Network


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

Hotspot


Dive into the research topics where Charlie Hatton is active.

Publication


Featured researches published by Charlie Hatton.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Assessing the significance of chromosomal aberrations in cancer: Methodology and application to glioma

Rameen Beroukhim; Gad Getz; Leia Nghiemphu; Jordi Barretina; Teli Hsueh; David Linhart; Igor Vivanco; Jeffrey C. Lee; Julie H. Huang; Sethu Alexander; Jinyan Du; Tweeny R. Kau; Roman K. Thomas; Kinjal Shah; Horacio Soto; Sven Perner; John R. Prensner; Ralph DeBiasi; Francesca Demichelis; Charlie Hatton; Mark A. Rubin; Levi A. Garraway; Stan F. Nelson; Linda M. Liau; Paul S. Mischel; T. Cloughesy; Matthew Meyerson; Todd Golub; Eric S. Lander; Ingo K. Mellinghoff

Comprehensive knowledge of the genomic alterations that underlie cancer is a critical foundation for diagnostics, prognostics, and targeted therapeutics. Systematic efforts to analyze cancer genomes are underway, but the analysis is hampered by the lack of a statistical framework to distinguish meaningful events from random background aberrations. Here we describe a systematic method, called Genomic Identification of Significant Targets in Cancer (GISTIC), designed for analyzing chromosomal aberrations in cancer. We use it to study chromosomal aberrations in 141 gliomas and compare the results with two prior studies. Traditional methods highlight hundreds of altered regions with little concordance between studies. The new approach reveals a highly concordant picture involving ≈35 significant events, including 16–18 broad events near chromosome-arm size and 16–21 focal events. Approximately half of these events correspond to known cancer-related genes, only some of which have been previously tied to glioma. We also show that superimposed broad and focal events may have different biological consequences. Specifically, gliomas with broad amplification of chromosome 7 have properties different from those with overlapping focalEGFR amplification: the broad events act in part through effects on MET and its ligand HGF and correlate with MET dependence in vitro. Our results support the feasibility and utility of systematic characterization of the cancer genome.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Drug-sensitive FGFR2 mutations in endometrial carcinoma

Amit Dutt; Helga B. Salvesen; Tzu-Hsiu Chen; Alex H. Ramos; Robert C. Onofrio; Charlie Hatton; Richard Nicoletti; Wendy Winckler; Rupinder Grewal; Megan Hanna; Nicolas Wyhs; Liuda Ziaugra; Daniel J. Richter; Jone Trovik; Ingeborg B. Engelsen; Ingunn Stefansson; Timothy Fennell; Kristian Cibulskis; Michael C. Zody; Lars A. Akslen; Stacey Gabriel; Kwok-Kin Wong; William R. Sellers; Matthew Meyerson; Heidi Greulich

Oncogenic activation of tyrosine kinases is a common mechanism of carcinogenesis and, given the druggable nature of these enzymes, an attractive target for anticancer therapy. Here, we show that somatic mutations of the fibroblast growth factor receptor 2 (FGFR2) tyrosine kinase gene, FGFR2, are present in 12% of endometrial carcinomas, with additional instances found in lung squamous cell carcinoma and cervical carcinoma. These FGFR2 mutations, many of which are identical to mutations associated with congenital craniofacial developmental disorders, are constitutively activated and oncogenic when ectopically expressed in NIH 3T3 cells. Inhibition of FGFR2 kinase activity in endometrial carcinoma cell lines bearing such FGFR2 mutations inhibits transformation and survival, implicating FGFR2 as a novel therapeutic target in endometrial carcinoma.


PLOS Genetics | 2012

Loss of ATRX, Genome Instability, and an Altered DNA Damage Response Are Hallmarks of the Alternative Lengthening of Telomeres Pathway

Courtney A. Lovejoy; Wendi Li; Steven Reisenweber; Supawat Thongthip; Joanne Bruno; Titia de Lange; Saurav De; John H.J. Petrini; Patricia Sung; Maria Jasin; Joseph Rosenbluh; Yaara Zwang; Barbara A. Weir; Charlie Hatton; Elena Ivanova; Laura E. MacConaill; Megan Hanna; William C. Hahn; Neal F. Lue; Roger R. Reddel; Yuchen Jiao; Kenneth W. Kinzler; Bert Vogelstein; Nickolas Papadopoulos; Alan K. Meeker

The Alternative Lengthening of Telomeres (ALT) pathway is a telomerase-independent pathway for telomere maintenance that is active in a significant subset of human cancers and in vitro immortalized cell lines. ALT is thought to involve templated extension of telomeres through homologous recombination, but the genetic or epigenetic changes that unleash ALT are not known. Recently, mutations in the ATRX/DAXX chromatin remodeling complex and histone H3.3 were found to correlate with features of ALT in pancreatic neuroendocrine cancers, pediatric glioblastomas, and other tumors of the central nervous system, suggesting that these mutations might contribute to the activation of the ALT pathway in these cancers. We have taken a comprehensive approach to deciphering ALT by applying genomic, molecular biological, and cell biological approaches to a panel of 22 ALT cell lines, including cell lines derived in vitro. Here we show that loss of ATRX protein and mutations in the ATRX gene are hallmarks of ALT–immortalized cell lines. In addition, ALT is associated with extensive genome rearrangements, marked micronucleation, defects in the G2/M checkpoint, and altered double-strand break (DSB) repair. These attributes will facilitate the diagnosis and treatment of ALT positive human cancers.


Chemistry & Biology | 2014

EZH2 inhibitor efficacy in non-Hodgkin's lymphoma does not require suppression of H3K27 monomethylation.

William D. Bradley; Shilpi Arora; Jennifer Busby; Srividya Balasubramanian; Victor S. Gehling; Christopher G. Nasveschuk; Rishi G. Vaswani; Chih-Chi Yuan; Charlie Hatton; Feng Zhao; Kaylyn E. Williamson; Priyadarshini Iyer; Jacqui Mendez; Robert E. Campbell; Nico Cantone; Shivani Garapaty-Rao; James E. Audia; Andrew Simon Cook; Les A. Dakin; Brian K. Albrecht; Jean-Christophe Harmange; Danette L. Daniels; Richard T. Cummings; Barbara M. Bryant; Emmanuel Normant; Patrick Trojer

The histone lysine methyltransferase (MT) Enhancer of Zeste Homolog 2 (EZH2) is considered an oncogenic driver in a subset of germinal center B-cell-like diffuse large B cell lymphoma (GCB-DLBCL) and follicular lymphoma due to the presence of recurrent, monoallelic mutations in the EZH2 catalytic domain. These genomic data suggest that targeting the EZH2 MT activity is a valid therapeutic strategy for the treatment of lymphoma patients with EZH2 mutations. Here we report the identification of highly potent and selective EZH2 small molecule inhibitors, their validation by a cellular thermal shift assay, application across a large cell panel representing various non-Hodgkins lymphoma (NHL) subtypes, and their efficacy in EZH2mutant-containing GCB-DLBCL xenograft models. Surprisingly, our EZH2 inhibitors selectively affect the turnover of trimethylated, but not monomethylated histone H3 lysine 27 at pharmacologically relevant doses. Importantly, we find that these inhibitors are broadly efficacious also in NHL models with wild-type EZH2.


Blood | 2011

High-throughput mutation profiling of CTCL samples reveals KRAS and NRAS mutations sensitizing tumors toward inhibition of the RAS/RAF/MEK signaling cascade.

Michael K. Kiessling; Patrick A. Oberholzer; Chandrani Mondal; Maria B. Karpova; Marie C. Zipser; William M. Lin; Michael Girardi; Laura E. MacConaill; Sarah M. Kehoe; Charlie Hatton; Lars E. French; Levi A. Garraway; Gernot Polier; Dorothee Süss; Claus-Detlev Klemke; Peter H. Krammer; Karsten Gülow; Reinhard Dummer

Cutaneous T-cell lymphomas (CTCLs) are malignancies of skin-homing lymphoid cells, which have so far not been investigated thoroughly for common oncogenic mutations. We screened 90 biopsy specimens from CTCL patients (41 mycosis fungoides, 36 Sézary syndrome, and 13 non-mycosis fungoides/Sézary syndrome CTCL) for somatic mutations using OncoMap technology. We detected oncogenic mutations for the RAS pathway in 4 of 90 samples. One mycosis fungoides and one pleomorphic CTCL harbored a KRAS(G13D) mutation; one Sézary syndrome and one CD30(+) CTCL harbored a NRAS(Q61K) amino acid change. All mutations were found in stage IV patients (4 of 42) who showed significantly decreased overall survival compared with stage IV patients without mutations (P = .04). In addition, we detected a NRAS(Q61K) mutation in the CTCL cell line Hut78. Knockdown of NRAS by siRNA induced apoptosis in mutant Hut78 cells but not in CTCL cell lines lacking RAS mutations. The NRAS(Q61K) mutation sensitized Hut78 cells toward growth inhibition by the MEK inhibitors U0126, AZD6244, and PD0325901. Furthermore, we found that MEK inhibitors exclusively induce apoptosis in Hut78 cells. Taken together, we conclude that RAS mutations are rare events at a late stage of CTCL, and our preclinical results suggest that such late-stage patients profit from MEK inhibitors.


The Journal of Molecular Diagnostics | 2014

Prospective Enterprise-Level Molecular Genotyping of a Cohort of Cancer Patients

Laura E. MacConaill; Elizabeth Garcia; Priyanka Shivdasani; Matthew Ducar; Ravali Adusumilli; Marc Breneiser; Mark W. Byrne; Lawrence Chung; Jodie Conneely; Lauren Crosby; Levi A. Garraway; Xin Gong; William C. Hahn; Charlie Hatton; Philip W. Kantoff; Michael J. Kluk; Frank C. Kuo; Yonghui Jia; Ruchi Joshi; Janina A. Longtine; Allison Manning; Emanuele Palescandolo; Nematullah Sharaf; Lynette M. Sholl; Paul Van Hummelen; Jacqueline E. Wade; Bruce M. Wollinson; Dimity Zepf; Barrett J. Rollins; Neal I. Lindeman

Ongoing cancer genome characterization studies continue to elucidate the spectrum of genomic abnormalities that drive many cancers, and in the clinical arena assessment of the driver genetic alterations in patients is playing an increasingly important diagnostic and/or prognostic role for many cancer types. However, the landscape of genomic abnormalities is still unknown for less common cancers, and the influence of specific genotypes on clinical behavior is often still unclear. To address some of these deficiencies, we developed Profile, a prospective cohort study to obtain genomic information on all patients at a large tertiary care medical center for cancer-related care. We enrolled patients with any cancer diagnosis, and, for each patient (unselected for cancer site or type) we applied mass spectrometric genotyping (OncoMap) of 471 common recurrent mutations in 41 cancer-related genes. We report the results of the first 5000 patients, of which 26% exhibited potentially actionable somatic mutations. These observations indicate the utility of genotyping in advancing the field of precision oncology.


eLife | 2016

Bromodomain inhibition of the transcriptional coactivators CBP/EP300 as a therapeutic strategy to target the IRF4 network in multiple myeloma

Andrew R. Conery; Richard C. Centore; Adrianne Neiss; Patricia J. Keller; Shivangi Joshi; Kerry L. Spillane; Peter Sandy; Charlie Hatton; Eneida Pardo; Laura Zawadzke; Archana Bommi-Reddy; Karen Gascoigne; Barbara M. Bryant; Jennifer A. Mertz; Robert J. Sims

Pharmacological inhibition of chromatin co-regulatory factors represents a clinically validated strategy to modulate oncogenic signaling through selective attenuation of gene expression. Here, we demonstrate that CBP/EP300 bromodomain inhibition preferentially abrogates the viability of multiple myeloma cell lines. Selective targeting of multiple myeloma cell lines through CBP/EP300 bromodomain inhibition is the result of direct transcriptional suppression of the lymphocyte-specific transcription factor IRF4, which is essential for the viability of myeloma cells, and the concomitant repression of the IRF4 target gene c-MYC. Ectopic expression of either IRF4 or MYC antagonizes the phenotypic and transcriptional effects of CBP/EP300 bromodomain inhibition, highlighting the IRF4/MYC axis as a key component of its mechanism of action. These findings suggest that CBP/EP300 bromodomain inhibition represents a viable therapeutic strategy for targeting multiple myeloma and other lymphoid malignancies dependent on the IRF4 network. DOI: http://dx.doi.org/10.7554/eLife.10483.001


Cancer Research | 2016

Pharmacological Inhibition of the Histone Lysine Demethylase KDM1A Suppresses the Growth of Multiple Acute Myeloid Leukemia Subtypes.

John P. McGrath; Kaylyn Williamson; Srividya Balasubramanian; Shobu Odate; Shilpi Arora; Charlie Hatton; Thomas M. Edwards; Thomas O'Brien; Steven Magnuson; David Stokoe; Danette L. Daniels; Barbara M. Bryant; Patrick Trojer

Lysine-specific demethylase 1 (KDM1A) is a transcriptional coregulator that can function in both the activation and repression of gene expression, depending upon context. KDM1A plays an important role in hematopoiesis and was identified as a dependency factor in leukemia stem cell populations. Therefore, we investigated the consequences of inhibiting KDM1A in a panel of cell lines representing all acute myelogenous leukemia (AML) subtypes using selective, reversible and irreversible KDM1A small-molecule inhibitors. Cell models of AML, CML, and T-ALL were potently affected by KDM1A inhibition, and cells bearing RUNX1-RUNX1T1 (AML1-ETO) translocations were especially among the most sensitive. RNAi-mediated silencing of KDM1A also effectively suppressed growth of RUNX1-RUNX1T1-containing cell lines. Furthermore, pharmacologic inhibition of KDM1A resulted in complete abrogation of tumor growth in an AML xenograft model harboring RUNX1-RUNX1T1 translocations. We unexpectedly found that KDM1A-targeting compounds not only inhibited the catalytic activity of the enzyme, but evicted KDM1A from target genes. Accordingly, compound-mediated KDM1A eviction was associated with elevated levels of local histone H3 lysine 4 dimethylation, and increased target gene expression, which was further accompanied by cellular differentiation and induction of cell death. Finally, our finding that KDM1A inhibitors effectively synergize with multiple conventional as well as candidate anti-AML agents affords a framework for potential future clinical application. Cancer Res; 76(7); 1975-88. ©2016 AACR.


Journal of Biological Chemistry | 2016

Regulatory T Cell Modulation by CBP/EP300 Bromodomain Inhibition

Srimoyee Ghosh; Alexander I. Taylor; Melissa Chin; Hon-Ren Huang; Andrew R. Conery; Jennifer A. Mertz; Andres Salmeron; Pranal J. Dakle; Deanna A. Mele; Alexandre Côté; Hari Jayaram; Jeremy W. Setser; Florence Poy; Georgia Hatzivassiliou; Denise DeAlmeida-Nagata; Peter Sandy; Charlie Hatton; F. Anthony Romero; Eugene Chiang; Thornik Reimer; Terry D. Crawford; Eneida Pardo; Venita G. Watson; Vickie Tsui; Andrea G. Cochran; Laura Zawadzke; Jean-Christophe Harmange; James E. Audia; Barbara M. Bryant; Richard T. Cummings

Covalent modification of histones is a fundamental mechanism of regulated gene expression in eukaryotes, and interpretation of histone modifications is an essential feature of epigenetic control. Bromodomains are specialized binding modules that interact with acetylated histones, linking chromatin recognition to gene transcription. Because of their ability to function in a domain-specific fashion, selective disruption of bromodomain:acetylated histone interactions with chemical probes serves as a powerful means for understanding biological processes regulated by these chromatin adaptors. Here we describe the discovery and characterization of potent and selective small molecule inhibitors for the bromodomains of CREBBP/EP300 that engage their target in cellular assays. We use these tools to demonstrate a critical role for CREBBP/EP300 bromodomains in regulatory T cell biology. Because regulatory T cell recruitment to tumors is a major mechanism of immune evasion by cancer cells, our data highlight the importance of CREBBP/EP300 bromodomain inhibition as a novel, small molecule-based approach for cancer immunotherapy.


Cancer Research | 2016

Preclinical Anticancer Efficacy of BET Bromodomain Inhibitors Is Determined by the Apoptotic Response

Andrew R. Conery; Richard C. Centore; Kerry L. Spillane; Nicole E. Follmer; Archana Bommi-Reddy; Charlie Hatton; Barbara M. Bryant; Patricia Greninger; Arnaud Amzallag; Cyril H. Benes; Jennifer A. Mertz; Robert J. Sims

Small-molecule inhibitors of the bromodomain and extraterminal (BET) family of proteins are being tested in clinical trials for a variety of cancers, but patient selection strategies remain limited. This challenge is partly attributed to the heterogeneous responses elicited by BET inhibition (BETi), including cellular differentiation, senescence, and death. In this study, we performed phenotypic and gene-expression analyses of treatment-naive and engineered tolerant cell lines representing human melanoma and leukemia to elucidate the dominant features defining response to BETi. We found that de novo and acquired tolerance to BETi is driven by the robustness of the apoptotic response, and that genetic or pharmacologic manipulation of the apoptotic signaling network can modify the phenotypic response to BETi. We further reveal that the expression signatures of the apoptotic genes BCL2, BCL2L1, and BAD significantly predict response to BETi. Taken together, our findings highlight the apoptotic program as a determinant of response to BETi, and provide a molecular basis for patient stratification and combination therapy development.

Collaboration


Dive into the Charlie Hatton's collaboration.

Top Co-Authors

Avatar

Barbara M. Bryant

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert J. Sims

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Patrick Trojer

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge