Network


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

Hotspot


Dive into the research topics where Karen Gascoigne is active.

Publication


Featured researches published by Karen Gascoigne.


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 | 2017

Therapeutic Targeting of the CBP/p300 Bromodomain Blocks the Growth of Castration-Resistant Prostate Cancer

Lingyan Jin; Jesse Garcia; Emily Chan; Cecile de la Cruz; Ehud Segal; Mark Merchant; Samir Kharbanda; Ryan Raisner; Peter M. Haverty; Zora Modrusan; Justin Ly; Edna F. Choo; Susan Kaufman; Maureen Beresini; F. Anthony Romero; Steven Magnuson; Karen Gascoigne

Resistance invariably develops to antiandrogen therapies used to treat newly diagnosed prostate cancers, but effective treatments for castration-resistant disease remain elusive. Here, we report that the transcriptional coactivator CBP/p300 is required to maintain the growth of castration-resistant prostate cancer. To exploit this vulnerability, we developed a novel small-molecule inhibitor of the CBP/p300 bromodomain that blocks prostate cancer growth in vitro and in vivo Molecular dissection of the consequences of drug treatment revealed a critical role for CBP/p300 in histone acetylation required for the transcriptional activity of the androgen receptor and its target gene expression. Our findings offer a preclinical proof of concept for small-molecule therapies to target the CBP/p300 bromodomain as a strategy to treat castration-resistant prostate cancer. Cancer Res; 77(20); 5564-75. ©2017 AACR.


Clinical Cancer Research | 2015

An Antibody–Drug Conjugate Directed against Lymphocyte Antigen 6 Complex, Locus E (LY6E) Provides Robust Tumor Killing in a Wide Range of Solid Tumor Malignancies

Jyoti Asundi; Lisa Crocker; Jarrod Tremayne; Peter Chang; Chie Sakanaka; Josh Tanguay; Susan D. Spencer; Sreedevi Chalasani; Elizabeth Luis; Karen Gascoigne; Rupal Desai; Rajiv Raja; Brad A. Friedman; Peter M. Haverty; Paul Polakis; Ron Firestein

Purpose: Chemotherapies are limited by a narrow therapeutic index resulting in suboptimal exposure of the tumor to the drug and acquired tumor resistance. One approach to overcome this is through antibody–drug conjugates (ADC) that facilitate greater potency via target-specific delivery of highly potent cytotoxic agents. Experimental Design: In this study, we used a bioinformatics approach to identify the lymphocyte antigen 6 complex locus E (LY6E), an IFN-inducible glycosylphosphatidylinositol (GPI)-linked cell membrane protein as a promising ADC target. We developed a monoclonal anti-LY6E antibody and characterized in situ LY6E expression in over 750 cancer specimens and normal tissues. Target-dependent anti-LY6E ADC killing was investigated both in vitro and in vivo using patient-derived xenograft models. Results: Using in silico approaches, we found that LY6E was significantly overexpressed and amplified in a wide array of different human solid tumors. IHC analysis revealed high LY6E protein expression in a number of tumor types, such as breast, lung, gastric, ovarian, pancreatic, kidney and head/neck carcinomas. Characterization of the endocytic pathways for LY6E revealed that the LY6E-specific antibody is internalized into cells leading to lysosomal accumulation. Consistent with this, a LY6E-specific ADC inhibited in vitro cell proliferation and produced durable tumor regression in vivo in clinically relevant LY6E-expressing xenograft models. Conclusions: Our results identify LY6E as a highly promising molecular ADC target for a variety of solid tumor types with current unmet medical need. Clin Cancer Res; 21(14); 3252–62. ©2015 AACR.


bioRxiv | 2018

Multiple-gene targeting and mismatch tolerance can confound analysis of genome-wide pooled CRISPR screens

Jean-Philippe Fortin; Karen Gascoigne; Peter M. Haverty; William F. Forrest; Michael Costa; Scott E. Martin

Genome-wide loss-of-function screens using the CRISPR/Cas9 system allow the efficient discovery of cancer cell vulnerabilities. While several studies have focused on correcting for DNA cleavage toxicity biases associated with copy number alterations, the effects of sgRNAs co-targeting multiple genomic loci in CRISPR screens have not been discussed yet. In this work, we analyze CRISPR essentiality screen data from 391 cancer cell lines to characterize biases induced by multi-target sgRNAs. We investigate two types of multi-targets: on-targets predicted through perfect sequence complementarity, and off-targets predicted through sequence complementarity with up to two nucleotide mismatches. We found that the number of on-targets and off-targets both increase sgRNA activity in a cell line-specific manner, and that existing additive models of gene knockout effects fail at capturing genetic interactions that may occur between co-targeted genes. We use synthetic lethality between paralog genes to show that genetic interactions can introduce biases in essentiality scores estimated from multi-target sgRNAs. We further show that single-mismatch tolerant sgRNAs can confound the analysis of gene essentiality and lead to incorrect co-essentiality functional networks.


Cancer Research | 2016

Abstract 4749: Bromodomain inhibition of the transcriptional coactivators CBP/EP300 as a therapeutic strategy to target the IRF4 network in multiple myeloma

Patricia J. Keller; Andrew R. Conery; Richard C. Centore; 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. 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. Citation Format: Patricia J. Keller, Andrew R. Conery, Richard C. Centore, Archana Bommi-Reddy, Karen E. Gascoigne, Barbara M. Bryant, Jennifer A. Mertz, Robert J. Sims. Bromodomain inhibition of the transcriptional coactivators CBP/EP300 as a therapeutic strategy to target the IRF4 network in multiple myeloma. [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 4749.


Journal of Medicinal Chemistry | 2016

Discovery of a Potent and Selective in Vivo Probe (GNE-272) for the Bromodomains of CBP/EP300

Terry D. Crawford; F. Anthony Romero; Kwong Wah Lai; Vickie Tsui; Alexander M. Taylor; Gladys de Leon Boenig; Cameron L. Noland; Jeremy Murray; Justin Ly; Edna F. Choo; Thomas Hunsaker; Emily Chan; Mark Merchant; Samir Kharbanda; Karen Gascoigne; Susan Kaufman; Maureen Beresini; Jiangpeng Liao; Wenfeng Liu; Kevin X. Chen; Zhongguo Chen; Andrew R. Conery; Alexandre Côté; Hariharan Jayaram; Ying Jiang; James R. Kiefer; Tracy Kleinheinz; Yingjie Li; Jonathan Maher; Eneida Pardo


Journal of Medicinal Chemistry | 2017

GNE-781, A Highly Advanced Potent and Selective Bromodomain Inhibitor of Cyclic Adenosine Monophosphate Response Element Binding Protein, Binding Protein (CBP)

F.A. Romero; Jeremy Murray; Kwong Wah Lai; Tsui; Brian K. Albrecht; L An; Maureen Beresini; G. de Leon Boenig; Sarah M. Bronner; Emily Chan; Kevin X. Chen; Zhongguo Chen; Edna F. Choo; K Clagg; K.R. Clark; Terry D. Crawford; Patrick Cyr; D de Almeida Nagata; Karen Gascoigne; J.L Grogan; G Hatzivassiliou; W Huang; Thomas Hunsaker; Susan Kaufman; S.G Koenig; R. Li; Yingjie Li; X. Liang; Jiangpeng Liao; Wenfeng Liu


Cell Reports | 2018

Enhancer Activity Requires CBP/P300 Bromodomain-Dependent Histone H3K27 Acetylation

Ryan Raisner; Samir Kharbanda; Lingyan Jin; Edwin Jeng; Emily Chan; Mark Merchant; Peter M. Haverty; Russell Bainer; Tommy K. Cheung; David Arnott; E. Megan Flynn; F. Anthony Romero; Steven Magnuson; Karen Gascoigne


Archive | 2015

Use of cbp/ep300 and bet inhibitors for treatment of cancer

Richard C. Centore; Andrew R. Conery; Karen Gascoigne; Iii Robert J. Sims


Cancer Research | 2018

Abstract 4329: Enhancer mapping in triple-negative breast cancer as a tool for novel biomarker and oncogene discovery

Ryan Raisner; Russ Bainer; Karen Gascoigne

Collaboration


Dive into the Karen Gascoigne's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark Merchant

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge