Rhamy Zeid
Harvard University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Rhamy Zeid.
Nature | 2012
Michael F. Berger; Eran Hodis; Timothy P. Heffernan; Yonathan Lissanu Deribe; Michael S. Lawrence; Alexei Protopopov; Elena S Ivanova; Ian R. Watson; Elizabeth Nickerson; Papia Ghosh; Hailei Zhang; Rhamy Zeid; Xiaojia Ren; Kristian Cibulskis; Andrey Sivachenko; Nikhil Wagle; Antje Sucker; Carrie Sougnez; Robert C. Onofrio; Lauren Ambrogio; Daniel Auclair; Timothy Fennell; Scott L. Carter; Yotam Drier; Petar Stojanov; Meredith A. Singer; Douglas Voet; Rui Jing; Gordon Saksena; Jordi Barretina
Melanoma is notable for its metastatic propensity, lethality in the advanced setting and association with ultraviolet exposure early in life. To obtain a comprehensive genomic view of melanoma in humans, we sequenced the genomes of 25 metastatic melanomas and matched germline DNA. A wide range of point mutation rates was observed: lowest in melanomas whose primaries arose on non-ultraviolet-exposed hairless skin of the extremities (3 and 14 per megabase (Mb) of genome), intermediate in those originating from hair-bearing skin of the trunk (5–55 per Mb), and highest in a patient with a documented history of chronic sun exposure (111 per Mb). Analysis of whole-genome sequence data identified PREX2 (phosphatidylinositol-3,4,5-trisphosphate-dependent Rac exchange factor 2)—a PTEN-interacting protein and negative regulator of PTEN in breast cancer—as a significantly mutated gene with a mutation frequency of approximately 14% in an independent extension cohort of 107 human melanomas. PREX2 mutations are biologically relevant, as ectopic expression of mutant PREX2 accelerated tumour formation of immortalized human melanocytes in vivo. Thus, whole-genome sequencing of human melanoma tumours revealed genomic evidence of ultraviolet pathogenesis and discovered a new recurrently mutated gene in melanoma.
Cancer Discovery | 2013
Alexandre Puissant; Stacey M. Frumm; Gabriela Alexe; Christopher F. Bassil; Jun Qi; Yvan Chanthery; Erin A. Nekritz; Rhamy Zeid; William Clay Gustafson; Patricia Greninger; Matthew J Garnett; Ultan McDermott; Cyril H. Benes; Andrew L. Kung; William A. Weiss; James E. Bradner; Kimberly Stegmaier
Bromodomain inhibition comprises a promising therapeutic strategy in cancer, particularly for hematologic malignancies. To date, however, genomic biomarkers to direct clinical translation have been lacking. We conducted a cell-based screen of genetically defined cancer cell lines using a prototypical inhibitor of BET bromodomains. Integration of genetic features with chemosensitivity data revealed a robust correlation between MYCN amplification and sensitivity to bromodomain inhibition. We characterized the mechanistic and translational significance of this finding in neuroblastoma, a childhood cancer with frequent amplification of MYCN. Genome-wide expression analysis showed downregulation of the MYCN transcriptional program accompanied by suppression of MYCN transcription. Functionally, bromodomain-mediated inhibition of MYCN impaired growth and induced apoptosis in neuroblastoma. BRD4 knockdown phenocopied these effects, establishing BET bromodomains as transcriptional regulators of MYCN. BET inhibition conferred a significant survival advantage in 3 in vivo neuroblastoma models, providing a compelling rationale for developing BET bromodomain inhibitors in patients with neuroblastoma.
Clinical Cancer Research | 2014
Pratiti Bandopadhayay; Guillaume Bergthold; Brian Nguyen; Simone Schubert; Sharareh Gholamin; Yujie Tang; Sara Bolin; Steven E. Schumacher; Rhamy Zeid; Sabran Masoud; Furong Yu; Nujsaubnusi Vue; William J. Gibson; Brenton R. Paolella; Siddhartha Mitra; Samuel H. Cheshier; Jun Qi; Kun-Wei Liu; Robert J. Wechsler-Reya; William A. Weiss; Fredrik J. Swartling; Mark W. Kieran; James E. Bradner; Rameen Beroukhim; Yoon-Jae Cho
Purpose: MYC-amplified medulloblastomas are highly lethal tumors. Bromodomain and extraterminal (BET) bromodomain inhibition has recently been shown to suppress MYC-associated transcriptional activity in other cancers. The compound JQ1 inhibits BET bromodomain-containing proteins, including BRD4. Here, we investigate BET bromodomain targeting for the treatment of MYC-amplified medulloblastoma. Experimental Design: We evaluated the effects of genetic and pharmacologic inhibition of BET bromodomains on proliferation, cell cycle, and apoptosis in established and newly generated patient- and genetically engineered mouse model (GEMM)-derived medulloblastoma cell lines and xenografts that harbored amplifications of MYC or MYCN. We also assessed the effect of JQ1 on MYC expression and global MYC-associated transcriptional activity. We assessed the in vivo efficacy of JQ1 in orthotopic xenografts established in immunocompromised mice. Results: Treatment of MYC-amplified medulloblastoma cells with JQ1 decreased cell viability associated with arrest at G1 and apoptosis. We observed downregulation of MYC expression and confirmed the inhibition of MYC-associated transcriptional targets. The exogenous expression of MYC from a retroviral promoter reduced the effect of JQ1 on cell viability, suggesting that attenuated levels of MYC contribute to the functional effects of JQ1. JQ1 significantly prolonged the survival of orthotopic xenograft models of MYC-amplified medulloblastoma (P < 0.001). Xenografts harvested from mice after five doses of JQ1 had reduced the expression of MYC mRNA and a reduced proliferative index. Conclusion: JQ1 suppresses MYC expression and MYC-associated transcriptional activity in medulloblastomas, resulting in an overall decrease in medulloblastoma cell viability. These preclinical findings highlight the promise of BET bromodomain inhibitors as novel agents for MYC-amplified medulloblastoma. Clin Cancer Res; 20(4); 912–25. ©2013 AACR.
Nature | 2016
Charles Y. Lin; Serap Erkek; Yiai Tong; Linlin Yin; Alexander J. Federation; Marc Zapatka; Parthiv Haldipur; Daisuke Kawauchi; Thomas Risch; Hans Jörg Warnatz; Barbara C. Worst; Bensheng Ju; Brent A. Orr; Rhamy Zeid; Donald R. Polaski; Maia Segura-Wang; Sebastian M. Waszak; David T. W. Jones; Marcel Kool; Volker Hovestadt; Ivo Buchhalter; Laura Sieber; Pascal Johann; Lukas Chavez; Stefan Gröschel; Marina Ryzhova; Andrey Korshunov; Wenbiao Chen; Victor V. Chizhikov; Kathleen J. Millen
Medulloblastoma is a highly malignant paediatric brain tumour, often inflicting devastating consequences on the developing child. Genomic studies have revealed four distinct molecular subgroups with divergent biology and clinical behaviour. An understanding of the regulatory circuitry governing the transcriptional landscapes of medulloblastoma subgroups, and how this relates to their respective developmental origins, is lacking. Here, using H3K27ac and BRD4 chromatin immunoprecipitation followed by sequencing (ChIP-seq) coupled with tissue-matched DNA methylation and transcriptome data, we describe the active cis-regulatory landscape across 28 primary medulloblastoma specimens. Analysis of differentially regulated enhancers and super-enhancers reinforced inter-subgroup heterogeneity and revealed novel, clinically relevant insights into medulloblastoma biology. Computational reconstruction of core regulatory circuitry identified a master set of transcription factors, validated by ChIP-seq, that is responsible for subgroup divergence, and implicates candidate cells of origin for Group 4. Our integrated analysis of enhancer elements in a large series of primary tumour samples reveals insights into cis-regulatory architecture, unrecognized dependencies, and cellular origins.
Nature Genetics | 2016
Pratiti Bandopadhayay; Lori A. Ramkissoon; Payal Jain; Guillaume Bergthold; Jeremiah Wala; Rhamy Zeid; Steven E. Schumacher; Laura M. Urbanski; Ryan O'Rourke; William J. Gibson; Kristine Pelton; Shakti Ramkissoon; Harry J. Han; Yuankun Zhu; Namrata Choudhari; Amanda Silva; Katie Boucher; Rosemary E. Henn; Yun Jee Kang; David Knoff; Brenton R. Paolella; Adrianne Gladden-Young; Pascale Varlet; Mélanie Pagès; Peleg Horowitz; Alexander J. Federation; Hayley Malkin; Adam Tracy; Sara Seepo; Matthew Ducar
Angiocentric gliomas are pediatric low-grade gliomas (PLGGs) without known recurrent genetic drivers. We performed genomic analysis of new and published data from 249 PLGGs, including 19 angiocentric gliomas. We identified MYB-QKI fusions as a specific and single candidate driver event in angiocentric gliomas. In vitro and in vivo functional studies show that MYB-QKI rearrangements promote tumorigenesis through three mechanisms: MYB activation by truncation, enhancer translocation driving aberrant MYB-QKI expression and hemizygous loss of the tumor suppressor QKI. To our knowledge, this represents the first example of a single driver rearrangement simultaneously transforming cells via three genetic and epigenetic mechanisms in a tumor.
Nature | 2017
Michael A. Erb; Thomas G. Scott; Bin E. Li; Huafeng Xie; Joshiawa Paulk; Hyuk-Soo Seo; Amanda Souza; Justin M. Roberts; Shiva Dastjerdi; Dennis L. Buckley; Neville E. Sanjana; Ophir Shalem; Behnam Nabet; Rhamy Zeid; Nana K. Offei-Addo; Sirano Dhe-Paganon; Feng Zhang; Stuart H. Orkin; Georg E. Winter; James E. Bradner
Recurrent chromosomal translocations producing a chimaeric MLL oncogene give rise to a highly aggressive acute leukaemia associated with poor clinical outcome. The preferential involvement of chromatin-associated factors as MLL fusion partners belies a dependency on transcription control. Despite recent progress made in targeting chromatin regulators in cancer, available therapies for this well-characterized disease remain inadequate, prompting the need to identify new targets for therapeutic intervention. Here, using unbiased CRISPR–Cas9 technology to perform a genome-scale loss-of-function screen in an MLL-AF4-positive acute leukaemia cell line, we identify ENL as an unrecognized gene that is specifically required for proliferation in vitro and in vivo. To explain the mechanistic role of ENL in leukaemia pathogenesis and dynamic transcription control, a chemical genetic strategy was developed to achieve targeted protein degradation. Acute loss of ENL suppressed the initiation and elongation of RNA polymerase II at active genes genome-wide, with pronounced effects at genes featuring a disproportionate ENL load. Notably, an intact YEATS chromatin-reader domain was essential for ENL-dependent leukaemic growth. Overall, these findings identify a dependency factor in acute leukaemia and suggest a mechanistic rationale for disrupting the YEATS domain in disease.
PLOS Pathogens | 2017
Jingwei Cheng; Donglim Esther Park; Christian Berrios; Elizabeth A. White; Reety Arora; Rosa Yoon; Timothy Branigan; Tengfei Xiao; Thomas Westerling; Alexander J. Federation; Rhamy Zeid; Benjamin Strober; Selene K. Swanson; Laurence Florens; James E. Bradner; Myles Brown; Peter M. Howley; Megha Padi; Michael P. Washburn; James A. DeCaprio
Merkel cell carcinoma (MCC) frequently contains integrated copies of Merkel cell polyomavirus DNA that express a truncated form of Large T antigen (LT) and an intact Small T antigen (ST). While LT binds RB and inactivates its tumor suppressor function, it is less clear how ST contributes to MCC tumorigenesis. Here we show that ST binds specifically to the MYC homolog MYCL (L-MYC) and recruits it to the 15-component EP400 histone acetyltransferase and chromatin remodeling complex. We performed a large-scale immunoprecipitation for ST and identified co-precipitating proteins by mass spectrometry. In addition to protein phosphatase 2A (PP2A) subunits, we identified MYCL and its heterodimeric partner MAX plus the EP400 complex. Immunoprecipitation for MAX and EP400 complex components confirmed their association with ST. We determined that the ST-MYCL-EP400 complex binds together to specific gene promoters and activates their expression by integrating chromatin immunoprecipitation with sequencing (ChIP-seq) and RNA-seq. MYCL and EP400 were required for maintenance of cell viability and cooperated with ST to promote gene expression in MCC cell lines. A genome-wide CRISPR-Cas9 screen confirmed the requirement for MYCL and EP400 in MCPyV-positive MCC cell lines. We demonstrate that ST can activate gene expression in a EP400 and MYCL dependent manner and this activity contributes to cellular transformation and generation of induced pluripotent stem cells.
Cancer Research | 2013
Alexandre Puissant; Stacey M. Frumm; Gabriela Alexe; Christopher F. Bassil; Jun Qi; Yvan H. Chanthery; Erin A. Nekritz; Rhamy Zeid; W. Clay Gustafson; Patricia Greninger; Matthew J. Garnett; Ultan McDermott; Cyril H. Benes; Andrew L. Kung; William A. Weiss; James E. Bradner; Kimberly Stegmaier
Bromodomain inhibition comprises a promising therapeutic strategy in cancer, particularly for hematologic malignancies. To date, however, genomic biomarkers to direct clinical translation have been lacking. We conducted a cell-based screen of genetically-defined cancer cell lines using a prototypical inhibitor of BET (bromodomain and extra-terminal domain) bromodomains. Integration of genetic features with chemosensitivity data revealed a robust correlation between MYCN amplification and sensitivity to bromodomain inhibition. We characterized the mechanistic and translational significance of this finding in neuroblastoma, a childhood cancer with frequent amplification of MYCN. Genome-wide expression analysis demonstrated downregulation of the MYCN transcriptional program accompanied by suppression of MYCN transcription, and a BET Bromodomain inhibitor was found to displace BRD4 from the MYCN promoter region in neuroblastoma cell lines. Functionally, bromodomain-mediated inhibition of MYCN impaired growth and induced apoptosis in neuroblastoma. BRD4 knock-down phenocopied these effects, establishing BET bromodomains as transcriptional regulators of MYCN. BET inhibition conferred a significant survival advantage in three in vivo neuroblastoma models, providing a compelling rationale for developing BET bromodomain inhibitors in patients with neuroblastoma. Citation Format: Alexandre Puissant, Stacey M. Frumm, Gabriela Alexe, Christopher F. Bassil, Jun Qi, Yvan H. Chanthery, Erin A. Nekritz, Rhamy Zeid, W. Clay Gustafson, Patricia Greninger, Matthew J. Garnett, Ultan McDermott, Cyril H. Benes, Andrew L. Kung, William A. Weiss, James E. Bradner, Kimberly Stegmaier. Targeting MYCN in Neuroblastoma by BET Bromodomain Inhibition. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4622. doi:10.1158/1538-7445.AM2013-4622
Proceedings of the National Academy of Sciences of the United States of America | 2018
Matthew L. Hemming; Matthew A. Lawlor; Rhamy Zeid; Tom Lesluyes; Jonathan A. Fletcher; Chandrajit P. Raut; Ewa Sicinska; Frédéric Chibon; Scott A. Armstrong; George D. Demetri; James E. Bradner
Significance Enhancers are regulatory regions in DNA that govern gene expression and orchestrate cellular phenotype. We describe the enhancer landscape of gastrointestinal stromal tumor (GIST), identifying established and unique GIST-associated genes that characterize this neoplasm. Focusing on transcriptional regulators, we identify a core group of transcription factors underlying GIST biology. Two transcription factors, BARX1 and HAND1, have mutually exclusive enhancers and expression in localized and metastatic GIST, respectively. HAND1 is necessary to sustain GIST proliferation and KIT expression, and binds to enhancers of GIST-associated genes. The relative expression of BARX1 and HAND1 is predictive of clinical behavior in GIST patients. These results expand our understanding of gene regulation in this disease and identify biomarkers that may be helpful in diagnosis and treatment. Activating mutations in the KIT or PDGFRA receptor tyrosine kinases are hallmarks of gastrointestinal stromal tumor (GIST). The biological underpinnings of recurrence following resection or disease progression beyond kinase mutation are poorly understood. Utilizing chromatin immunoprecipitation with sequencing of tumor samples and cell lines, we describe the enhancer landscape of GIST, highlighting genes that reinforce and extend our understanding of these neoplasms. A group of core transcription factors can be distinguished from others unique to localized and metastatic disease. The transcription factor HAND1 emerges in metastatic disease, binds to established GIST-associated enhancers, and facilitates GIST cell proliferation and KIT gene expression. The pattern of transcription factor expression in primary tumors is predictive of metastasis-free survival in GIST patients. These results provide insight into the enhancer landscape and transcription factor network underlying GIST, and define a unique strategy for predicting clinical behavior of this disease.
Molecular Cancer Therapeutics | 2015
Charles Y. Lin; Serap Erkek; Yiai Tong; Linlin Yang; Alexander J. Federation; Marc Zapatka; Parthiv Haldipur; Daisuke Kawauchi; Thomas Risch; Hans-Jörg Warnatz; Barbara C. Worst; Bensheng Ju; Brent A. Orr; Rhamy Zeid; Donald R. Polaski; Maia Segura-Wang; Sebastian M. Waszak; David T. W. Jones; Marcel Kool; Volker Hovestadt; Ivo Buchhalter; Laura Sieber; Pascal Johann; Stefan Gröschel; Marina Ryzhova; Andrey Korshunov; Wenbiao Chen; Victor V. Chizhikov; Kathleen J. Millen; Vyacheslav Amstislavskiy
Medulloblastoma is a highly malignant paediatric brain tumour, often inflicting devastating consequences on the developing child. Genomic studies have revealed four distinct molecular subgroups with divergent biology and clinical behaviour. An understanding of the regulatory circuitry governing the transcriptional landscapes of medulloblastoma subgroups, and how this relates to their respective developmental origins, is currently lacking. Using H3K27ac and BRD4 ChIP-Seq, coupled with tissue-matched DNA methylation and transcriptome data, we describe the active cis-regulatory landscape across 28 primary medulloblastoma specimens. Analysis of differentially regulated enhancers and super-enhancers reinforced inter-subgroup heterogeneity and revealed novel, clinically relevant insights into medulloblastoma biology. Computational reconstruction of core regulatory circuitry identified a master set of transcription factors responsible for subgroup divergence that validated by ChIP-Seq and implicated candidate cells-of-origin for Group 4. Our integrated analysis of cis-regulatory elements in a large series of primary tumour samples reveals insights into cis-regulatory architecture, unrecognized dependencies, and cellular origins. Citation Format: Charles Y. Lin, Serap Erkek, Yiai Tong, Linlin Yang, Alexander J. Federation, Marc Zapatka, Parthiv Haldipur, Daisuke Kawauchi, Thomas Risch, Hans-Jorg Warnatz, Barbara Worst, Bensheng Ju, Brent A. Orr, Rhamy Zeid, Donald R. Polaski, Maia Segura-Wang, Sebastian M. Waszak, David TW Jones, Marcel Kool, Volker Hovestadt, Ivo Buchhalter, Laura Sieber, Pascal Johann, Stefan Groschel, Marina Ryzhova, Andrey Korshunov, Wenbiao Chen, Victor V. Chizhikov, Kathleen J. Millen, Vyacheslav Amstislavskiy, Hans Lehrach, Marie-Laure Yaspo, Roland Eils, Peter Lichter, Jan O. Korbel, Stefan Pfister, James E. Bradner, Paul A. Northcott. Medulloblastoma regulatory circuitries reveal subgroup-specific cellular origins. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr LB-B23.