Network


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

Hotspot


Dive into the research topics where Igor Dolgalev is active.

Publication


Featured researches published by Igor Dolgalev.


Cancer Cell | 2010

Integrative Genomic Profiling of Human Prostate Cancer

Barry S. Taylor; Nikolaus Schultz; Haley Hieronymus; Anuradha Gopalan; Yonghong Xiao; Brett S. Carver; Vivek K. Arora; Poorvi Kaushik; Ethan Cerami; Boris Reva; Yevgeniy Antipin; Nicholas Mitsiades; Thomas Landers; Igor Dolgalev; John Major; Manda Wilson; Nicholas D. Socci; Alex E. Lash; Adriana Heguy; James A. Eastham; Howard I. Scher; Victor E. Reuter; Peter T. Scardino; Chris Sander; Charles L. Sawyers; William L. Gerald

Annotation of prostate cancer genomes provides a foundation for discoveries that can impact disease understanding and treatment. Concordant assessment of DNA copy number, mRNA expression, and focused exon resequencing in 218 prostate cancer tumors identified the nuclear receptor coactivator NCOA2 as an oncogene in approximately 11% of tumors. Additionally, the androgen-driven TMPRSS2-ERG fusion was associated with a previously unrecognized, prostate-specific deletion at chromosome 3p14 that implicates FOXP1, RYBP, and SHQ1 as potential cooperative tumor suppressors. DNA copy-number data from primary tumors revealed that copy-number alterations robustly define clusters of low- and high-risk disease beyond that achieved by Gleason score. The genomic and clinical outcome data from these patients are now made available as a public resource.


The New England Journal of Medicine | 2012

Prognostic relevance of integrated genetic profiling in acute myeloid leukemia

Jay Patel; Mithat Gonen; Maria E. Figueroa; Hugo F. Fernandez; Zhuoxin Sun; Janis Racevskis; Pieter Van Vlierberghe; Igor Dolgalev; Sabrena Thomas; Olga Aminova; Kety Huberman; Janice Cheng; Agnes Viale; Nicholas D. Socci; Adriana Heguy; Athena M. Cherry; Gail H. Vance; Rodney R. Higgins; Rhett P. Ketterling; Robert E. Gallagher; Mark R. Litzow; Marcel R.M. van den Brink; Hillard M. Lazarus; Jacob M. Rowe; Selina M. Luger; Adolfo A. Ferrando; Elisabeth Paietta; Martin S. Tallman; Ari Melnick; Omar Abdel-Wahab

BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous disease with respect to presentation and clinical outcome. The prognostic value of recently identified somatic mutations has not been systematically evaluated in a phase 3 trial of treatment for AML. METHODS We performed a mutational analysis of 18 genes in 398 patients younger than 60 years of age who had AML and who were randomly assigned to receive induction therapy with high-dose or standard-dose daunorubicin. We validated our prognostic findings in an independent set of 104 patients. RESULTS We identified at least one somatic alteration in 97.3% of the patients. We found that internal tandem duplication in FLT3 (FLT3-ITD), partial tandem duplication in MLL (MLL-PTD), and mutations in ASXL1 and PHF6 were associated with reduced overall survival (P=0.001 for FLT3-ITD, P=0.009 for MLL-PTD, P=0.05 for ASXL1, and P=0.006 for PHF6); CEBPA and IDH2 mutations were associated with improved overall survival (P=0.05 for CEBPA and P=0.01 for IDH2). The favorable effect of NPM1 mutations was restricted to patients with co-occurring NPM1 and IDH1 or IDH2 mutations. We identified genetic predictors of outcome that improved risk stratification among patients with AML, independently of age, white-cell count, induction dose, and post-remission therapy, and validated the significance of these predictors in an independent cohort. High-dose daunorubicin, as compared with standard-dose daunorubicin, improved the rate of survival among patients with DNMT3A or NPM1 mutations or MLL translocations (P=0.001) but not among patients with wild-type DNMT3A, NPM1, and MLL (P=0.67). CONCLUSIONS We found that DNMT3A and NPM1 mutations and MLL translocations predicted an improved outcome with high-dose induction chemotherapy in patients with AML. These findings suggest that mutational profiling could potentially be used for risk stratification and to inform prognostic and therapeutic decisions regarding patients with AML. (Funded by the National Cancer Institute and others.).


The New England Journal of Medicine | 2010

Mutations in GNA11 in Uveal Melanoma

Catherine D. Van Raamsdonk; Klaus G. Griewank; Michelle B. Crosby; Maria C. Garrido; Swapna Vemula; Thomas Wiesner; Anna C. Obenauf; Werner Wackernagel; Gary G. R. Green; Nancy Bouvier; M. Mert Sozen; Gail Baimukanova; Ritu Roy; Adriana Heguy; Igor Dolgalev; Raya Khanin; Michael R. Speicher; Joan M. O'Brien; Boris C. Bastian

BACKGROUND Uveal melanoma is the most common intraocular cancer. There are no effective therapies for metastatic disease. Mutations in GNAQ, the gene encoding an alpha subunit of heterotrimeric G proteins, are found in 40% of uveal melanomas. METHODS We sequenced exon 5 of GNAQ and GNA11, a paralogue of GNAQ, in 713 melanocytic neoplasms of different types (186 uveal melanomas, 139 blue nevi, 106 other nevi, and 282 other melanomas). We sequenced exon 4 of GNAQ and GNA11 in 453 of these samples and in all coding exons of GNAQ and GNA11 in 97 uveal melanomas and 45 blue nevi. RESULTS We found somatic mutations in exon 5 (affecting Q209) and in exon 4 (affecting R183) in both GNA11 and GNAQ, in a mutually exclusive pattern. Mutations affecting Q209 in GNA11 were present in 7% of blue nevi, 32% of primary uveal melanomas, and 57% of uveal melanoma metastases. In contrast, we observed Q209 mutations in GNAQ in 55% of blue nevi, 45% of uveal melanomas, and 22% of uveal melanoma metastases. Mutations affecting R183 in either GNAQ or GNA11 were less prevalent (2% of blue nevi and 6% of uveal melanomas) than the Q209 mutations. Mutations in GNA11 induced spontaneously metastasizing tumors in a mouse model and activated the mitogen-activated protein kinase pathway. CONCLUSIONS Of the uveal melanomas we analyzed, 83% had somatic mutations in GNAQ or GNA11. Constitutive activation of the pathway involving these two genes appears to be a major contributor to the development of uveal melanoma. (Funded by the National Institutes of Health and others.).


Blood | 2009

Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies

Omar Abdel-Wahab; Ann Mullally; Cyrus V. Hedvat; Guillermo Garcia-Manero; Jay Patel; Martha Wadleigh; Sébastien Malinge; Jin Juan Yao; Outi Kilpivaara; Rukhmi Bhat; Kety Huberman; Sabrena Thomas; Igor Dolgalev; Adriana Heguy; Elisabeth Paietta; Michelle M. Le Beau; Miloslav Beran; Martin S. Tallman; Benjamin L. Ebert; Hagop M. Kantarjian; Richard Stone; D. Gary Gilliland; John D. Crispino; Ross L. Levine

Disease alleles that activate signal transduction are common in myeloid malignancies; however, there are additional unidentified mutations that contribute to myeloid transformation. Based on the recent identification of TET2 mutations, we evaluated the mutational status of TET1, TET2, and TET3 in myeloproliferative neoplasms (MPNs), chronic myelomonocytic leukemia (CMML), and acute myeloid leukemia (AML). Sequencing of TET2 in 408 paired tumor/normal samples distinguished between 68 somatic mutations and 6 novel single nucleotide polymorphisms and identified TET2 mutations in MPN (27 of 354, 7.6%), CMML (29 of 69, 42%), AML (11 of 91, 12%), and M7 AML (1 of 28, 3.6%) samples. We did not identify somatic TET1 or TET3 mutations or TET2 promoter hypermethylation in MPNs. TET2 mutations did not cluster in genetically defined MPN, CMML, or AML subsets but were associated with decreased overall survival in AML (P = .029). These data indicate that TET2 mutations are observed in different myeloid malignancies and may be important in AML prognosis.


Nature Genetics | 2013

The integrated landscape of driver genomic alterations in glioblastoma

Veronique Frattini; Vladimir Trifonov; Joseph Chan; Angelica Castano; Marie Lia; Francesco Abate; Stephen T. Keir; Alan X. Ji; Pietro Zoppoli; Francesco Niola; Carla Danussi; Igor Dolgalev; Paola Porrati; Serena Pellegatta; Adriana Heguy; Gaurav Gupta; David Pisapia; Peter Canoll; Jeffrey N. Bruce; Roger E. McLendon; Hai Yan; Kenneth D. Aldape; Gaetano Finocchiaro; Tom Mikkelsen; Gilbert G. Privé; Darell D. Bigner; Anna Lasorella; Raul Rabadan; Antonio Iavarone

Glioblastoma is one of the most challenging forms of cancer to treat. Here we describe a computational platform that integrates the analysis of copy number variations and somatic mutations and unravels the landscape of in-frame gene fusions in glioblastoma. We found mutations with loss of heterozygosity in LZTR1, encoding an adaptor of CUL3-containing E3 ligase complexes. Mutations and deletions disrupt LZTR1 function, which restrains the self renewal and growth of glioma spheres that retain stem cell features. Loss-of-function mutations in CTNND2 target a neural-specific gene and are associated with the transformation of glioma cells along the very aggressive mesenchymal phenotype. We also report recurrent translocations that fuse the coding sequence of EGFR to several partners, with EGFR-SEPT14 being the most frequent functional gene fusion in human glioblastoma. EGFR-SEPT14 fusions activate STAT3 signaling and confer mitogen independence and sensitivity to EGFR inhibition. These results provide insights into the pathogenesis of glioblastoma and highlight new targets for therapeutic intervention.


Nature Genetics | 2013

The mutational landscape of adenoid cystic carcinoma

Allen S. Ho; Kasthuri Kannan; David M Roy; Luc G. T. Morris; Ian Ganly; Nora Katabi; Deepa Ramaswami; Logan A. Walsh; Stephanie Eng; Jason T. Huse; Jianan Zhang; Igor Dolgalev; Kety Huberman; Adriana Heguy; Agnes Viale; Marija Drobnjak; Margaret Leversha; Christine E Rice; Bhuvanesh Singh; N. Gopalakrishna Iyer; C. René Leemans; Elisabeth Bloemena; Robert L. Ferris; Raja R. Seethala; Benjamin E. Gross; Yupu Liang; Rileen Sinha; Luke Peng; Benjamin J. Raphael; Sevin Turcan

Adenoid cystic carcinomas (ACCs) are among the most enigmatic of human malignancies. These aggressive salivary gland cancers frequently recur and metastasize despite definitive treatment, with no known effective chemotherapy regimen. Here we determined the ACC mutational landscape and report the exome or whole-genome sequences of 60 ACC tumor-normal pairs. These analyses identified a low exonic somatic mutation rate (0.31 non-silent events per megabase) and wide mutational diversity. Notably, we found mutations in genes encoding chromatin-state regulators, such as SMARCA2, CREBBP and KDM6A, suggesting that there is aberrant epigenetic regulation in ACC oncogenesis. Mutations in genes central to the DNA damage response and protein kinase A signaling also implicate these processes. We observed MYB-NFIB translocations and somatic mutations in MYB-associated genes, solidifying the role of these aberrations as critical events in ACC. Lastly, we identified recurrent mutations in the FGF-IGF-PI3K pathway (30% of tumors) that might represent new avenues for therapy. Collectively, our observations establish a molecular foundation for understanding and exploring new treatments for ACC.


Oncogene | 2012

Concurrent loss of the PTEN and RB1 tumor suppressors attenuates RAF dependence in melanomas harboring V600E BRAF

Feng Xing; Yogindra Persaud; Christine A. Pratilas; Barry S. Taylor; Manickam Janakiraman; Qing-Bai She; H. Gallardo; Cailian Liu; Taha Merghoub; B. Hefter; Igor Dolgalev; Agnes Viale; Adriana Heguy; E. de Stanchina; David Cobrinik; Gideon Bollag; Jedd D. Wolchok; Alan N. Houghton; David B. Solit

Identifying the spectrum of genetic alterations that cooperate with critical oncogenes to promote transformation provides a foundation for understanding the diversity of clinical phenotypes observed in human cancers. Here, we performed integrated analyses to identify genomic alterations that co-occur with oncogenic BRAF in melanoma and abrogate cellular dependence upon this oncogene. We identified concurrent mutational inactivation of the PTEN and RB1 tumor suppressors as a mechanism for loss of BRAF/MEK dependence in melanomas harboring V600EBRAF mutations. RB1 alterations were mutually exclusive with loss of p16INK4A, suggesting that whereas p16INK4A and RB1 may have overlapping roles in preventing tumor formation, tumors with loss of RB1 exhibit diminished dependence upon BRAF signaling for cell proliferation. These findings provide a genetic basis for the heterogeneity of clinical outcomes in patients treated with targeted inhibitors of the mitogen-activated protein kinase pathway. Our results also suggest a need for comprehensive screening for RB1 and PTEN inactivation in patients treated with RAF and MEK-selective inhibitors to determine whether these alterations are associated with diminished clinical benefit in patients whose cancers harbor mutant BRAF.


Cancer Research | 2015

Whole exome sequencing reveals frequent genetic alterations in BAP1, NF2, CDKN2A and CUL1 in malignant pleural mesothelioma

Guangwu Guo; Juliann Chmielecki; Chandra Goparaju; Adriana Heguy; Igor Dolgalev; Michele Carbone; Sara Seepo; Matthew Meyerson; Harvey I. Pass

Malignant pleural mesothelioma (MPM) is an aggressive neoplasm associated with asbestos exposure. Although previous studies based on candidate gene approaches have identified important common somatic mutations in MPM, these studies have focused on small sets of genes and have provided a limited view of the genetic alterations underlying this disease. Here, we performed whole-exome sequencing on DNA from 22 MPMs and matched blood samples, and identified 517 somatic mutations across 490 mutated genes. Integrative analysis of mutations and somatic copy-number alterations revealed frequent genetic alterations in BAP1, NF2, CDKN2A, and CUL1. Our study presents the first unbiased view of the genomic basis of MPM.


Cancer Discovery | 2016

Diverse and Targetable Kinase Alterations Drive Histiocytic Neoplasms

Eli L. Diamond; Benjamin H. Durham; Julien Haroche; Zhan Yao; Jing Ma; Sameer A. Parikh; Zhaoming Wang; John K. Choi; Eunhee Kim; Fleur Cohen-Aubart; Stanley Chun-Wei Lee; Yijun Gao; Jean Baptiste Micol; Patrick Campbell; Michael P. Walsh; Brooke E. Sylvester; Igor Dolgalev; Olga Aminova; Adriana Heguy; Paul Zappile; Joy Nakitandwe; Chezi Ganzel; James Dalton; David W. Ellison; Juvianee Estrada-Veras; Mario E. Lacouture; William A. Gahl; Philip J. Stephens; Vincent A. Miller; Jeffrey S. Ross

UNLABELLED Histiocytic neoplasms are clonal, hematopoietic disorders characterized by an accumulation of abnormal, monocyte-derived dendritic cells or macrophages in Langerhans cell histiocytosis (LCH) and non-Langerhans cell histiocytosis (non-LCH), respectively. The discovery of BRAF(V600E) mutations in approximately 50% of these patients provided the first molecular therapeutic target in histiocytosis. However, recurrent driving mutations in the majority of patients with BRAF(V600E)-wild-type non-LCH are unknown, and recurrent cooperating mutations in non-MAP kinase pathways are undefined for the histiocytic neoplasms. Through combined whole-exome and transcriptome sequencing, we identified recurrent kinase fusions involving BRAF, ALK, and NTRK1, as well as recurrent, activating MAP2K1 and ARAF mutations in patients with BRAF(V600E)-wild-type non-LCH. In addition to MAP kinase pathway lesions, recurrently altered genes involving diverse cellular pathways were identified. Treatment of patients with MAP2K1- and ARAF-mutated non-LCH using MEK and RAF inhibitors, respectively, resulted in clinical efficacy, demonstrating the importance of detecting and targeting diverse kinase alterations in these disorders. SIGNIFICANCE We provide the first description of kinase fusions in systemic histiocytic neoplasms and activating ARAF and MAP2K1 mutations in non-Langerhans histiocytic neoplasms. Refractory patients with MAP2K1- and ARAF-mutant histiocytoses had clinical responses to MEK inhibition and sorafenib, respectively, highlighting the importance of comprehensive genomic analysis of these disorders.


Cell Reports | 2014

Control of embryonic stem cell identity by BRD4-dependent transcriptional elongation of super-enhancer-associated pluripotency genes.

Raffaella Di Micco; Barbara Fontanals-Cirera; Vivien Low; Panagiotis Ntziachristos; Stephanie Yuen; Claudia D. Lovell; Igor Dolgalev; Yoshiya Yonekubo; Guangtao Zhang; Elena Rusinova; Guillermo Gerona-Navarro; Marta Cañamero; Michael Ohlmeyer; Iannis Aifantis; Ming-Ming Zhou; Aristotelis Tsirigos; Eva Hernando

SUMMARY Transcription factors and chromatin-remodeling complexes are key determinants of embryonic stem cell (ESC) identity. Here, we demonstrate that BRD4, a member of the bromodomain and extraterminal domain (BET) family of epigenetic readers, regulates the self-renewal ability and pluripotency of ESCs. BRD4 inhibition resulted in induction of epithelial-tomesenchymal transition (EMT) markers and commitment to the neuroectodermal lineage while reducing the ESC multidifferentiation capacity in teratoma as-says. BRD4 maintains transcription of core stem cell genes such as OCT4 and PRDM14 by occupying their super-enhancers (SEs), large clusters of regulatory elements, and recruiting to them Mediator and CDK9, the catalytic subunit of the positive transcription elongation factor b (P-TEFb), to allow Pol-II-dependent productive elongation. Our study describes a mechanism of regulation of ESC identity that could be applied to improve the efficiency of ESC differentiation.

Collaboration


Dive into the Igor Dolgalev's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Agnes Viale

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge