ,Histone-Lysine N-Methyltransferase ,Hematology ,Gene rearrangement ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Survival Analysis ,DNA-Binding Proteins ,Repressor Proteins ,Leukemia ,ETV6 ,Child, Preschool ,Cancer research ,Myeloid-Lymphoid Leukemia Protein ,Carrier Proteins ,Chromosomes, Human, Pair 9 ,Gene Deletion ,Transcription Factors - Abstract
To determine the frequency and prognostic significance of recently described genetic lesions in pediatric acute lymphoblastic leukemia (ALL), all cases with available leukemic cell samples treated on St Jude Study XII were analyzed by molecular techniques for alterations of the p16, MLL and ETV6 genes. Homozygous p16 deletion was seen in 36 of 155 cases, including 14 of 23 T cell cases, but had no prognostic value. Rearrangement of MLL was seen in nine of 170 cases (5%) and conferred a poor prognosis, with a 5-year EFS estimate of only 11 +/- 7%, compared with 74 +/- 5% for the germline MLL group (P=0.0001). By contrast, rearrangement of ETV6 was found in 35 cases (21%) and was significantly associated with a better outcome (5-year EFS estimates: 87 +/- 7% vs 64 +/- 6%). In a Cox regression model adjusted for age, DNA index, race, leukocyte count, treatment group, and CNS status, ETV6 rearrangement retained independent prognostic significance (two-sided P value 0.012). Thus, in this uniformly treated group of patients, we confirmed the unfavorable prognostic significance of MLL rearrangement and demonstrated the favorable impact of ETV6 rearrangement, suggesting that these factors be added to ALL risk classification schemes.
- Published
- 1997
224. Disseminated Intrathoracic Desmoplastic Small Round-Cell Tumor: A Case Report
- Author
-
Alberto S. Pappo, James R. Downing, Jesse J. Jenkins, Sheila A. Shurtleff, Sue C. Kaste, and Lakshmi Venkateswaran
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Genes, Wilms Tumor ,Adolescent ,Desmoplastic small-round-cell tumor ,Soft Tissue Neoplasms ,Sarcoma, Ewing ,Malignancy ,Metastasis ,Fatal Outcome ,medicine ,Humans ,Neoplasm ,business.industry ,Soft tissue ,Hematology ,medicine.disease ,Primary tumor ,Desmoplasia ,medicine.anatomical_structure ,Oncology ,Sarcoma, Small Cell ,Pediatrics, Perinatology and Child Health ,Abdomen ,medicine.symptom ,business - Abstract
Purpose : Recently recognized as a distinct clinicopathologic entity, desmoplastic small round-cell tumors typically affect young men. These aggressive tumors usually arise in the abdomen; other sites of primary disease have been described only rarely. We report the case of an extraabdominal primary tumor with widespread dissemination, including the subcutaneous tissue, a previously unrecognized metastatic site. Patient and Methods: We describe the case of 16-year-old boy with a primary extraabdominal metastatic desmoplastic small round-cell tumor. Results: Our patient had a primary intrathoracic desmoplastic small round-cell tumor and widespread dissemination involving the subcutaneous tissue, kidney, liver, bone, and lymph nodes. Histopathologic analysis found intense desmoplasia and polyphenotypic expression of neural, muscle, and epithelial markers. Reverse transcriptase-polymerase chain reaction analysis of fresh tumor tissue confirmed the characteristic EWS-WT1 transcript. Conclusions: Broader than originally anticipated, the clinical spectrum of desmoplastic small round-cell tumors continues to evolve. Primary intrathoracic tumors with soft-tissue dissemination and polyphenotypic expression should prompt suspicion of this malignancy. Molecular analysis of fresh tumor tissue is an important adjunct to diagnosing this rare neoplasm.
- Published
- 1997
225. Case-Control Study Suggests a Favorable Impact of TEL Rearrangement in Patients With B-Lineage Acute Lymphoblastic Leukemia Treated With Antimetabolite-Based Therapy: A Pediatric Oncology Group Study
- Author
-
D. Jeanette Pullen, Jeffrey E. Rubnitz, Michael P. Link, Vita J. Land, Jonathan J. Shuster, Ching-Hon Pui, Bruce M. Camitta, James R. Downing, and Frederick G. Behm
- Subjects
Oncology ,Chemotherapy ,medicine.medical_specialty ,business.industry ,medicine.drug_class ,medicine.medical_treatment ,Immunology ,Case-control study ,Retrospective cohort study ,Cell Biology ,Hematology ,Gene rearrangement ,medicine.disease ,Biochemistry ,Antimetabolite ,Germline ,El Niño ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,Internal medicine ,Medicine ,business - Abstract
TEL gene rearrangement is the most common genetic lesion in pediatric acute lymphoblastic leukemia (ALL), occurring in about 25% of B-lineage cases. We previously showed that, among patients treated on St Jude protocols, TEL rearrangement independently conferred an excellent prognosis. To extend these results to patients treated with antimetabolite-based therapy, we performed Southern blot analysis to determine the TEL gene status of 104 cases of B-lineage ALL treated on Pediatric Oncology Group 8602, matched on age, gender, and leukocyte count. There were 52 failures among the 77 patients with germline TEL, compared with only 8 failures among 27 patients in the rearranged group. Based on a two-sided logistic regression analysis, stratified for age (subdivided at 10 years), leukocyte count (subdivided at 50,000), and gender, the estimated odds of failing by 4 years in the germline TEL group is 5.4 times that of the rearranged TEL group, with 95% confidence from 1.9 to 15.6, two-sided P = .0009. Thus, the presence of a rearranged TEL gene is also associated with an improved survival among patients treated with antimetabolite-based therapy. Our results indicate that all newly diagnosed ALL patients should be screened for TEL gene rearrangements and suggest that these patients are candidates for less intensive therapy.
- Published
- 1997
226. Identification of a recurrent germline PAX5 mutation and susceptibility to pre-B cell acute lymphoblastic leukemia
- Author
-
Katherine L. Tucker, Samantha Hansford, Guangchun Song, Tom Walsh, Paul A. Meyers, Martin Fleisher, Kenan Onel, Vundavalli V. Murty, Nicholas D. Socci, Deepa Bhojwani, Sharon E. Plon, Susana C. Raimondi, Jeremy Wechsler, Ming Lee, Hamish S. Scott, Jinghui Zhang, Peter Maslak, Andrew E. Timms, Sohela Shah, Megan Harlan Fleischut, Agnes Viale, Jing Ma, Annet Simons, Rohini Rau-Murthy, Lily Offit, Xiaoni Gao, Kathryn G. Roberts, Robert J. Klein, Georgia Chenevix-Trench, Kenneth Offit, Scott W. Lowe, Esmé Waanders, Rosemary Sutton, Mary Claire King, Jun Li, David S. Ziegler, Suresh C. Jhanwar, Deborah I. Ritter, Mark J. Daly, Shann Ching Chen, Joseph Vijai, Sarah M. Lo, Marshall S. Horwitz, Jason Littman, John T. Sandlund, Cornelius Miething, Joshua D. Schiffman, James E. Hayes, Michael Rusch, David A. Wheeler, Jinjun Cheng, Gang Wu, Rajmohan Murali, David Altshuler, Kasmintan A. Schrader, Roland P. Kuiper, Katherine L. Nathanson, Steven M. Lipkin, Charles G. Mullighan, Marina Corines, Christopher N. Hahn, Panduka Nagahawatte, David G. Huntsman, Ross L. Levine, Lei Wei, Christopher Manschreck, Janine Senz, James R. Downing, Thomas Kitzing, and Jun J. Yang
- Subjects
Somatic cell ,Biology ,medicine.disease_cause ,Polymorphism, Single Nucleotide ,Germline ,Article ,Loss of heterozygosity ,Germline mutation ,immune system diseases ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,hemic and lymphatic diseases ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,Genetics and epigenetic pathways of disease Translational research [NCMLS 6] ,B cell ,Germ-Line Mutation ,Mutation ,Leukemia ,PAX5 Transcription Factor ,medicine.disease ,medicine.anatomical_structure ,FOS: Biological sciences ,Human chromosome abnormalities--Diagnosis ,Cancer research - Abstract
Contains fulltext : 126772.pdf (Publisher’s version ) (Closed access) Somatic alterations of the lymphoid transcription factor gene PAX5 (also known as BSAP) are a hallmark of B cell precursor acute lymphoblastic leukemia (B-ALL), but inherited mutations of PAX5 have not previously been described. Here we report a new heterozygous germline variant, c.547G>A (p.Gly183Ser), affecting the octapeptide domain of PAX5 that was found to segregate with disease in two unrelated kindreds with autosomal dominant B-ALL. Leukemic cells from all affected individuals in both families exhibited 9p deletion, with loss of heterozygosity and retention of the mutant PAX5 allele at 9p13. Two additional sporadic ALL cases with 9p loss harbored somatic PAX5 substitutions affecting Gly183. Functional and gene expression analysis of the PAX5 mutation demonstrated that it had significantly reduced transcriptional activity. These data extend the role of PAX5 alterations in the pathogenesis of pre-B cell ALL and implicate PAX5 in a new syndrome of susceptibility to pre-B cell neoplasia.
- Published
- 2013
227. Loss of oncogenic Notch1 with resistance to a PI3K inhibitor in T-cell leukaemia
- Author
-
Jon C. Aster, Kevin Shannon, Leslie Lee, Shann Ching Chen, Joy Nakitandwe, Jin Xu, Monique Dail, Keiko Akagi, Jessica Lawrence, Jason Wong, James R. Downing, Daniel O'Connor, Qing Li, Deepak Sampath, and Warren S. Pear
- Subjects
Male ,Drug Resistance ,Drug resistance ,Mitogen-activated protein kinase kinase ,Inbred C57BL ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Mice ,hemic and lymphatic diseases ,Receptor, Notch1 ,ras ,Cancer ,Phosphoinositide-3 Kinase Inhibitors ,Pediatric ,Sulfonamides ,Multidisciplinary ,MEK inhibitor ,Drug Synergism ,Hematology ,5.1 Pharmaceuticals ,embryonic structures ,Benzamides ,cardiovascular system ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Development of treatments and therapeutic interventions ,Receptor ,Signal Transduction ,Protein Structure ,Indazoles ,Childhood Leukemia ,Pediatric Cancer ,General Science & Technology ,Notch signaling pathway ,Down-Regulation ,Biology ,Article ,Rare Diseases ,Downregulation and upregulation ,Animals ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Mitogen-Activated Protein Kinase Kinases ,Notch1 ,Phosphoinositide 3-kinase ,Diphenylamine ,Clone Cells ,Protein Structure, Tertiary ,Mice, Inbred C57BL ,Genes, ras ,Genes ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Neoplasm ,sense organs ,Proto-Oncogene Proteins c-akt ,Tertiary - Abstract
Mutations that deregulate Notch1 and Ras/phosphoinositide 3 kinase (PI3K)/Akt signalling are prevalent in T-cell acute lymphoblastic leukaemia (T-ALL), and often coexist. Here we show that the PI3K inhibitor GDC-0941 is active against primary T-ALLs from wild-type and KrasG12D mice, and addition of the MEK inhibitor PD0325901 increases its efficacy. Mice invariably relapsed after treatment with drug-resistant clones, most of which unexpectedly had reduced levels of activated Notch1 protein, downregulated many Notch1 target genes, and exhibited cross-resistance to γ-secretase inhibitors. Multiple resistant primary T-ALLs that emerged in vivo did not contain somatic Notch1 mutations present in the parental leukaemia. Importantly, resistant clones upregulated PI3K signalling. Consistent with these data, inhibiting Notch1 activated the PI3K pathway, providing a likely mechanism for selection against oncogenic Notch1 signalling. These studies validate PI3K as a therapeutic target in T-ALL and raise the unexpected possibility that dual inhibition of PI3K and Notch1 signalling could promote drug resistance in T-ALL. Mutations that dysregulate Notch1 and Ras/PI3K signalling are common in T-cell acute lymphoblastic leukaemia; here, treatment with a PI3K inhibitor is shown to induce drug resistance that is associated with downregulation of activated Notch1 signalling, suggesting that inhibition of both Notch1 and PI3K could promote drug resistance. T-cell acute lymphoblastic leukaemia (T-ALL) is closely associated with mutations that deregulate Notch1 and Ras/phosphoinositide 3 kinase (PI3K) signalling. Kevin Shannon and colleagues show that treatment with GDC-0941 (pictilisib), a PI3K inhibitor that is advancing in clinical development, induces resistance associated with downregulation of activated Notch signalling, cross-resistance to γ-secretase inhibitors and upregulated PI3K signalling. Inhibition of Notch1 activates the PI3K pathway, suggesting that the rational strategy of concurrently targeting activated Notch1 and PI3K in T-ALL is likely to accelerate drug resistance.
- Published
- 2013
228. Integrated genetic and epigenetic analysis of childhood acute lymphoblastic leukemia
- Author
-
James R. Downing, Yushan Li, Shann Ching Chen, Mondira Kundu, Charles G. Mullighan, Ari Melnick, Maria E. Figueroa, Jason Sotzen, Letha A. Phillips, and Anna Andersson
- Subjects
Genetics ,DNA Copy Number Variations ,Microarray analysis techniques ,Gene Expression Regulation, Leukemic ,General Medicine ,Biology ,DNA Methylation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Epigenesis, Genetic ,Epigenetics of physical exercise ,Cell Transformation, Neoplastic ,DNA methylation ,Gene expression ,Cluster Analysis ,Humans ,Epigenetics ,Child ,Transcriptome ,Gene ,RNA-Directed DNA Methylation ,Epigenomics ,Research Article ,Genes, Neoplasm ,Oligonucleotide Array Sequence Analysis ,Signal Transduction - Abstract
Acute lymphoblastic leukemia (ALL) is the commonest childhood malignancy and is characterized by recurring structural genetic alterations. Previous studies of DNA methylation suggest epigenetic alterations may also be important, but an integrated genome-wide analysis of genetic and epigenetic alterations in ALL has not been performed. We analyzed 137 B-lineage and 30 T-lineage childhood ALL cases using microarray analysis of DNA copy number alterations and gene expression, and genome-wide cytosine methylation profiling using the HpaII tiny fragment enrichment by ligation-mediated PCR (HELP) assay. We found that the different genetic subtypes of ALL are characterized by distinct DNA methylation signatures that exhibit significant correlation with gene expression profiles. We also identified an epigenetic signature common to all cases, with correlation to gene expression in 65% of these genes, suggesting that a core set of epigenetically deregulated genes is central to the initiation or maintenance of lymphoid transformation. Finally, we identified aberrant methylation in multiple genes also targeted by recurring DNA copy number alterations in ALL, suggesting that these genes are inactivated far more frequently than suggested by structural genomic analyses alone. Together, these results demonstrate subtype- and disease-specific alterations in cytosine methylation in ALL that influence transcriptional activity, and are likely to exert a key role in leukemogenesis.
- Published
- 2013
229. Global chromatin profiling reveals NSD2 mutations in pediatric acute lymphoblastic leukemia
- Author
-
Robert Schlegel, Levi A. Garraway, Jing Ma, James R. Downing, Min Hu, Yan Wang, Ho Man Chan, Feng Yan, E. Robert McDonald, Giordano Caponigro, Veronica Gibaja, Nicholas Keen, Robert Huether, Charles G. Mullighan, John Easton, Vesselina G. Cooke, Jacob D. Jaffe, Steven A. Carr, Rosalie deBeaumount, Nathan P. Englund, Zhengtian Yu, Hyo-eun C. Bhang, Frank Stegmeier, Lei Wei, William R. Sellers, Jun Liu, Jordan E. Taylor, Zhaofu Wang, Jordi Barretina, Kavitha Venkatesan, Gregory V. Kryukov, and Jinghui Zhang
- Subjects
Methyltransferase ,Mice, SCID ,Histones ,chemistry.chemical_compound ,Mice ,Cell Line, Tumor ,Genetics ,Animals ,Humans ,Genetic Predisposition to Disease ,Epigenetics ,Child ,Gene knockdown ,biology ,Base Sequence ,Genetic Variation ,Histone-Lysine N-Methyltransferase ,Sequence Analysis, DNA ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Molecular biology ,Pediatric cancer ,Xenograft Model Antitumor Assays ,Chromatin ,Repressor Proteins ,Histone ,chemistry ,biology.protein ,NIH 3T3 Cells ,Ectopic expression ,Female ,DNA ,Neoplasm Transplantation - Abstract
Epigenetic dysregulation is an emerging hallmark of cancers. We developed a high-information-content mass spectrometry approach to profile global histone modifications in human cancers. When applied to 115 lines from the Cancer Cell Line Encyclopedia1, this approach identified distinct molecular chromatin signatures. One signature was characterized by increased histone 3 lysine 36 (H3K36) dimethylation, exhibited by several lines harboring translocations in NSD2, which encodes a methyltransferase. A previously unknown NSD2 p.Glu1099Lys (p.E1099K) variant was identified in nontranslocated acute lymphoblastic leukemia (ALL) cell lines sharing this signature. Ectopic expression of the variant induced a chromatin signature characteristic of NSD2 hyperactivation and promoted transformation. NSD2 knockdown selectively inhibited the proliferation of NSD2-mutant lines and impaired the in vivo growth of an NSD2-mutant ALL xenograft. Sequencing analysis of >1,000 pediatric cancer genomes identified the NSD2 p.E1099K alteration in 14% of t(12;21) ETV6-RUNX1–containing ALLs. These findings identify NSD2 as a potential therapeutic target for pediatric ALL and provide a general framework for the functional annotation of cancer epigenomes.
- Published
- 2013
230. Molecular analysis of t(11;19) breakpoints in childhood acute leukemias
- Author
-
S. A. Shurtleff, A. M. Curcio-Brint, Jeffrey E. Rubnitz, Andrew J. Carroll, James R. Downing, Frederick G. Behm, Susana C. Raimondi, and RP Pinheiro
- Subjects
medicine.medical_specialty ,Acute leukemia ,Immunology ,Breakpoint ,Cytogenetics ,Myeloid leukemia ,Chromosomal translocation ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Virology ,Leukemia ,hemic and lymphatic diseases ,Chromosome 19 ,medicine ,Cancer research ,Myeloid-Lymphoid Leukemia Protein ,neoplasms - Abstract
MLL is fused to ENL or ELL in acute leukemias that contain t(ll;19)(q23;p13). Although ENL and ELL localize to chromosome 19, bands p13.3 and p13.1, respectively, these breakpoints are not always readily distinguished by standard cytogenetics. We therefore used reverse transcriptase-polymerase chain reaction (RT-PCR) assays to analyze 26 cases of childhood acute leukemia containing t(11;19) to determine the frequencies of ENL and ELL involvement. All 17 cases of acute lymphoblastic leukemia (ALL) had MLL/ENL fusion transcripts. By contrast, of the 9 cases of acute myeloid leukemia (AML) analyzed, 6 had MLL/ENL fusions, 2 had MLL/ELL fusions, and 1 case had no RT-PCR- detectable MLL fusion mRNA. These data suggest that the majority of 11;19 translocations involve ENL, whereas involvement of ELL is relatively uncommon in childhood acute leukemia and may be restricted to AML.
- Published
- 1996
231. Rearrangement of the MLL gene confers a poor prognosis in childhood acute lymphoblastic leukemia, regardless of presenting age
- Author
-
Gaston K. Rivera, C H Pui, Qing Liu, James R. Downing, Frederick G. Behm, J. H. Kersey, William M. Crist, J. L. Frestedt, and Susana C. Raimondi
- Subjects
Poor prognosis ,Immunology ,Chromosome ,Chromosomal translocation ,Cell Biology ,Hematology ,Gene rearrangement ,Biology ,medicine.disease ,Biochemistry ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,Cancer research ,medicine ,Myeloid-Lymphoid Leukemia Protein ,Clinical significance ,neoplasms ,Childhood Acute Lymphoblastic Leukemia - Abstract
MLL gene rearrangements are associated with an extremely poor prognosis in infants with acute lymphoblastic leukemia (ALL), but little is known about their clinical significance in older children. Therefore, we studied 45 cases of childhood ALL with abnormalities of chromosome 11q23 for rearrangement of the MLL gene to determine if this feature confers a uniformly poor prognosis. MLL gene rearrangements were detected in all 18 cases with the common t(4;11), t(9;11) or t(11;19) translocations, whereas only 5 of 12 patients with either unbalanced or uncommon balanced translocations demonstrated a rearrangement. Abnormalities of the MLL gene were not detected in any of the 15 cases with a deletion or inversion of the chromosomes 11q23 region. The presence of an MLL rearrangement was significantly associated with age less than 1 year (P < .001), leukocyte count –>50 x 10(9)/L (P = .003), and the absence of leukemic cell CD10 expression (P < .001). In a stratified statistical analysis adjusted for age and treatment protocol, MLL gene rearrangement was correlated with an inferior treatment outcome (P = .028). The 4-year event-free survival estimate (+/-SE) was 10% +/-6.5% for cases with a rearranged MLL gene and 64% +/-19.2% for other cases. When infants were excluded from the analysis, MLL rearrangement was still significantly associated with a poor outcome (P = .02), and remained so with the exclusion of t(4;11)-positive cases (P = .05). Thus, regardless of presenting age, MLL gene rearrangement identifies a high-risk subgroup of patients who are not likely to be cured with conventional treatment.
- Published
- 1996
232. The t(12;21) Translocation Converts AML-1B from an Activator to a Repressor of Transcription
- Author
-
D G Gilliland, J N Davis, M F Roussell, S. A. Shurtleff, Gerard Grosveld, Weili Sun, A Buijs, James R. Downing, Todd R. Golub, Shari Meyers, Noel Lenny, and Scott W. Hiebert
- Subjects
Transcription, Genetic ,Chromosomes, Human, Pair 21 ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Repressor ,Biology ,DNA-binding protein ,Translocation, Genetic ,Transactivation ,hemic and lymphatic diseases ,Humans ,Enhancer ,neoplasms ,Molecular Biology ,Transcription factor ,Sequence Deletion ,Chromosomes, Human, Pair 12 ,Leukemia ,Base Sequence ,Proto-Oncogene Proteins c-ets ,Basic helix-loop-helix ,Activator (genetics) ,Helix-Loop-Helix Motifs ,Cell Biology ,Molecular biology ,Fusion protein ,DNA-Binding Proteins ,Repressor Proteins ,Enhancer Elements, Genetic ,Transcription Factors ,Research Article - Abstract
The t(12;21) translocation is present in up to 30% of childhood B-cell acute lymphoblastic and fuses a potential dimerization motif from the ets-related factor TEL to the N terminus of AML1. The t(12;21) translocation encodes a 93-kDa fusion protein that localizes to a high-salt- and detergent-resistant nuclear compartment. This protein binds the enhancer core motif, TGTGGT, and interacts with the AML-1-binding protein, core-binding factor beta. Although TEL/AML-1B retains the C-terminal domain of AML-1B that is required for transactivation of the T-cell receptor beta enhancer, it fails to activate transcription but rather inhibits the basal activity of this enhancer. TEL/AML-1B efficiently interferes with AML-1B dependent transactivation of the T-cell receptor beta enhancer, and coexpression of wild-type TEL does not reverse this inhibition. The N-terminal TEL helix-loop-helix domain is essential for TEL/AML-1B-mediated repression. Thus, the t(12;21) fusion protein dominantly interferes with AML-1B-dependent transcription, suggesting that the inhibition of expression of AML-1 genes is critical for B-cell leukemogenesis.
- Published
- 1996
233. TGF-β Signaling, Tumor Suppression, and Acute Lymphoblastic Leukemia
- Author
-
James R. Downing
- Subjects
T-Lymphocytes ,Genes, myc ,Gene Expression ,Smad Proteins ,Protein Serine-Threonine Kinases ,Transforming Growth Factor beta1 ,Transforming Growth Factor beta ,Acute lymphocytic leukemia ,Gene expression ,medicine ,Genes, Tumor Suppressor ,Smad3 Protein ,Permissive ,Receptor ,Acute leukemia ,business.industry ,Receptor, Transforming Growth Factor-beta Type II ,General Medicine ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,DNA-Binding Proteins ,Immunology ,Cancer cell ,Trans-Activators ,Cancer research ,Signal transduction ,business ,Receptors, Transforming Growth Factor beta ,Signal Transduction ,Transcription Factors ,Transforming growth factor - Abstract
Dr. James R. Downing describes the transforming growth factor β signaling pathway that supresses the early development of cancer cells. There is mounting evidence of a permissive role of TGF-β in t...
- Published
- 2004
234. Molecular Cloning, Expression Pattern, and Chromosomal Localization of Human CDKN2D/INK4d,an Inhibitor of Cyclin D-Dependent Kinases
- Author
-
James R. Downing, Jill M. Lahti, Tsukasa Okuda, Vincent J. Kidd, Virginia Valentine, Hiroshi Hirai, Sheila A. Shurtleff, and Charles J. Sherr
- Subjects
Male ,Cyclin D ,Molecular Sequence Data ,Gene Expression ,Cell Cycle Proteins ,Polymerase Chain Reaction ,Translocation, Genetic ,Mice ,Fetus ,Consensus Sequence ,Genetics ,Animals ,Humans ,Gene family ,Amino Acid Sequence ,Cloning, Molecular ,Cyclin-Dependent Kinase Inhibitor p19 ,Enzyme Inhibitors ,CDKN2D ,Child ,Gene ,Cyclin-Dependent Kinase Inhibitor p16 ,In Situ Hybridization, Fluorescence ,DNA Primers ,Cyclin ,Base Sequence ,Sequence Homology, Amino Acid ,biology ,Kinase ,Cell Cycle ,Homozygote ,Chromosome Mapping ,Genetic Variation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Cell cycle ,Molecular biology ,Cyclin-Dependent Kinases ,Chromosomes, Human, Pair 1 ,Organ Specificity ,biology.protein ,Cyclin-dependent kinase 6 ,Carrier Proteins ,Chromosomes, Human, Pair 19 ,Gene Deletion ,HeLa Cells - Abstract
Progression through the G1 phase of the cell cycle is dependent on the activity of holoenzymes formed between D-type cyclins and their catalytic partners, the cyclin-dependent kinases cdk4 and cdk6. p16{sup INK4a} p15{sup INK4b}, and p18{sup INK4c}, a group of structurally related proteins, function as specific inhibitors of the cyclin D-dependent kinases and are likely to play physiologic roles as specific regulators of these kinases in vivo. A new member of the INK4 gene family, murine INK4d, has recently been identified. Here we report the isolation of human INK4d (gene symbol CDKN2D), which is 86 identical at the amino acid level to the murine clone and {approximately}44% identical to each of the other human INK4 family members. The INK4d gene is ubiquitously expressed as a single 1.4-kb mRNA with the highest levels detected in thymus, spleen, peripheral blood leukocytes, fetal liver, brain, and testes. The abundance of INK4d mRNA oscillates in a cell-cycle-dependent manner with expression lowest at mid G1 and maximal during S phase. Using a P1-phage genomic clone of INK4d for fluorescence in situ hybridization analysis, the location of this gene was mapped to chromosome 19p13. No rearrangements or deletions of the INK4d gene were observed in Southernmore » blot analysis of selected cases of pediatric acute lymphoblastic leukemia (ALL) containing a variant (1;19)(q23;p13) translocation that lacks rearrangement of either E2A or PBX1, or in ALL cases containing homozygous or hemizygous deletions of the related genes, INK4a and INK4b. 39 refs., 3 figs.« less
- Published
- 1995
235. Acute lymphoblastic leukemias with deletion of 11q23 or a novel inversion (11)(p13q23) lack MLL gene rearrangements and have favorable clinical features
- Author
-
C H Pui, J. L. Frestedt, David R. Head, J. H. Kersey, James R. Downing, Frederick G. Behm, and Susana C. Raimondi
- Subjects
Genetics ,Immunology ,Karyotype ,Chromosomal translocation ,Cell Biology ,Hematology ,Gene rearrangement ,Biology ,medicine.disease ,Biochemistry ,Antigen ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,Cancer research ,medicine ,Myeloid-Lymphoid Leukemia Protein ,Childhood Acute Lymphoblastic Leukemia ,Chromosomal inversion - Abstract
Balanced translocations affecting the 11q23 region are among the most frequent chromosomal abnormalities in childhood acute lymphoblastic leukemia (ALL), comprising 5% to 6%. These cases consistently have a rearranged MLL gene and are associated with high-risk presenting features, hyperleukocytosis and younger age, and a poor treatment outcome. To assess the clinical and biologic significance of 11q23- associated structural chromosomal abnormalities other than translocations, we studied 17 cases of childhood ALL [14 with del(11)(q23) and 3 with inv(11)(p12q23)] that were identified among 785 cases with successful chromosome analysis. In contrast to reported cases with 11q23 and MLL gene rearrangement, our series was characterized by relatively low leukocyte counts (median, 15.1 x 10(9)/L), expression of CD10 antigen but not myeloid-associated CD15 and CDw65 antigens, a relatively high frequency of T-cell immunophenotypes, and a generally favorable prognosis. All 13 cases with interpretable molecular analysis lacked MLL gene rearrangements. We suggest that most cases with deletions or inversions affecting the 11q23 region represent clinically and biologically different entities as compared with those defined by 11q23 translocation.
- Published
- 1995
236. Molecular detection of the (2;5) translocation of non-Hodgkin's lymphoma by reverse transcriptase-polymerase chain reaction
- Author
-
A. M. Curcio-Brint, Adonis Lorenzana, A.E. Kossakowska, Sheila A. Shurtleff, David R. Head, John T. Sandlund, Dennis D. Weisenburger, Maria Zielenska, Marc Ladanyi, Paul S. Thorner, James R. Downing, Stephan W. Morris, and Frederick G. Behm
- Subjects
medicine.medical_specialty ,Nucleophosmin ,integumentary system ,CD30 ,Immunology ,Cytogenetics ,Chromosomal translocation ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Reverse transcriptase ,Non-Hodgkin's lymphoma ,Lymphoma ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Anaplastic lymphoma kinase - Abstract
The t(2;5)(p23;q35) translocation was initially identified in cases of anaplastic large-cell lymphoma (ALCL) that expressed the Ki-1 (CD30) antigen. We have recently cloned this translocation and shown it to encode a chimeric product consisting of the N-terminal portion of a nonribosomal nucleolar phosphoprotein, nucleophosmin (NPM), from chromosome 5, fused to the kinase domain of a novel transmembrane tyrosine-specific protein kinase, anaplastic lymphoma kinase (ALK), from chromosome 2. To better define the spectrum of lymphomas that contain this translocation, we have analyzed 70 cases of non-Hodgkin's lymphoma (NHL) for expression of the t(2;5)-derived NPM/ALK chimeric message by reverse transcriptase-polymerase chain reaction (RT-PCR). Using a previously described set of oligonucleotide primers, NPM/ALK chimeric transcripts were detected in 21 of 22 cases that contained the t(2;5) by cytogenetic analysis and in 10 of 48 cases that either lacked evidence of the t(2;5) or had unsuccessful cytogenetics. In all but 1 case, the NPM/ALK PCR products were of identical size and sequence, suggesting that the genomic chromosome breaks are clustered in a single intron in both NPM and ALK. The NPM/ALK-expressing cases were not confined to NHLs with anaplastic morphology and included 15 ALCLs, 6 immunoblastic lymphomas, and 10 diffuse large-cell lymphomas. Moreover, only slightly greater than half of the cases with anaplastic morphology and 59% of CD30-expressing cases were NPM/ALK positive. Thus, neither anaplastic morphology nor the expression of CD30 accurately predicted the presence of this molecular genetic subtype of lymphoma.
- Published
- 1995
237. Childhood acute lymphoblastic leukemia with equivocal chromosome markers of the t(I;I9) translocation
- Author
-
Leonid V. Filatov, James R. Downing, Frederick G. Behm, Ching-Hon Pui, Susana C. Raimondi, and David R. Head
- Subjects
Cancer Research ,medicine.diagnostic_test ,fungi ,Chromosome ,Chromosomal translocation ,Biology ,Fusion gene ,Fusion transcript ,hemic and lymphatic diseases ,Chromosome 19 ,Immunology ,Genetics ,medicine ,Cancer research ,Hyperdiploidy ,Childhood Acute Lymphoblastic Leukemia ,Fluorescence in situ hybridization - Abstract
The t(1;19)(q23;p13) or its derivative encodes an E2A-PBX1 fusion transcript and protein that has been shown to have important prognostic and therapeutic implications in patients with acute lymphoblastic leukemia (ALL). We describe two childhood cases in which a der(22)t( 1;22)(q21-23;p13) cytogenetically mimicked a der( 19)t( 1; 19)(q23;p 13). In one case, which was phenotyped as early pre-B ALL with hyperdiploidy but lacked evidence of an E2A-PBX1 gene fusion by molecular study, the poor banding quality of chromosomes led to misinterpretation of the cytogenetic findings; a correct diagnosis was established only after analysis by the fluorescence in situ hybridization (FISH) method. The second case, which was classified as pseudodiploid pre-B ALL, had both a derivative 19 and a derivative 22 but lacked sufficient cells for evaluation of E2A-PBX1 gene fusion. This case was included in order to compare the der(19)t(1;19) and the der(22)t(1;22) and to pinpoint the difficulty in distinguishing these markers. FISH analysis can resolve diagnostic uncertainty in cases of ALL with equivocal chromosome 19 markers. © 1995 Wiley-Liss, Inc.
- Published
- 1995
238. Abstract 2628: Molecular diagnosis for pediatric cancer through integrative analysis of whole-genome, whole-exome and transcriptome sequencing
- Author
-
Michael Rusch, Bhavin Vadodaria, Zhaojie Zhang, Jiali Gu, Yongjin Li, Donald Yergeau, Andrew Thrasher, Xiaotu Ma, David W. Ellison, Erin Hedlund, John Easton, Sheila A. Shurtleff, Xiang Chen, Michael N. Edmonson, Aman Patel, Rose B. McGee, James R. Downing, Joy Nakitandwe, Tanja A. Gruber, Matthew Parker, Jared Becksfort, and Jinghui Zhang
- Subjects
Transcriptome ,Loss of heterozygosity ,Structural variation ,Cancer genome sequencing ,Genetics ,Cancer Research ,Germline mutation ,Oncology ,Genome project ,Biology ,Exome ,Pediatric cancer - Abstract
Next-generation sequencing (NGS) of the whole genome, whole exome, and transcriptome has enabled characterization of genetic landscapes of multiple cancers. By analyzing over 2,000 pediatric cancer patients, we have developed a comprehensive database for recurrent somatic alterations and pathogenic germline mutations as part of the St. Jude/Washington University Pediatric Cancer Genome Project. However, there is no systematic evaluation on whether NGS is able to identify germline and somatic lesions reported by existing molecular diagnostic assays and what combination of NGS platforms is best suited for clinical sequencing. Here we report the first comprehensive study that employs whole-genome sequencing at 30-45X coverage, whole-exome sequencing at 100X coverage and transcriptome sequencing using matched tumor/normal samples from cancer patients. A pilot study was carried out to perform NGS analysis on 78 children of leukemia, solid tumor or brain tumor with a total of 112 diagnostic or prognostic biomarkers previously characterized by multiple molecular diagnostic assays. We implemented an analysis pipeline that integrates the genetic lesions detected by all three NGS platforms to characterize somatic and germline single nucleotide variations (SNVs), short insertions and deletions (indels), structural variations including fusions, karyotypes, copy number alterations, loss of heterozygosity, tumor purity and tumor-in-normal contamination. The turn-around time for data analysis is 2 weeks with an overall sensitivity of 99% on detecting known biomarkers. Extensive validation of >3,000 somatic sequence mutations or structural variations from 38 cases shows that the specificity for somatic SNV, indel and structural variation is at 98%, 95% and 84% across the genome. We demonstrate that in addition to providing cross-validation, multi-platform NGS is required for detecting all genetic lesions of pathological significance including complex re-arrangements such as chromothripsis. In addition to known pathogenic or likely pathogenic mutations, our analysis has also unveiled novel pathogenic mutations (e.g. a germline deletion in TP53 in one patient with medulloblastoma) and identified multiple variants of unknown significance that may be worth further exploration (e.g. an in-frame deletion of exons 3-9 of DNMT3A in one neuroblastoma). Our study demonstrates that NGS is able to detect a wide range of genetic lesions currently characterized by multiple molecular diagnostic assays, providing critical insight into the design of clinical sequencing for ongoing studies. Citation Format: Jinghui Zhang, Michael Rusch, Joy Nakitandwe, Zhaojie Zhang, Michael N. Edmonson, Matthew Parker, Xiaotu Ma, Jared Becksfort, Andrew Thrasher, Jiali Gu, Yongjin Li, Erin Hedlund, Aman Patel, John Easton, Donald Yergeau, Bhavin Vadodaria, Xiang Chen, Tanja A. Gruber, Rose McGee, David Ellison, Sheila Shurtleff, James R. Downing. Molecular diagnosis for pediatric cancer through integrative analysis of whole-genome, whole-exome and transcriptome sequencing. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2628.
- Published
- 2016
239. Abstract 2436: Exploring genomic alterations in pediatric cancer using ProteinPaint
- Author
-
Aman Patel, Mark R. Wilkinson, Zhaojie Zhang, James R. Downing, Xin Zhou, Jinghui Zhang, Yongjin Li, Matthew Parker, Michael N. Edmonson, Jared Becksfort, Gang Wu, Michael Rusch, and Yu Liu
- Subjects
Genetics ,Gene expression profiling ,Cancer Research ,Germline mutation ,COSMIC cancer database ,Oncology ,Biology ,Indel ,Somatic evolution in cancer ,Gene ,Pediatric cancer ,Germline - Abstract
Current cancer genome data portals have focused primarily on presenting data generated from adult cancer studies. These portals typically lack features for exploring pathogenic germline mutations, somatic gene fusions, and gene expression profiling, all of which are important biomarkers for risk stratification of pediatric cancer. We have developed ProteinPaint (https://pecan.stjude.org/proteinpaint/), a web service hosting 30,000+ validated somatic SNV/indels and fusion transcripts detected in 1,654 pediatric tumor samples from 17 subtypes, 252 pathogenic or loss-of-function germline lesions detected in >1000 pediatric cancer patients of 21 subtypes, and gene expression profiles derived from RNA-Seq of 928 pediatric tumors. Cancer genomic alterations are shown on novel “disc-on-stem” skewer graphs which were designed to depict the diverse prevalence, complex allelic alteration, and temporal origin of sequence mutations and gene fusions. Adult somatic cancer mutation data from the COSMIC database can be displayed in parallel with pediatric cancer data sets for cross-study comparison. We will demonstrate examples of how ProteinPaint's integrative view of genomic alteration, gene expression and pediatric-adult data comparison has facilitated the evaluation of somatic and germline mutation pathogenicity in a clinical setting. Custom data including sequence mutations in the MAF format used by the Cancer Genome Atlas (TCGA) project, copy number alterations, and structural variations can all be imported and visualized alongside published pediatric and adult cancer data sets. Furthermore, ProteinPaint supports curation and annotation of fusion transcripts predicted from RNASeq data and analysis of tumor clonal evolution with a 2-D plot of mutation frequency of paired diagnosis and relapse samples. ProteinPaint delivers a premium user experience with animation and interactive features for visualizing large cancer mutation datasets, and can serve as a workbench to import, explore and interpret user data. Its framework continues to expand as its intuitive visualization has enabled non-bioinformatics scientists and clinicians to access and manipulate genomic data for discovery and clinical reporting. Citation Format: Xin Zhou, Michael N. Edmonson, Mark R. Wilkinson, Aman Patel, Gang Wu, Yu Liu, Yongjin Li, Zhaojie Zhang, Michael Rusch, Matthew Parker, Jared Becksfort, James R. Downing, Jinghui Zhang. Exploring genomic alterations in pediatric cancer using ProteinPaint. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2436.
- Published
- 2016
240. LG-10GENETIC ALTERATIONS IN UNCOMMON LOW-GRADE NEUROEPITHELIAL TUMORS: BRAF, FGFR1, AND MYB MUTATIONS OCCUR AT HIGH FREQUENCY AND ALIGN WITH MORPHOLOGY
- Author
-
Bo Tang, Chandanamali Punchihewa, James R. Downing, Teresa Santiago, Jinghui Zhang, Frederick A. Boop, Ruth G. Tatevossian, Ibrahim Qaddoumi, Ji Wen, Amar Gajjar, James Dalton, John Easton, Paul Klimo, Sheila A. Shurtleff, Wilda Orisme, Suzanne J. Baker, Gang Wu, Kelly Haupfear, and David W. Ellison
- Subjects
Genetics ,Abstracts ,Cancer Research ,Oncology ,Fibroblast growth factor receptor 1 ,Neuroepithelial tumors ,Mutation (genetic algorithm) ,Cancer research ,Morphology (biology) ,MYB ,Neurology (clinical) ,Biology - Published
- 2016
241. High-resolution genomic profiling of chronic lymphocytic leukemia reveals new recurrent genomic alterations
- Author
-
Jing Ma, Peter Lichter, Michael W.M. Kühn, Lars Bullinger, Hartmut Döhner, Michael Hallek, Xiaoping Su, Stephan Stilgenbauer, Florian Miller, Thorsten Zenz, Karlheinz Holzmann, Jan O. Korbel, Dirk Winkler, Raymonde Busch, Andreas Gerhardinger, Johannes Bloehdorn, James R. Downing, Daniel Mertens, Jennifer Edelmann, Ina Radtke, Andreas Bühler, and Stanley Pounds
- Subjects
Male ,DNA Copy Number Variations ,Sequence analysis ,Chronic lymphocytic leukemia ,Immunology ,Immunoglobulin Variable Region ,Loss of Heterozygosity ,Locus (genetics) ,Kaplan-Meier Estimate ,Biology ,Biochemistry ,Polymorphism, Single Nucleotide ,Loss of heterozygosity ,medicine ,Humans ,Gene ,In Situ Hybridization, Fluorescence ,Oligonucleotide Array Sequence Analysis ,Chromosome Aberrations ,medicine.diagnostic_test ,Gene Expression Profiling ,Cell Biology ,Hematology ,Genomics ,medicine.disease ,Molecular biology ,Leukemia, Lymphocytic, Chronic, B-Cell ,Gene expression profiling ,Mutation ,Immunoglobulin heavy chain ,Female ,Tumor Suppressor Protein p53 ,Immunoglobulin Heavy Chains ,Fluorescence in situ hybridization - Abstract
To identify genomic alterations in chronic lymphocytic leukemia (CLL), we performed single-nucleotide polymorphism–array analysis using Affymetrix Version 6.0 on 353 samples from untreated patients entered in the CLL8 treatment trial. Based on paired-sample analysis (n = 144), a mean of 1.8 copy number alterations per patient were identified; approximately 60% of patients carried no copy number alterations other than those detected by fluorescence in situ hybridization analysis. Copy-neutral loss-of-heterozygosity was detected in 6% of CLL patients and was found most frequently on 13q, 17p, and 11q. Minimally deleted regions were refined on 13q14 (deleted in 61% of patients) to the DLEU1 and DLEU2 genes, on 11q22.3 (27% of patients) to ATM, on 2p16.1-2p15 (gained in 7% of patients) to a 1.9-Mb fragment containing 9 genes, and on 8q24.21 (5% of patients) to a segment 486 kb proximal to the MYC locus. 13q deletions exhibited proximal and distal breakpoint cluster regions. Among the most common novel lesions were deletions at 15q15.1 (4% of patients), with the smallest deletion (70.48 kb) found in the MGA locus. Sequence analysis of MGA in 59 samples revealed a truncating mutation in one CLL patient lacking a 15q deletion. MNT at 17p13.3, which in addition to MGA and MYC encodes for the network of MAX-interacting proteins, was also deleted recurrently.
- Published
- 2012
242. Comparative analysis of different approaches to measure treatment response in acute myeloid leukemia
- Author
-
Dario Campana, Ching-Hon Pui, Hiroto Inaba, Susana C. Raimondi, Xueyuan Cao, Jeffrey W. Taub, Mihaela Onciu, Sheila A. Shurtleff, Gary V. Dahl, Jeffrey E. Rubnitz, James R. Downing, Barbara A. Degar, Elaine Coustan-Smith, Raul C. Ribeiro, Stanley Pounds, W. Paul Bowman, Kathleen Y. Wang, Jeffrey Jacobsen, and Wing Leung
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Pathology ,Myeloid ,Neoplasm, Residual ,Adolescent ,Kaplan-Meier Estimate ,Sensitivity and Specificity ,Severity of Illness Index ,Disease-Free Survival ,Flow cytometry ,Cohort Studies ,Sex Factors ,Bone Marrow ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,Child ,Survival analysis ,Retrospective Studies ,Analysis of Variance ,medicine.diagnostic_test ,business.industry ,Reverse Transcriptase Polymerase Chain Reaction ,Age Factors ,Myeloid leukemia ,ORIGINAL REPORTS ,medicine.disease ,Flow Cytometry ,Minimal residual disease ,Immunohistochemistry ,Survival Analysis ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Female ,Bone marrow ,business ,Monte Carlo Method - Abstract
Purpose In acute myeloid leukemia (AML), initial treatment response by morphologic analysis of bone marrow predicts long-term outcome. Response can now be assessed by minimal residual disease (MRD) monitoring with flow cytometry or polymerase chain reaction (PCR). We determined the relation among the results of these approaches and their prognostic value. Patients and Methods In the multicenter AML02 study, follow-up bone marrow samples from 203 children and adolescents with newly diagnosed AML were examined by flow cytometry (n = 1,514), morphology (n = 1,382), and PCR amplification of fusion transcripts (n = 508). Results were correlated with treatment outcome. Results Among 1,215 samples with less than 5% leukemic myeloblasts by morphology, 100 (8.2%) were MRD positive (≥ 0.1%) by flow cytometry, whereas 96 (57.5%) of the 167 samples with ≥ 5% blasts were MRD negative. Virtually all (308 of 311; 99.0%) MRD-negative samples by PCR were also MRD negative by flow cytometry. However, only 19 (9.6%) of the 197 PCR-positive samples were flow cytometry positive, with analyses of AML1-ETO and CBFβ-MYH11 accounting for most discrepancies, whereas eight of 13 MLL-positive samples had detectable MRD by flow cytometry. MRD by flow cytometry after induction 1 or 2 predicted lower event-free survival and higher relapse rate (P < .001) and was an independent prognostic factor in a multivariable analysis; prediction was not improved by morphologic information or molecular findings. Conclusion In childhood AML, morphologic assessment of treatment response has limited value if MRD is measured by flow cytometry. MLL fusion transcripts can provide prognostic information in some patients, whereas monitoring of AML1-ETO and CBFβ-MYH11 transcripts is largely uninformative.
- Published
- 2012
243. Molecular genetics of acute myeloid leukemia
- Author
-
Tanja A. Gruber and James R. Downing
- Published
- 2012
244. The Pediatric Cancer Genome Project
- Author
-
Richard K. Wilson, Li Ding, Jinghui Zhang, Ching-Hon Pui, Timothy J. Ley, James R. Downing, Elaine R. Mardis, and William E. Evans
- Subjects
Lineage (genetic) ,Adolescent ,Biology ,medicine.disease_cause ,Article ,Acute megakaryoblastic leukemia ,Neoplasms ,Databases, Genetic ,Human Genome Project ,Genetics ,medicine ,Humans ,Rhabdomyosarcoma ,Child ,Mutation ,Genome, Human ,Myeloid leukemia ,Cancer ,Infant ,Sequence Analysis, DNA ,medicine.disease ,Pediatric cancer ,Leukemia ,Child, Preschool - Abstract
Subject terms: Cancer genomics• Paediatric cancer• Sequencing At a glance Figures View all figures Figure 1: Frequency of cancer diagnoses and leukemia subtypes in children and adults. (a) The frequency of cancer types in children (left) and adults (right) on the basis of 2012 Surveillance, Epidemiology and End Results (SEER) data. Each chart is organized with cancers listed from the most common to the least common in a clockwise fashion. (b) The frequency of T-cell lineage (blue text) and B-cell lineage (black text) subtypes of acute lymphoblastic leukemia (ALL) in children (left) and adults (right). Each chart is organized with ALL subtypes listed from the most common to the least common in a clockwise fashion. iAMP21, intrachromosomal amplification of chromosome 21. Full size image View in article Figure 2: Genetic landscape of 15 different types of pediatric cancers determined from whole-genome sequencing of 260 tumors and matching germline samples. The number of somatic mutations in each sample, including single-nucleotide variations (SNVs), insertion and/or deletion events (indels) and structural variations, is shown as the height in the three-dimensional graph. Only high-quality variations or validated somatic mutations are included in the summary. CDS, protein-coding regions; tier 1, mutations in annotated genes; tier 2, mutations in non-coding conserved or regulatory regions; tier 3, mutations in non-repetitive, non-coding and non-conserved regions; tier 4, mutations in repetitive regions. Tier 2 and tier 3/tier 4 mutations were rescaled to 1/10 and 1/100 of the original counts to maintain a consistent scale with the results for other somatic lesions. INF, infant ALL; CBF, core-binding-factor acute myeloid leukemia; TALL, T-cell ALL; AMLM7, acute megakaryoblastic leukemia; HYPO, hypodiploid ALL; PHALL, Philadelphia chromosome–positive BCR-ABL1 ALL; RB, retinoblastoma; RHB, rhabdomyosarcoma; NBL, neuroblastoma; OS, osteosarcoma; ACT, adrenocortical carcinoma; HGG, high-grade glioblastoma; LGG, low-grade glioma; EPD, ependymoma; MB, medulloblastoma. Full size image View in article
- Published
- 2012
245. Association of Age at Diagnosis and Genetic Mutations in Patients with Neuroblastoma
- Author
-
Alberto S. Pappo, Armita Bahrami, Jared Becksfort, James R. Downing, Jianmin Wang, James T. Dalton, Robert S. Fulton, David H. Ellison, Charles Lu, Irene Y. Cheung, Catherine A. Billups, Adriana Heguy, Matthew Parker, Sara M. Federico, Richard K. Wilson, Michael A. Dyer, Xiang Chen, Jinghui Zhang, Elaine R. Mardis, Nai-Kong V. Cheung, Li Ding, Jianrong Wu, and Satish K. Tickoo
- Subjects
Male ,medicine.medical_specialty ,Pathology ,X-linked Nuclear Protein ,Adolescent ,DNA Mutational Analysis ,Context (language use) ,Article ,Cohort Studies ,Neuroblastoma ,Young Adult ,Internal medicine ,medicine ,Humans ,Young adult ,Stage (cooking) ,Age of Onset ,Neoplasm Metastasis ,Child ,ATRX ,In Situ Hybridization, Fluorescence ,business.industry ,DNA Helicases ,Infant, Newborn ,Infant ,Nuclear Proteins ,Telomere Homeostasis ,General Medicine ,Sequence Analysis, DNA ,medicine.disease ,Immunohistochemistry ,Child, Preschool ,Cohort ,Mutation ,Female ,Age of onset ,business ,Cohort study - Abstract
Neuroblastoma is diagnosed over a wide age range from birth through young adulthood, and older age at diagnosis is associated with a decline in survivability.To identify genetic mutations that are associated with age at diagnosis in patients with metastatic neuroblastoma.Whole genome sequencing was performed on DNA from diagnostic tumors and their matched germlines from 40 patients with metastatic neuroblastoma obtained between 1987 and 2009. Age groups at diagnosis included infants (0-18 months), children (18 months-12 years), and adolescents and young adults (≥12 years). To confirm the findings from this discovery cohort, validation testing using tumors from an additional 64 patients obtained between 1985 and 2009 also was performed. Formalin-fixed, paraffin-embedded tumor tissue was used for immunohistochemistry and fluorescence in situ hybridization. Telomere lengths were analyzed using whole genome sequencing data, quantitative polymerase chain reaction, and fluorescent in situ hybridization.Somatic recurrent mutations in tumors from patients with neuroblastoma correlated with the age at diagnosis and telomere length.In the discovery cohort (n = 40), mutations in the ATRX gene were identified in 100% (95% CI, 50%-100%) of tumors from patients in the adolescent and young adult group (5 of 5), in 17% (95% CI, 7%-36%) of tumors from children (5 of 29), and 0% (95% CI, 0%-40%) of tumors from infants (0 of 6). In the validation cohort (n = 64), mutations in the ATRX gene were identified in 33% (95% CI, 17%-54%) of tumors from patients in the adolescent and young adult group (9 of 27), in 16% (95% CI, 6%-35%) of tumors from children (4 of 25), and in 0% (95% CI, 0%-24%) of tumors from infants (0 of 12). In both cohorts (N = 104), mutations in the ATRX gene were identified in 44% (95% CI, 28%-62%) of tumors from patients in the adolescent and young adult group (14 of 32), in 17% (95% CI, 9%-29%) of tumors from children (9 of 54), and in 0% (95% CI, 0%-17%) of tumors from infants (0 of 18). ATRX mutations were associated with an absence of the ATRX protein in the nucleus and with long telomeres.ATRX mutations were associated with age at diagnosis in children and young adults with stage 4 neuroblastoma.clinicaltrials.gov Identifier: NCT00588068.
- Published
- 2012
246. High-resolution genomic profiling of adult and pediatric core-binding factor acute myeloid leukemia reveals new recurrent genomic alterations
- Author
-
Verena I. Gaidzik, Arnold Ganser, Peter Paschka, Jing Ma, James R. Downing, Richard F. Schlenk, Jennifer Edelmann, Raul C. Ribeiro, Stanley Pounds, Jürgen Krauter, Jeffrey E. Rubnitz, Ina Radtke, Jan Krönke, Lars Bullinger, Karlheinz Holzmann, Sheila A. Shurtleff, Michael W.M. Kühn, Jinjun Cheng, Juliane Gohlke, Hartmut Döhner, Konstanze Döhner, Asmaa Aq Quessar, Salil Goorha, and Xiaoping Su
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Adult ,Tumor suppressor gene ,DNA Copy Number Variations ,Oncogene Proteins, Fusion ,Sequence analysis ,Immunology ,Loss of Heterozygosity ,Biology ,Biochemistry ,Polymorphism, Single Nucleotide ,Loss of heterozygosity ,Recurrence ,medicine ,Humans ,Child ,Core binding factor acute myeloid leukemia ,Oligonucleotide Array Sequence Analysis ,Chromosome Aberrations ,Myeloid Neoplasia ,Gene Expression Regulation, Leukemic ,Gene Expression Profiling ,Core Binding Factors ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease ,Molecular biology ,Minimal residual disease ,Leukemia ,Leukemia, Myeloid ,Acute Disease ,Mutation ,Chromosome Deletion - Abstract
To identify cooperating lesions in core-binding factor acute myeloid leukemia, we performed single-nucleotide polymorphism-array analysis on 300 diagnostic and 41 relapse adult and pediatric leukemia samples. We identified a mean of 1.28 copy number alterations per case at diagnosis in both patient populations. Recurrent minimally deleted regions (MDRs) were identified at 7q36.1 (7.7%), 9q21.32 (5%), 11p13 (2.3%), and 17q11.2 (2%). Approximately one-half of the 7q deletions were detectable only by single-nucleotide polymorphism-array analysis because of their limited size. Sequence analysis of MLL3, contained within the 7q36.1 MDR, in 46 diagnostic samples revealed one truncating mutation in a leukemia lacking a 7q deletion. Recurrent focal gains were identified at 8q24.21 (4.7%) and 11q25 (1.7%), both containing a single noncoding RNA. Recurrent regions of copy-neutral loss-of-heterozygosity were identified at 1p (1%), 4q (0.7%), and 19p (0.7%), with known mutated cancer genes present in the minimally altered region of 1p (NRAS) and 4q (TET2). Analysis of relapse samples identified recurrent MDRs at 3q13.31 (12.2%), 5q (4.9%), and 17p (4.9%), with the 3q13.31 region containing only LSAMP, a putative tumor suppressor. Determining the role of these lesions in leukemogenesis and drug resistance should provide important insights into core-binding factor acute myeloid leukemia.
- Published
- 2012
247. Expression of an Oncogenic ERG isoform Characterizes a Distinct Subtype of B-Progenitor Acute Lymphoblastic Leukemia
- Author
-
Stephen P. Hunger, Martin S. Tallman, Panagiotis Ntziachristos, Lei Wei, Janis Racevskis, John Easton, Michael N. Edmonson, Michelle L. Churchman, Kathryn G. Roberts, James R. Downing, Cheng Cheng, Marcus L Valentine, Kerri Ochoa, Selina M. Luger, Hiroki Yoshihara, Meenakshi Devidas, Elaine R. Mardis, Steven M. Kornblau, Peter M. Voorhees, Deqing Pei, Chunxu Qu, Guido Marcucci, Debbie Payne-Turner, Jessica Kohlschmidt, Jing Ma, Jinghui Zhang, Richard C. Harvey, Yongjin Li, Heather L. Mulder, Robert S. Fulton, Mignon L. Loh, Charles Lu, William E. Evans, Wendy Stock, Iannis Aifantis, Michael Rusch, Jacob M. Rowe, Xiang Chen, Esmé Waanders, Kelly McCastlain, Clara D. Bloomfield, Kristy Boggs, Li Ding, Ching-Hon Pui, Charles G. Mullighan, Guangchun Song, Susana C. Raimondi, Richard K. Wilson, Elisabeth Paietta, I-Ming L. Chen, Sheila A. Shurtleff, Cheryl L. Willman, Bhavin Vadodaria, Krzysztof Mrózek, Lucinda Fulton, Gang Wu, James Dalton, Yashodan Tabib, and Sima Jeha
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Gene expression profiling ,Leukemia ,Exon ,SETD2 ,medicine ,Gene ,Exome ,Exome sequencing - Abstract
Introduction. Prior studies have described a subset of B-progenitor ALL cases with a distinct gene expression profile and/or deletions involving ERG (encoding the ETS family member v-ets avian erythroblastosis virus E26 oncogene), however the relationship of these alterations and their role in leukemogenesis are poorly understood. We performed integrated genomic and epigenetic analyses, biochemical studies and leukemogenesis assays to define the genetic basis of this form of ALL. Methods. We studied 1674 childhood, adolescent and young adult B-progenitor ALL cases with microarray gene expression profiling and/or RNA-sequencing data to enable the identification of ERG ALL by unsupervised clustering and predictive analysis of microarrays. Detailed genomic analysis was performed for 144 ERG ALL cases, including whole genome (N=38), exome (n=46) and/or RNA-sequencing (n=57) cases, and single nucleotide polymorphism array analysis. Epigenetic profiling, including whole genome bisulfite sequencing, chromatin immunoprecipitation and sequencing for ERG and histone modifications and ATAC-sequencing were performed for a subset of 8 xenografted ERG tumors and reference cell lines. ERG transcript expression was measured by analysis of RNA-seq analysis and quantitative RT-PCR assays, and by interrogation of TCGA and PCGP RNA-seq data. The function of ERG isoforms was evaluated by EMSA and transcriptional reporter assays, immunofluoresence, colony forming assays and retroviral bone marrow transplant assays. Results. One hundred and forty four cases (8.6%) of B-ALL cases exhibited a distinct gene expression profile and lacked known chromosomal rearrangements (ERG ALL). Such cases had favorable outcome. Eighty cases (55.6%) had focal deletions of ERG with no evidence of oncogenic or chimeric ERG fusions. The deletions were most commonly heterozygous and involving exons 3-7 (n=27) or 3-9 (n=22) of 10 coding exons, and less commonly involving exon 1, or a larger region of the gene. No ERG deletions were identified in non-ERG ALL. Two cases harbored missense mutations in the ETS domain. Analysis of whole genome and exome sequencing data of 71 cases identified a high frequency of alterations of lymphoid transcription factors (46.5%; IKZF1 36.7%, PAX5 11.3%); mutation of transcription factors otherwise uncommon in ALL (21%; MYC, MYCBP2, MGA, ZEB2, GATA3); activation of signaling pathways, most commonly NRAS or KRAS (35.2%); cell cycle regulation (22.5%); and epigenetic modifiers (56.3%), most commonly KMT2D, SETD2, ARID2 and NCOR1. Notably, the five year event-free survival of ERG ALL cases with IKZF1 alterations exceeded 85% in both St Jude and Children's Oncology Group cohorts. We observed striking transcriptional deregulation at the ERG locus. Most (51/56) ERG- deleted cases expressed an ERG isoform encoded by a novel exon in intron 6 that splices in frame to distal exons, resulting in expression of a truncated C-terminal ERG protein that lacks the pointed and central regulatory domains, but retains the ETS and transactivation domain (ERGalt). ERGalt was also present in most (36/44) cases lacking an ERG deletion, and was strongly associated with presence of ERGalt protein in leukemic cells. We also identified expression of an Antisense Long non-coding RNA associated with the ERG locus (ALE) in ERG ALL. ERGalt and ALE were absent, or uncommonly expressed at very low levels in non-ERG ALL. ERGalt was absent, and ALE rarely expressed in non-ALL PCGP and TCGA samples. ERGalt and point mutant ERG were retained in the nucleus, bound DNA targets and acted as competitive inhibitors of wild type (WT) ERG in transcriptional reporter assays. Lineage-negative Arf -null bone marrow cells transduced with ERG WT induced an aggressive erythro-megakaryoblastic leukemia; in contrast ERGalt induced an immature lymphoid progenitor leukemia. Conclusions. Genomic alterations drive aberrant transcription of ERG, resulting on expression of a truncated, C-terminal oncogenic ERG protein. This represents a novel mechanism of transcription factor deregulation in leukemia. As a subset of ERG ALL cases lack ERG deletion, and as IKZF1 alterations are not associated with inferior outcome in this form of ALL, diagnostic approaches must incorporate gene expression profiling in addition to identification of ERG and IKZF1 alterations to accurately identify this form of leukemia. Disclosures Evans: Prometheus Labs: Patents & Royalties: Royalties from licensing TPMT genotyping. Stock:Gilead: Membership on an entity's Board of Directors or advisory committees. Voorhees:Oncopeptides: Consultancy; Onyx Pharmaceuticals: Research Funding; GSK: Consultancy; Oncopeptides: Research Funding; Janssen: Research Funding; A Takeda Oncology Company: Consultancy, Research Funding; Celgene: Consultancy; Millennium Pharmaceuticals: Consultancy, Research Funding; Acetylon Pharmaceuticals, Inc.: Research Funding; Novartis: Consultancy; Array BioPharma: Consultancy; GSK: Research Funding; Celgene: Research Funding. Hunger:Spectrum Pharmaceuticals: Consultancy; Jazz Pharmaceuticals: Consultancy; Sigma Tau: Consultancy; Merck: Equity Ownership. Mullighan:Incyte: Consultancy; Amgen: Honoraria.
- Published
- 2015
248. The Genomic Landscape of Childhood T-Lineage Acute Lymphoblastic Leukemia
- Author
-
Meenakshi Devidas, James R. Downing, Kimberly P. Dunsmore, Xiaotu Ma, Ying Shao, Brent L. Wood, Mark R. Wilkinson, John Easton, Mary V. Relling, Daniela S. Gerhard, Yu Liu, Jaime M. Guidry Auvil, Mignon L. Loh, Jinghui Zhang, Richard C. Harvey, William L. Carroll, Michael Rusch, Naomi J. Winick, Elizabeth A. Raetz, Stuart S. Winter, Cheryl L. Willman, Malcolm A. Smith, Stephen P. Hunger, Charles G. Mullighan, and Michael N. Edmonson
- Subjects
Genetics ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Molecular biology ,Exon ,CDKN2A ,Missense mutation ,MYB ,DDX3X ,Exome ,Gene ,Exome sequencing - Abstract
Comprehensive studies examining the genomic landscape of T-lineage ALL are lacking, but are important to identify all oncogenic drivers. Here we report sequencing of 264 T-ALL consecutive cases treated on the Children's Oncology Group AALL0434 clinical trial. Whole exome sequencing, copy number analysis using exome and single nucleotide polymorphism array analysis of tumor and remission DNA, and RNA-sequencing of tumor RNA were performed. Cases with immunophenotypic data (N=189) included 19 early T-cell precursor (ETP) cases, 24 near-ETP (with normal CD5 expression) and 146 Non-ETP cases. Median exomic coverage was 89% (72%-96%) of exons with at least 20-fold coverage. We identified 4657 non-synonymous clonal and subclonal somatic mutations (3926 single nucleotide variants (SNV) and 731 insertion-deletion mutations; indels) in 3030 genes, with a mean of 17.6 per case (range 1-50). 176 potential driver genes were identified statistical analysis or by known pathogenic role in cancer. These included NOTCH1 (n=194, 73%), FBXW7 (n=64, 24%), PHF6 (n=50, 19%), PTEN (n=37, 14%), USP7 (n=32, 12%), DNM2 (n=29, 11%) and BCL11B (n=27, 10%). New mutations in T-ALL included CCND3 (n=15, 6%), MYB (n=13, 5%), CTCF (n=13, 5%), MED12 (n=7, 3%), USP9X (n=7, 3%), SMARCA4 (n=7, 3%) and CREBBP (n=6, 2%). In addition to MYB amplification, we identified missense mutations and in-frame protein insertions at the N-terminus of MYB, with a hotspot at codon 14 in a region of six acidic residues in an otherwise hydrophilic N-terminal tail. These mutations resulted in a disordered region that is predicted to affect nuclear localization. The MYB mutations detected were enriched in non-ETP cases (n=13; 8 non-ETP, 1 near-ETP, 4 unknown). Other genes enriched in non-ETP cases included RPL10, CNOT3, MYCN and DDX3X. MED12 mutations were more common in ETP ALL. Sub-clonal mutations (mutant allele fraction of less than 30%) were identified in 111 of 176 driver genes in 198 (75%) cases including NOTCH1 (n=94), FBXW7 (n=29) and PTEN (n=17) indicating that sub-clonal evolution is a hallmark of T-ALL. In addition, multiple mutations in individual genes were commonly observed in single cases. For example, up to 3 different somatic NOTCH1 mutations were detected in each of 9 patients, with 2 different NOTCH1 mutations in 49 cases. Integration of sequence mutations with copy number aberration data showed the following pathways to be most frequently mutated: cell cycle/tumor suppression (N=225; CDKN2A/B (n=206), CDKN1B (n=35), RB1 (n=28)); NOTCH1/FBXW7 (n=212), PI3K-AKT (n=130), JAK-STAT (n=99) and Ras (n=51). Mutations in the PI3K-AKT, JAK-STAT and Ras signaling pathways were mutually exclusive. We identified a high frequency of mutations in transcriptional regulators in 222 cases, including 108 cases with mutations in a core regulatory complex comprising TAL1 (n=51), MYB (n=45) RUNX1 (n=18) and GATA3 (n=13). In 90 of these 108 cases (83%), only a single mutation was present in any of the four genes, consistent with a central role of this complex in leukemogenesis. Epigenetic alterations were identified in 178 cases, including PHF6 (n=63), SMARCA4 (n=23), KDM6A (n=22) and EZH2 (n=18), and new deletions and mutations in KMT2A (MLL; n=11). Interim analysis of transcriptome sequencing data of 126 T-ALL cases detected fusions in 61 (48%) samples, which could be separated into two categories. One weres in-frame fusions resulting in a chimeric protein. The most frequent included MLLT10 fusions (PICALM-MLLT10 (n=3), DDX3X-MLLT10 (n=2) and NAP1L1-MLLT10 (n=1)), KMT2A fusions (KMT2A-MLLT1 (n=4), MLLT6-KMT2A (n=1) and MKT2A-MLLT4 (n=1)), as well as internal tandem duplication mutations involving FLT3 (n=6; 3 ETP, 1 near-ETP, 1 non-ETP, 1 unknown) and NOTCH1 (n=2). We also identified novel fusions including ETV6-CTNNB1 and STMN1-SPI1 (n=1 each). The other category contains rearrangement-driven aberrant expression, including rearrangements in TLX1 (n=11), TLX3 (n=4), TAL1 (n=2), and TAL2 (n=3). Moreover, we found a novel TAL2 transcript in all the 3 cases with TAL2 rearrangement, hijacking a new exon 6kb upstream of the canonical TAL2 transcription start site and extending approximate 3.5kb downstream. These findings provide the first comprehensive landscape of genomic alterations in T-ALL and have provided new insights into the genes and pathways mutated in this disease, their interaction, and the nature of clonal heterogeneity in T-ALL. Disclosures Hunger: Spectrum Pharmaceuticals: Consultancy; Jazz Pharmaceuticals: Consultancy; Sigma Tau: Consultancy; Merck: Equity Ownership. Mullighan:Cancer Science Institute: Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Speakers Bureau; Incyte: Consultancy, Honoraria; Loxo Oncology: Research Funding.
- Published
- 2015
249. Genomic Landscape of Relapsed Acute Lymphoblastic Leukemia
- Author
-
Scott R. Olsen, Sima Jeha, Jinghui Zhang, Yiping Fan, Stephanie M. Dobson, William E. Evans, Pankaj Gupta, Gang Wu, Esmé Waanders, Xiaotu Ma, Geoffrey Neale, Colin Bailey, John E. Dick, Ying Shao, Ilaria Iacobucci, Kathryn G. Roberts, Kelly McCastlain, Jun J. Yang, Ching-Hon Pui, Charles G. Mullighan, Ji Wen, Jing Ma, James R. Downing, Mary V. Relling, John Easton, Matthew Lear, Deanna Naeve, Guangchun Song, Michael Rusch, Shann-Ching Chen, Zhaohui Gu, and Debbie Payne-Turner
- Subjects
Oncology ,Neuroblastoma RAS viral oncogene homolog ,medicine.medical_specialty ,business.industry ,Immunology ,Cancer ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease_cause ,medicine.disease ,Biochemistry ,Somatic evolution in cancer ,Leukemia ,Internal medicine ,medicine ,KRAS ,business ,EP300 ,Exome sequencing - Abstract
Introduction: Despite risk stratification according to presenting clinical and genetic features, 10-25% of children with acute lymphoblastic leukemia (ALL) relapse, which is associated with a poor prognosis. Here, we sought to provide a comprehensive overview of the genetic alterations associated with relapse in ALL. Methods: We studied 93 children (27 female, 43 male) diagnosed with ALL (62 B-progenitor, 25 T-lineage) between 1987 and 2008 and treated on total therapy studies XI-XVI who experienced relapse and/or a second tumor. Age at diagnosis ranged from 3 months to 18 years. Median time to relapse was 3 years (range 3 months to 10 years). Seventy patients had a single relapse, 15 cases had 2 relapses, and 8 cases developed a second tumor of different lineage (B-cell lymphoma, chronic myeloid leukemia (n=1 each) and acute myeloid leukemia (n=6)). Diagnosis, relapse and matched normal samples (n=299) were studied using Affymetrix SNP 6.0 microarrays and whole genome or whole exome sequencing. Results: We found 2692 copy number aberrations (CNAs) with a median of 9 (range 0-109) in the diagnosis samples (n=91) compared to a median of 10 (range 0-112) in the relapse samples (n=89) and 12 (range 0-70) in subsequent samples (n=20). The number of CNAs did not differ significantly between diagnosis, relapse or subsequent samples. We identified a 7286 non-silent single nucleotide variants (SNVs) and small insertions or deletions (indels) in 5002 genes, 1392 of which were recurrent. The median number of variants was 12 (range 0-70) at diagnosis (n=91), 21 (range 0-858) at relapse (n=91; P=0.0029 v. diagnosis) and 60 (range 10-650) in subsequent samples (n=20; P The most frequently mutated genes were NOTCH1 (n=33), NRAS (n=24), CREBBP (n=20) and KRAS (n=16). Of the recurrently altered genes, only 87 genes were known to be affected in cancer (Cancer Gene Census, COSMIC database), of which 59 were affected in leukemia and lymphoma tissues, indicating that we have identified 1306 novel recurrently affected genes, most commonly C13orf40 and MKI67. Mutations in epigenetic regulators were particularly frequent, with genes mutated in at least 3 cases altered in over 60% of the cohort (e.g. CREBBP, EP300, MLL2, MLL3, KDM6A/B, CTCF, SETD2, TET2/3, and EZH2). Clonal evolution analyses showed multiple patterns of evolution, with relapses sharing either few or many variants with the diagnosis sample in a frequency that reflects both predominant clones and minor subclones propagating relapse. Variants in NOTCH1, NRAS, and CREBBP were preserved from a major clone at diagnosis in 4, 6, and 5 cases respectively, but acquired at relapse or grown out from a minor subclone at diagnosis in 3, 5, and 8 cases respectively. In contrast, variants in USH2A (n=4), FOXA1 (n=3), and purine/pyrimidine synthesis pathway genes NT5C2 (n=3), PRPS1 (n=3) and NT5C1B (n=1) were exclusively found in relapse samples. Notably, the NT5C2 mutations, which are thought to confer resistance to thiopurines, were subclonal at relapse in the majority of cases. We identified 13 cases (10 B-lineage, 3 T-lineage) in which the diagnosis and relapse were fully discordant for all CNAs and sequence mutations, only 4 of which showed a prolonged remission time (>5 years). This suggests that these patients developed a second primary malignancy and may be predisposed to leukemia development. Indeed, one case revealed focal amplifications on chromosome 1q21.1 encompassing the neuroblastoma breakpoint family genes, which are implicated in cancer development. Comprehensive germline analyses are underway. Conclusion: This study has provided detailed insight into the genetic basis of relapse, implicating multiple new genes and pathways involved in treatment resistance, demonstrating multiple patterns of clonal evolution, and revealing an unexpectedly high frequency of genetically discordant second malignancy in relapse in ALL. Disclosures Evans: Prometheus Labs: Patents & Royalties: Royalties from licensing TPMT genotyping. Mullighan:Amgen: Honoraria, Speakers Bureau; Cancer Science Institute: Membership on an entity's Board of Directors or advisory committees; Incyte: Consultancy, Honoraria; Loxo Oncology: Research Funding.
- Published
- 2015
250. Next Generation Sequencing Identifies a Novel Subset of Non-Down Syndrome Acute Megakaryoblastic Leukemia Characterized By Chimeric Transcripts Involving HOX Cluster Genes
- Author
-
Jinjun Cheng, Jing Ma, Heather L. Mulder, Tanja A. Gruber, Michael Rusch, Allen Eng Juh Yeoh, Yongjin Li, Jasmijn D.E. de Rooij, Katarina Reinhardt, C. Michel Zwaan, John Easton, Maarten Forenerod, Stephanie Halene, Cary Koss, Jinghui Zhang, James R. Downing, Franco Locatelli, Dirk Reinhardt, Joshua Yew Suang Lim, Cristyn Branstetter, Der-Cherng Liang, Diane S. Krause, Lee-Yung Shih, Lonneke J. Verboon, and Marry M. van den Heuvel-Eibrink
- Subjects
Genetics ,Acute leukemia ,Immunology ,GATA1 ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Fusion gene ,Acute megakaryoblastic leukemia ,medicine ,Hox gene ,Exome ,Gene ,Exome sequencing - Abstract
Acute Megakaryoblastic Leukemia (AMKL) is a subtype of acute myeloid leukemia (AML) that morphologically resembles abnormal megakaryoblasts. While extremely rare in adults, pediatric cases comprise 4-15% of newly diagnosed AML patients. Clinical outcomes for Down syndrome (DS) patients with AMKL are uniformly excellent, whereas studies on non-DS patients (non-DS-AMKL) are more variable with the majority reporting inferior survival rates compared to other AML subtypes. Furthermore, the recommendation for stem cell transplant (SCT) in first remission for non-DS-AMKL patients is not uniform among pediatric cooperative groups. Previous efforts have identified chimeric oncogenes in non-DS-AMKL cases, including RBM15-MKL1, CBFA2T3-GLIS2, MLL gene rearrangements and NUP98-KDM5A. The etiology of 30-40% of cases, however, remains unknown. To better understand the genomic landscape of non-DS-AMKL and its contribution to clinical outcomes, we performed RNA and exome sequencing on specimens from 115 patients compiled from eight institutions and three cooperative groups including 90 pediatric and 25 adult cases. Of the 104 patients for whom RNA was available, 27.8% (5/18) adult and 72% (62/86) pediatric cases carried a high confidence fusion event by RNAseq. The most frequent fusions in the pediatric cohort when combining RNAseq data, cytogenetics and RT-PCR include CBFA2T3-GLIS2 (17/90), MLL r (13/90), NUP98-KDM5A (9/90), and RBM15-MKL1 (9/90). Previously described low frequency fusions identified in this expanded cohort, include a case of NIPBL-HOXB9 and a novel but functionally analogous NIPBL-HOXA9 fusion. Similarly, a case carrying GATA2-HOXA10 was identified, which is functionally equivalent to the GATA2-HOXA9 fusion that has been reported in a single case. Chimeric transcripts not previously described include several fusions involving genes within the HOX cluster (HOTAIRM1-HOXA3, HOXA_AS3-HOXA9, EWSR1-HOXB8, PLEK-HOXA11-AS, and BMP2K-HOXD10 each in a single case). Collectively, fusions involving a HOX cluster gene (HOX r) occurred in 11% of the pediatric cohort. Single Nucleotide Variation (SNV) analysis of exome and RNAseq data on the cohort revealed the presence of truncating GATA1 mutations in one adult and 10 pediatric specimens lacking fusion genes. Patients carrying GATA1 mutations did not have stigmata of DS or evidence of mutant reads in germline DNA, suggesting they are not mosaics. To determine if these fusion events contribute significantly to gene expression patterns, samples with greater than 60% purity were subjected to unsupervised clustering. Confirming the strength of the fusions in altering gene expression signatures, samples clustered according to fusion subtype and were distinct from those carrying GATA1 mutations. Specifically MLL r, HOX r, NUP98-KDM5A, and CBFA2T3-GLIS2 cases formed distinct clusters. When analyzing differentially upregulated genes within these subgroups, HOX r cases demonstrated upregulation of a HOX gene signature. Combined with MLL r and NUP98-KDM5A, chimeric oncogenes also known to upregulate HOX cluster genes, roughly one-third of pediatric non-DS-AMKL patients carry a HOX gene expression program. These cases were distinct from those carrying the CBFA2T3-GLIS2 inversion. HOX genes play a significant role in normal hematopoietic development and data suggests that deregulated expression has a central role in the etiology of several subtypes of acute leukemia, in part through the acquisition of enhanced self-renewal. We evaluated our identified HOXr for their ability to serially replate in murine colony formation assays as a surrogate marker of this characteristic. Confirming their pathogenicity, chimeric transcripts conferred an enhanced ability to replate. We conclude that chimeric transcripts involving HOX cluster genes comprise a distinct subset of pediatric AMKL. Clinical outcome analyses between genomic subgroups of this heterogeneous malignancy may allow us to more effectively risk stratify these patients and determine those that may benefit from SCT in first remission. JdR and CB contributed equally FL, DR, MH-E, MF, CMZ, and TAG co-corresponding authors on behalf of AIEOP, BFM, DCOG, and SJCRH study groups Disclosures Shih: Novartis: Research Funding.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.