Network


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

Hotspot


Dive into the research topics where Brinton Seashore-Ludlow is active.

Publication


Featured researches published by Brinton Seashore-Ludlow.


Cancer Discovery | 2015

Harnessing Connectivity in a Large-Scale Small-Molecule Sensitivity Dataset

Brinton Seashore-Ludlow; Matthew G. Rees; Jaime H. Cheah; Murat Cokol; Edmund V. Price; Matthew E. Coletti; Victor Victor Jones; Nicole E. Bodycombe; Christian K. Soule; Joshua Gould; Benjamin Alexander; Ava Li; Philip Montgomery; Mathias J. Wawer; Nurdan Kuru; Joanne Kotz; C. Suk-Yee Hon; Benito Munoz; Ted Liefeld; Vlado Dančík; Joshua Bittker; Michelle Palmer; James E. Bradner; Alykhan F. Shamji; Paul A. Clemons; Stuart L. Schreiber

UNLABELLED Identifying genetic alterations that prime a cancer cell to respond to a particular therapeutic agent can facilitate the development of precision cancer medicines. Cancer cell-line (CCL) profiling of small-molecule sensitivity has emerged as an unbiased method to assess the relationships between genetic or cellular features of CCLs and small-molecule response. Here, we developed annotated cluster multidimensional enrichment analysis to explore the associations between groups of small molecules and groups of CCLs in a new, quantitative sensitivity dataset. This analysis reveals insights into small-molecule mechanisms of action, and genomic features that associate with CCL response to small-molecule treatment. We are able to recapitulate known relationships between FDA-approved therapies and cancer dependencies and to uncover new relationships, including for KRAS-mutant cancers and neuroblastoma. To enable the cancer community to explore these data, and to generate novel hypotheses, we created an updated version of the Cancer Therapeutic Response Portal (CTRP v2). SIGNIFICANCE We present the largest CCL sensitivity dataset yet available, and an analysis method integrating information from multiple CCLs and multiple small molecules to identify CCL response predictors robustly. We updated the CTRP to enable the cancer research community to leverage these data and analyses.


Cell | 2014

Predicting cancer-specific vulnerability via data-driven detection of synthetic lethality.

Livnat Jerby-Arnon; Nadja Pfetzer; Yedael Y. Waldman; Lynn McGarry; Daniel James; Emma Shanks; Brinton Seashore-Ludlow; Adam Weinstock; Tamar Geiger; Paul A. Clemons; Eyal Gottlieb; Eytan Ruppin

Synthetic lethality occurs when the inhibition of two genes is lethal while the inhibition of each single gene is not. It can be harnessed to selectively treat cancer by identifying inactive genes in a given cancer and targeting their synthetic lethal (SL) partners. We present a data-driven computational pipeline for the genome-wide identification of SL interactions in cancer by analyzing large volumes of cancer genomic data. First, we show that the approach successfully captures known SL partners of tumor suppressors and oncogenes. We then validate SL predictions obtained for the tumor suppressor VHL. Next, we construct a genome-wide network of SL interactions in cancer and demonstrate its value in predicting gene essentiality and clinical prognosis. Finally, we identify synthetic lethality arising from gene overactivation and use it to predict drug efficacy. These results form a computational basis for exploiting synthetic lethality to uncover cancer-specific susceptibilities.


Nature Chemical Biology | 2016

Correlating chemical sensitivity and basal gene expression reveals mechanism of action

Matthew G. Rees; Brinton Seashore-Ludlow; Jaime H. Cheah; Drew J. Adams; Edmund Price; Shubhroz Gill; Sarah Javaid; Matthew E. Coletti; Victor Victor Jones; Nicole E Bodycombe; Christian K. Soule; Benjamin Alexander; Ava Li; Philip Montgomery; Joanne Kotz; C. Suk-Yee Hon; Benito Munoz; Ted Liefeld; Vlado Dančík; Daniel A. Haber; Clary B. Clish; Joshua Bittker; Michelle Palmer; Bridget K. Wagner; Paul A. Clemons; Alykhan F. Shamji; Stuart L. Schreiber

Changes in cellular gene expression in response to small-molecule or genetic perturbations have yielded signatures that can connect unknown mechanisms of action (MoA) to ones previously established. We hypothesized that differential basal gene expression could be correlated with patterns of small-molecule sensitivity across many cell lines to illuminate the actions of compounds whose MoA are unknown. To test this idea, we correlated the sensitivity patterns of 481 compounds with ~19,000 basal transcript levels across 823 different human cancer cell lines and identified selective outlier transcripts. This process yielded many novel mechanistic insights, including the identification of activation mechanisms, cellular transporters, and direct protein targets. We found that ML239, originally identified in a phenotypic screen for selective cytotoxicity in breast cancer stem-like cells, most likely acts through activation of fatty acid desaturase 2 (FADS2). These data and analytical tools are available to the research community through the Cancer Therapeutics Response Portal.


Nature | 2017

Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway

Vasanthi Viswanathan; Matthew J. Ryan; Harshil Dhruv; Shubhroz Gill; Ossia M. Eichhoff; Brinton Seashore-Ludlow; Samuel D. Kaffenberger; John K. Eaton; Kenichi Shimada; Andrew J. Aguirre; Srinivas R. Viswanathan; Shrikanta Chattopadhyay; Pablo Tamayo; Wan Seok Yang; Matthew G. Rees; Sixun Chen; Zarko V. Boskovic; Sarah Javaid; Cherrie Huang; Xiaoyun Wu; Yuen Yi Tseng; Elisabeth Roider; Dong Gao; James M. Cleary; Brian M. Wolpin; Jill P. Mesirov; Daniel A. Haber; Jeffrey A. Engelman; Jesse S. Boehm; Joanne Kotz

Plasticity of the cell state has been proposed to drive resistance to multiple classes of cancer therapies, thereby limiting their effectiveness. A high-mesenchymal cell state observed in human tumours and cancer cell lines has been associated with resistance to multiple treatment modalities across diverse cancer lineages, but the mechanistic underpinning for this state has remained incompletely understood. Here we molecularly characterize this therapy-resistant high-mesenchymal cell state in human cancer cell lines and organoids and show that it depends on a druggable lipid-peroxidase pathway that protects against ferroptosis, a non-apoptotic form of cell death induced by the build-up of toxic lipid peroxides. We show that this cell state is characterized by activity of enzymes that promote the synthesis of polyunsaturated lipids. These lipids are the substrates for lipid peroxidation by lipoxygenase enzymes. This lipid metabolism creates a dependency on pathways converging on the phospholipid glutathione peroxidase (GPX4), a selenocysteine-containing enzyme that dissipates lipid peroxides and thereby prevents the iron-mediated reactions of peroxides that induce ferroptotic cell death. Dependency on GPX4 was found to exist across diverse therapy-resistant states characterized by high expression of ZEB1, including epithelial–mesenchymal transition in epithelial-derived carcinomas, TGFβ-mediated therapy-resistance in melanoma, treatment-induced neuroendocrine transdifferentiation in prostate cancer, and sarcomas, which are fixed in a mesenchymal state owing to their cells of origin. We identify vulnerability to ferroptic cell death induced by inhibition of a lipid peroxidase pathway as a feature of therapy-resistant cancer cells across diverse mesenchymal cell-state contexts.


ACS Chemical Biology | 2014

NAMPT Is the Cellular Target of STF-31-Like Small-Molecule Probes

Drew J. Adams; Daisuke Ito; Matthew G. Rees; Brinton Seashore-Ludlow; Xiaoling Puyang; Alex H. Ramos; Jaime H. Cheah; Paul A. Clemons; Markus Warmuth; Ping Zhu; Alykhan F. Shamji; Stuart L. Schreiber

The small-molecule probes STF-31 and its analogue compound 146 were discovered while searching for compounds that kill VHL-deficient renal cell carcinoma cell lines selectively and have been reported to act via direct inhibition of the glucose transporter GLUT1. We profiled the sensitivity of 679 cancer cell lines to STF-31 and found that the pattern of response is tightly correlated with sensitivity to three different inhibitors of nicotinamide phosphoribosyltransferase (NAMPT). We also performed whole-exome next-generation sequencing of compound 146-resistant HCT116 clones and identified a recurrent NAMPT-H191R mutation. Ectopic expression of NAMPT-H191R conferred resistance to both STF-31 and compound 146 in cell lines. We further demonstrated that both STF-31 and compound 146 inhibit the enzymatic activity of NAMPT in a biochemical assay in vitro. Together, our cancer-cell profiling and genomic approaches identify NAMPT inhibition as a critical mechanism by which STF-31-like compounds inhibit cancer cells.


Clinical Cancer Research | 2016

DiSCoVERing Innovative Therapies for Rare Tumors: Combining Genetically Accurate Disease Models with In Silico Analysis to Identify Novel Therapeutic Targets.

Allison Hanaford; Tenley C. Archer; Antoinette Price; Ulf D. Kahlert; Jarek Maciaczyk; Guido Nikkhah; Jong Wook Kim; Tobias Ehrenberger; Paul A. Clemons; Vlado Dančík; Brinton Seashore-Ludlow; Vasanthi Viswanathan; Michelle L. Stewart; Matthew G. Rees; Alykhan F. Shamji; Stuart L. Schreiber; Ernest Fraenkel; Scott L. Pomeroy; Jill P. Mesirov; Pablo Tamayo; Charles G. Eberhart; Eric Raabe

Purpose: We used human stem and progenitor cells to develop a genetically accurate novel model of MYC-driven Group 3 medulloblastoma. We also developed a new informatics method, Disease-model Signature versus Compound-Variety Enriched Response (“DiSCoVER”), to identify novel therapeutics that target this specific disease subtype. Experimental Design: Human neural stem and progenitor cells derived from the cerebellar anlage were transduced with oncogenic elements associated with aggressive medulloblastoma. An in silico analysis method for screening drug sensitivity databases (DiSCoVER) was used in multiple drug sensitivity datasets. We validated the top hits from this analysis in vitro and in vivo. Results: Human neural stem and progenitor cells transformed with c-MYC, dominant-negative p53, constitutively active AKT and hTERT formed tumors in mice that recapitulated Group 3 medulloblastoma in terms of pathology and expression profile. DiSCoVER analysis predicted that aggressive MYC-driven Group 3 medulloblastoma would be sensitive to cyclin-dependent kinase (CDK) inhibitors. The CDK 4/6 inhibitor palbociclib decreased proliferation, increased apoptosis, and significantly extended the survival of mice with orthotopic medulloblastoma xenografts. Conclusions: We present a new method to generate genetically accurate models of rare tumors, and a companion computational methodology to find therapeutic interventions that target them. We validated our human neural stem cell model of MYC-driven Group 3 medulloblastoma and showed that CDK 4/6 inhibitors are active against this subgroup. Our results suggest that palbociclib is a potential effective treatment for poor prognosis MYC-driven Group 3 medulloblastoma tumors in carefully selected patients. Clin Cancer Res; 22(15); 3903–14. ©2016 AACR.


Journal of Medicinal Chemistry | 2016

Inhibitors of the Cysteine Synthase CysM with Antibacterial Potency against Dormant Mycobacterium tuberculosis

Katharina Brunner; Selma Maric; Rudraraju Srilakshmi Reshma; Helena Almqvist; Brinton Seashore-Ludlow; Anna-Lena Gustavsson; Ömer Poyraz; Perumal Yogeeswari; Thomas Lundbäck; Michaela Vallin; Dharmarajan Sriram; Robert Schnell; Gunter Schneider

Cysteine is an important amino acid in the redox defense of Mycobacterium tuberculosis, primarily as a building block of mycothiol. Genetic studies have implicated de novo cysteine biosynthesis in pathogen survival in infected macrophages, in particular for persistent M. tuberculosis. Here, we report on the identification and characterization of potent inhibitors of CysM, a critical enzyme in cysteine biosynthesis during dormancy. A screening campaign of 17 312 compounds identified ligands that bind to the active site with micromolar affinity. These were characterized in terms of their inhibitory potencies and structure-activity relationships through hit expansion guided by three-dimensional structures of enzyme-inhibitor complexes. The top compound binds to CysM with 300 nM affinity and displays selectivity over the mycobacterial homologues CysK1 and CysK2. Notably, two inhibitors show significant potency in a nutrient-starvation model of dormancy of Mycobacterium tuberculosis, with little or no cytotoxicity toward mammalian cells.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Prediction of intracellular exposure bridges the gap between target- and cell-based drug discovery

André Mateus; Laurie J. Gordon; Gareth Wayne; Helena Almqvist; Hanna Axelsson; Brinton Seashore-Ludlow; Andrea Treyer; Pär Matsson; Thomas Lundbäck; Andrew West; Michael M. Hann; Per Artursson

Significance Exposure at the site of action has been identified as one of the three most important factors for success in drug discovery and the design of chemical probes. Modern drug discovery programs have, to a great extent, shifted to intracellular targets, but methods to determine intracellular drug concentrations have been lacking. Here, we use a methodology for predicting intracellular exposure of small-molecule drugs to understand their potency toward intracellular targets. We show that our approach is generally applicable to multiple targets, cell types, and therapeutic areas. We expect that routine measurements of intracellular drug concentration will contribute to reducing the high attrition observed in drug discovery and the design of both better chemical probes and medicines. Inadequate target exposure is a major cause of high attrition in drug discovery. Here, we show that a label-free method for quantifying the intracellular bioavailability (Fic) of drug molecules predicts drug access to intracellular targets and hence, pharmacological effect. We determined Fic in multiple cellular assays and cell types representing different targets from a number of therapeutic areas, including cancer, inflammation, and dementia. Both cytosolic targets and targets localized in subcellular compartments were investigated. Fic gives insights on membrane-permeable compounds in terms of cellular potency and intracellular target engagement, compared with biochemical potency measurements alone. Knowledge of the amount of drug that is locally available to bind intracellular targets provides a powerful tool for compound selection in early drug discovery.


Journal of Biomolecular Screening | 2016

Early Perspective: Microplate Applications of the Cellular Thermal Shift Assay (CETSA)

Brinton Seashore-Ludlow; Thomas Lundbäck

The cellular thermal shift assay (CETSA) was introduced in 2013 as a means to assess drug binding in complex environments such as cell lysates, live cells, and even tissues. The assay principle relies on the well-proven biophysical concept of ligand-induced thermal stabilization of proteins, which in CETSA applications is measured as a persistent presence of soluble protein at elevated temperatures. Given its recent development, we have just started to learn about the benefits and pitfalls of the method as it is applied to a growing number of protein target classes, the majority of which are intracellular soluble proteins. One of the early technology developments concerned the transfer of the original assay procedure from PCR tubes and Western blot detection of soluble protein to a homogeneous assay in high-density microplates. A move to high-throughput formats is essential for a more systematic application in drug discovery settings, as well as in academic efforts for validating chemical probes through studies of structure–activity relationships. This perspective aims at providing an overview of knowledge gained in microplate formatting of CETSA and makes an attempt at forecasting future applications.


Journal of the American Chemical Society | 2015

Discovery of a Small-Molecule Probe for V-ATPase Function

Leslie N. Aldrich; Szu Yu Kuo; Adam B. Castoreno; Gautam Goel; Petric Kuballa; Matthew G. Rees; Brinton Seashore-Ludlow; Jaime H. Cheah; Isabel Latorre; Stuart L. Schreiber; Alykhan F. Shamji; Ramnik J. Xavier

Lysosomes perform a critical cellular function as a site of degradation for diverse cargoes including proteins, organelles, and pathogens delivered through distinct pathways, and defects in lysosomal function have been implicated in a number of diseases. Recent studies have elucidated roles for the lysosome in the regulation of protein synthesis, metabolism, membrane integrity, and other processes involved in homeostasis. Complex small-molecule natural products have greatly contributed to the investigation of lysosomal function in cellular physiology. Here we report the discovery of a novel, small-molecule modulator of lysosomal acidification derived from diversity-oriented synthesis through high-content screening.

Collaboration


Dive into the Brinton Seashore-Ludlow'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

Pablo Tamayo

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric Raabe

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ernest Fraenkel

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge