Network


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

Hotspot


Dive into the research topics where Patrick W. Faloon is active.

Publication


Featured researches published by Patrick W. Faloon.


Nature Chemical Biology | 2016

Identification of cancer-cytotoxic modulators of PDE3A by predictive chemogenomics

Luc de Waal; Tim Lewis; Matthew G. Rees; Aviad Tsherniak; Xiaoyun Wu; Peter S. Choi; Lara Gechijian; Christina R. Hartigan; Patrick W. Faloon; Mark Hickey; Nicola Tolliday; Steven A. Carr; Paul A. Clemons; Benito Munoz; Bridget K. Wagner; Alykhan F. Shamji; Angela N. Koehler; Monica Schenone; Alex B. Burgin; Stuart L. Schreiber; Heidi Greulich; Matthew Meyerson

High cancer death rates indicate the need for new anti-cancer therapeutic agents. Approaches to discover new cancer drugs include target-based drug discovery and phenotypic screening. Here, we identified phosphodiesterase 3A modulators as cell-selective cancer cytotoxic compounds by phenotypic compound library screening and target deconvolution by predictive chemogenomics. We found that sensitivity to 6-(4-(diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one, or DNMDP, across 766 cancer cell lines correlates with expression of the phosphodiesterase 3A gene, PDE3A. Like DNMDP, a subset of known PDE3A inhibitors kill selected cancer cells while others do not. Furthermore, PDE3A depletion leads to DNMDP resistance. We demonstrated that DNMDP binding to PDE3A promotes an interaction between PDE3A and Schlafen 12 (SLFN12), suggesting a neomorphic activity. Co-expression of SLFN12 with PDE3A correlates with DNMDP sensitivity, while depletion of SLFN12 results in decreased DNMDP sensitivity. Our results implicate PDE3A modulators as candidate cancer therapeutic agents and demonstrate the power of predictive chemogenomics in small-molecule discovery.


Journal of Biomolecular Screening | 2014

Assay Development and High-Throughput Screening for Inhibitors of Kaposi’s Sarcoma–Associated Herpesvirus N-Terminal Latency-Associated Nuclear Antigen Binding to Nucleosomes

Chantal Beauchemin; Nathan J. Moerke; Patrick W. Faloon; Kenneth M. Kaye

Kaposi’s sarcoma–associated herpesvirus (KSHV) has a causative role in several human malignancies, especially in immunocompromised hosts. KSHV latently infects tumor cells and persists as an extrachromosomal episome (plasmid). KSHV latency-associated nuclear antigen (LANA) mediates KSHV episome persistence. LANA binds specific KSHV sequence to replicate viral DNA. In addition, LANA tethers KSHV genomes to mitotic chromosomes to efficiently segregate episomes to daughter nuclei after mitosis. N-terminal LANA (N-LANA) binds histones H2A and H2B to attach to chromosomes. Currently, there are no specific inhibitors of KSHV latent infection. To enable high-throughput screening (HTS) of inhibitors of N-LANA binding to nucleosomes, here we develop, miniaturize, and validate a fluorescence polarization (FP) assay that detects fluorophore-labeled N-LANA peptide binding to nucleosomes. We also miniaturize a counterscreen to identify DNA intercalators that nonspecifically inhibit N-LANA binding to nucleosomes, and also develop an enzyme-linked immunosorbent assay to assess N-LANA binding to nucleosomes in the absence of fluorescence. HTS of libraries containing more than 350,000 compounds identified multiple compounds that inhibited N-LANA binding to nucleosomes. No compounds survived all counterscreens, however. More complex small-molecule libraries will likely be necessary to identify specific inhibitors of N-LANA binding to histones H2A and H2B; these assays should prove useful for future screens.


Journal of the American Chemical Society | 2015

Kinase-Independent Small-Molecule Inhibition of JAK-STAT Signaling.

Danny Hung-Chieh Chou; Amedeo Vetere; Amit Choudhary; Stephen S. Scully; Monica Schenone; Alicia Tang; Rachel Gomez; Sean M. Burns; Morten Lundh; Tamara Vital; Eamon Comer; Patrick W. Faloon; Vlado Dančík; Christie Ciarlo; Joshiawa Paulk; Mingji Dai; Clark Reddy; Hanshi Sun; Matthew A. Young; Nicholas J. Donato; Jacob D. Jaffe; Paul A. Clemons; Michelle Palmer; Steven A. Carr; Stuart L. Schreiber; Bridget K. Wagner

Phenotypic cell-based screening is a powerful approach to small-molecule discovery, but a major challenge of this strategy lies in determining the intracellular target and mechanism of action (MoA) for validated hits. Here, we show that the small-molecule BRD0476, a novel suppressor of pancreatic β-cell apoptosis, inhibits interferon-gamma (IFN-γ)-induced Janus kinase 2 (JAK2) and signal transducer and activation of transcription 1 (STAT1) signaling to promote β-cell survival. However, unlike common JAK-STAT pathway inhibitors, BRD0476 inhibits JAK-STAT signaling without suppressing the kinase activity of any JAK. Rather, we identified the deubiquitinase ubiquitin-specific peptidase 9X (USP9X) as an intracellular target, using a quantitative proteomic analysis in rat β cells. RNAi-mediated and CRISPR/Cas9 knockdown mimicked the effects of BRD0476, and reverse chemical genetics using a known inhibitor of USP9X blocked JAK-STAT signaling without suppressing JAK activity. Site-directed mutagenesis of a putative ubiquitination site on JAK2 mitigated BRD0476 activity, suggesting a competition between phosphorylation and ubiquitination to explain small-molecule MoA. These results demonstrate that phenotypic screening, followed by comprehensive MoA efforts, can provide novel mechanistic insights into ostensibly well-understood cell signaling pathways. Furthermore, these results uncover USP9X as a potential target for regulating JAK2 activity in cellular inflammation.


Bioorganic & Medicinal Chemistry Letters | 2015

Benzo-fused lactams from a diversity-oriented synthesis (DOS) library as inhibitors of scavenger receptor BI (SR-BI)-mediated lipid uptake

Chris Dockendorff; Patrick W. Faloon; Jun Pu; Miao Yu; Stephen Johnston; Melissa Bennion; Marsha Penman; Thomas J.F. Nieland; Sivaraman Dandapani; Jose R. Perez; Benito Munoz; Michelle Palmer; Stuart L. Schreiber; Monty Krieger

We report a new series of 8-membered benzo-fused lactams that inhibit cellular lipid uptake from HDL particles mediated by Scavenger Receptor, Class B, Type I (SR-BI). The series was identified via a high-throughput screen of the National Institutes of Health Molecular Libraries Small Molecule Repository (NIH MLSMR), measuring the transfer of the fluorescent lipid DiI from HDL particles to CHO cells overexpressing SR-BI. The series is part of a previously reported diversity-oriented synthesis (DOS) library prepared via a build-couple-pair approach. Detailed structure-activity relationship (SAR) studies were performed with a selection of the original library, as well as additional analogs prepared via solution phase synthesis. These studies demonstrate that the orientation of the substituents on the aliphatic ring have a critical effect on activity. Additionally, a lipophilic group is required at the western end of the molecule, and a northern hydroxyl group and a southern sulfonamide substituent also proved to be optimal. Compound 2p was found to possess a superior combination of potency (av IC50=0.10μM) and solubility (79μM in PBS), and it was designated as probe ML312.


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery of bisamide-heterocycles as inhibitors of scavenger receptor BI (SR-BI)-mediated lipid uptake.

Chris Dockendorff; Patrick W. Faloon; Andrew Germain; Miao Yu; Willmen Youngsaye; Partha Nag; Melissa Bennion; Marsha Penman; Thomas J.F. Nieland; Sivaraman Dandapani; Jose R. Perez; Benito Munoz; Michelle Palmer; Stuart L. Schreiber; Monty Krieger

A new series of potent inhibitors of cellular lipid uptake from HDL particles mediated by scavenger receptor, class B, type I (SR-BI) was identified. The series was identified via a high-throughput screen of the National Institutes of Health Molecular Libraries Small Molecule Repository (NIH MLSMR) that measured the transfer of the fluorescent lipid DiI from HDL particles to CHO cells overexpressing SR-BI. The series is characterized by a linear peptidomimetic scaffold with two adjacent amide groups, as well as an aryl-substituted heterocycle. Analogs of the initial hit were rapidly prepared via Ugi 4-component reaction, and select enantiopure compounds were prepared via a stepwise sequence. Structure-activity relationship (SAR) studies suggest an oxygenated arene is preferred at the western end of the molecule, as well as highly lipophilic substituents on the central and eastern nitrogens. Compound 5e, with (R)-stereochemistry at the central carbon, was designated as probe ML279. Mechanistic studies indicate that ML279 stabilizes the interaction of HDL particles with SR-BI, and its effect is reversible. It shows good potency (IC50=17 nM), is non-toxic, plasma stable, and has improved solubility over our alternative probe ML278.


ACS Chemical Biology | 2018

Functionally biased D2R antagonists: Targeting the β-arrestin pathway to improve antipsychotic treatment

Michel Weiwer; Qihong Xu; Jennifer Gale; Michael C. Lewis; Arthur J. Campbell; Frederick A. Schroeder; Genevieve C. Van de Bittner; Michelle Walk; Aldo Amaya; Ping Su; Luka D̵ordevic; Joshua Sacher; Adam Skepner; David Fei; Kelly Dennehy; Shannon Nguyen; Patrick W. Faloon; Jose R. Perez; Jeffrey R. Cottrell; Fang Liu; Michelle Palmer; Jen Q. Pan; Jacob M. Hooker; Yan-Ling Zhang; Edward M. Scolnick; Florence F. Wagner; Edward B. Holson

Schizophrenia is a severe neuropsychiatric disease that lacks completely effective and safe therapies. As a polygenic disorder, genetic studies have only started to shed light on its complex etiology. To date, the positive symptoms of schizophrenia are well-managed by antipsychotic drugs, which primarily target the dopamine D2 receptor (D2R). However, these antipsychotics are often accompanied by severe side effects, including motoric symptoms. At D2R, antipsychotic drugs antagonize both G-protein dependent (Gαi/o) signaling and G-protein independent (β-arrestin) signaling. However, the relevant contributions of the distinct D2R signaling pathways to antipsychotic efficacy and on-target side effects (motoric) are still incompletely understood. Recent evidence from mouse genetic and pharmacological studies point to β-arrestin signaling as the major driver of antipsychotic efficacy and suggest that a β-arrestin biased D2R antagonist could achieve an additional level of selectivity at D2R, increasing the therapeutic index of next generation antipsychotics. Here, we characterize BRD5814, a highly brain penetrant β-arrestin biased D2R antagonist. BRD5814 demonstrated good target engagement via PET imaging, achieving efficacy in an amphetamine-induced hyperlocomotion mouse model with strongly reduced motoric side effects in a rotarod performance test. This proof of concept study opens the possibility for the development of a new generation of pathway selective antipsychotics at D2R with reduced side effect profiles for the treatment of schizophrenia.


Molecular Cancer Therapeutics | 2015

Abstract C136: Identification of selective cancer cytotoxic modulators of phosphodiesterase 3a by predictive chemogenomics

Lucian de Waal; Tim Lewis; Matthew G. Rees; Aviad Tsherniak; Xiaoyun Wu; Peter S. Choi; Lara Gechijian; Christina R. Hartigan; Patrick W. Faloon; Mark Hickey; Nicola Tolliday; Steven A. Carr; Paul A. Clemons; Benito Munoz; Bridget K. Wagner; Alykhan F. Shamji; Angela N. Koehler; Monica Schenone; Alex B. Burgin; Stuart L. Schreiber; Heidi Greulich; Matthew Meyerson

High cancer death rates indicate the need for new anti-cancer therapeutic agents. Approaches to discover new cancer drugs include target-based drug discovery and phenotypic screening. Here, we identify phosphodiesterase 3A modulators as cell-selective cancer cytotoxic compounds by phenotypic compound library screening and target deconvolution by predictive chemogenomics. We found that sensitivity to 6-(4-(diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one, or DNMDP, across 766 cancer cell lines correlates with expression of the phosphodiesterase 3A gene, PDE3A. Like DNMDP, a subset of known PDE3A inhibitors kill selected cancer cells while others do not. Furthermore, PDE3A depletion leads to DNMDP resistance. We demonstrate that DNMDP binding to PDE3A promotes an interaction between PDE3A and Schlafen 12 (SLFN12), suggesting a neomorphic activity. Co-expression of SLFN12 with PDE3A correlates with DNMDP sensitivity, while depletion of SLFN12 results in decreased sensitivity to DNMDP. Our results implicate PDE3A modulators as candidate cancer therapeutic agents and demonstrate the power of predictive chemogenomics in small-molecule discovery. Citation Format: Lucian de Waal, Timothy A. Lewis, Matthew G. Rees, Aviad Tsherniak, Xiaoyun Wu, Peter S. Choi, Lara Gechijian, Christina Hartigan, Patrick W. Faloon, Mark J. Hickey, Nicola Tolliday, Steven A. Carr, Paul A. Clemons, Benito Munoz, Bridget K. Wagner, Alykhan F. Shamji, Angela N. Koehler, Monica Schenone, Alex B. Burgin, Stuart L. Schreiber, Heidi Greulich, Matthew Meyerson. Identification of selective cancer cytotoxic modulators of phosphodiesterase 3a by predictive chemogenomics. [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 C136.


Cancer Research | 2014

Abstract 4596: An integrated genomic characterization of the target of a small molecule identifies a novel cancer dependency

Luc de Waal; Tim Lewis; Lara Gechijian; Aviad Tsherniak; Willmen Youngsaye; Matthew G. Rees; Oliver R. Mikse; Mark Hickey; Patrick W. Faloon; Nicola Tolliday; Angela N. Koehler; Monica Schenone; Kwok K. Wong; Alykhan F. Shamji; Benito Munoz; Stuart L. Schreiber; Heidi Greulich; Matthew Meyerson

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Recent large sequencing and cancer dependency studies have accelerated the identification of candidate targets for precision medicine. However, the current drug development paradigm starting with target identification and validation can be slow and has thus far yielded a limited variety of successful targets. We sought to return to an empirical approach to drug discovery and performed a high throughput screen to identify small molecules that were both potent and selective. In a primary screen of 2000 compounds in two cell-lines: A549 and H1734, three compounds only affected H1734 viability. One of which validated in a dose-response experiment with great potency and specificity, we called this small molecule ‘Compound 1B’. In an effort to identify the target of Compound 1B, we profiled 766 genomically-characterized cancer cell lines and found that approximately 4% were sensitive to our compound. Sensitivity was not restricted to a particular tissue of origin. Interestingly, expression of Phosphodiesterase 3A (PDE3A) correlated with cytotoxicity. We further showed that Compound 1B specifically inhibited the enzymatic activity of PDE3A and PDE3B in a panel of 11 different phosphodiesterase family members. However, only a subset of other PDE3 inhibitors shared the same cytotoxic phenotype of Compound 1B. In a rescue screen of 1600 bioactive compounds, we identified the non-lethal PDE3 inhibitors as compounds that were able to rescue cell death induced by Compound 1B. Biochemical assays showed that both Compound 1B, cytotoxic and non-cytotoxic PDE3 inhibitors compete for binding to PDE3A. Knockdown of PDE3A did not affect cell viability and inhibited response of sensitive cell lines to Compound 1B. Thus we have identified a potent and selective small molecule that likely acts through PDE3A to induce cancer cell-line cytotoxicity. Our data suggest a hyper- or neomorphic function of PDE3A induced upon binding of Compound 1B. By cross-referencing integrative datasets with compound-sensitivity data, we show that reversal of the current drug-development paradigm can elucidate novel cancer targets, which are not yet identifiable by analysis of large next-generation sequencing datasets. Citation Format: Luc M. de Waal, Tim A. Lewis, Lara Gechijian, Aviad Tsherniak, Willmen Youngsaye, Matthew Rees, Oliver Mikse, Mark Hickey, Patrick Faloon, Nicola Tolliday, Angela Koehler, Monica Schenone, Kwok Wong, Alykhan Shamji, Benito Munoz, Stuart L. Schreiber, Heidi Greulich, Matthew L. Meyerson. An integrated genomic characterization of the target of a small molecule identifies a novel cancer dependency. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4596. doi:10.1158/1538-7445.AM2014-4596


ACS Medicinal Chemistry Letters | 2011

Synthesis of a Novel Suppressor of β-Cell Apoptosis via Diversity-Oriented Synthesis

Danny Hung-Chieh Chou; Jeremy R. Duvall; Baudouin Gerard; Haibo Liu; Bhaumik A. Pandya; Byung-Chul Suh; Erin Forbeck; Patrick W. Faloon; Bridget K. Wagner; Lisa A. Marcaurelle


ACS Medicinal Chemistry Letters | 2015

Indolinyl-Thiazole Based Inhibitors of Scavenger Receptor-BI (SR-BI)-Mediated Lipid Transport

Chris Dockendorff; Patrick W. Faloon; Miao Yu; Willmen Youngsaye; Marsha Penman; Thomas J.F. Nieland; Partha Nag; Tim Lewis; Jun Pu; Melissa Bennion; Joseph Negri; Conor Paterson; Garrett Lam; Sivaraman Dandapani; Jose R. Perez; Benito Munoz; Michelle Palmer; Stuart L. Schreiber; Monty Krieger

Collaboration


Dive into the Patrick W. Faloon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stuart L Schreiber

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge