Network


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

Hotspot


Dive into the research topics where Richard Marcellus is active.

Publication


Featured researches published by Richard Marcellus.


Nature Communications | 2012

Catalytic site remodelling of the DOT1L methyltransferase by selective inhibitors.

Wenyu Yu; Emma J. Chory; Amy K. Wernimont; Wolfram Tempel; Alex Scopton; Alexander J. Federation; Jason J. Marineau; Jun Qi; Dalia Barsyte-Lovejoy; Joanna Yi; Richard Marcellus; Roxana E. Iacob; John R. Engen; Carly Griffin; Ahmed Aman; Erno Wienholds; Fengling Li; Javier Pineda; Guillermina Estiu; Tatiana Shatseva; Taraneh Hajian; Rima Al-awar; John E. Dick; Masoud Vedadi; Peter J. Brown; C.H. Arrowsmith; James E. Bradner; Matthieu Schapira

Selective inhibition of protein methyltransferases is a promising new approach to drug discovery. An attractive strategy towards this goal is the development of compounds that selectively inhibit binding of the cofactor, S-adenosylmethionine, within specific protein methyltransferases. Here we report the three-dimensional structure of the protein methyltransferase DOT1L bound to EPZ004777, the first S-adenosylmethionine-competitive inhibitor of a protein methyltransferase with in vivo efficacy. This structure and those of four new analogues reveal remodelling of the catalytic site. EPZ004777 and a brominated analogue, SGC0946, inhibit DOT1L in vitro and selectively kill mixed lineage leukaemia cells, in which DOT1L is aberrantly localized via interaction with an oncogenic MLL fusion protein. These data provide important new insight into mechanisms of cell-active S-adenosylmethionine-competitive protein methyltransferase inhibitors, and establish a foundation for the further development of drug-like inhibitors of DOT1L for cancer therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2014

(R)-PFI-2 is a potent and selective inhibitor of SETD7 methyltransferase activity in cells

Dalia Barsyte-Lovejoy; Fengling Li; Menno J. Oudhoff; John Howard Tatlock; Aiping Dong; Hong Zeng; Hong Wu; Spencer A. Freeman; Matthieu Schapira; Guillermo Senisterra; Ekaterina Kuznetsova; Richard Marcellus; Abdellah Allali-Hassani; Steven Kennedy; Jean-Philippe Lambert; Amber L. Couzens; Ahmed Aman; Anne-Claude Gingras; Rima Al-awar; Paul V. Fish; Brian S. Gerstenberger; Lee R. Roberts; Caroline L. Benn; Rachel L. Grimley; Mitchell J.S. Braam; Fabio Rossi; Marius Sudol; Peter J. Brown; Mark Edward Bunnage; Dafydd R. Owen

Significance Protein methyltransferases constitute an emerging but undercharacterized class of therapeutic targets with diverse roles in normal human biology and disease. Small-molecule “chemical probes” can be powerful tools for the functional characterization of such enzymes, and here we report the discovery of (R)-PFI-2—a first-in-class, potent, highly selective, and cell-active inhibitor of the methyltransferase activity of SETD7 [SET domain containing (lysine methyltransferase) 7]—and two related compounds for control and chemoproteomics studies. We used these compounds to characterize the role of SETD7 in signaling, in the Hippo pathway, that controls cell growth and organ size. Our work establishes a chemical biology tool kit for the study of the diverse roles of SETD7 in cells and further validates protein methyltransferases as a druggable target class. SET domain containing (lysine methyltransferase) 7 (SETD7) is implicated in multiple signaling and disease related pathways with a broad diversity of reported substrates. Here, we report the discovery of (R)-PFI-2—a first-in-class, potent (Kiapp = 0.33 nM), selective, and cell-active inhibitor of the methyltransferase activity of human SETD7—and its 500-fold less active enantiomer, (S)-PFI-2. (R)-PFI-2 exhibits an unusual cofactor-dependent and substrate-competitive inhibitory mechanism by occupying the substrate peptide binding groove of SETD7, including the catalytic lysine-binding channel, and by making direct contact with the donor methyl group of the cofactor, S-adenosylmethionine. Chemoproteomics experiments using a biotinylated derivative of (R)-PFI-2 demonstrated dose-dependent competition for binding to endogenous SETD7 in MCF7 cells pretreated with (R)-PFI-2. In murine embryonic fibroblasts, (R)-PFI-2 treatment phenocopied the effects of Setd7 deficiency on Hippo pathway signaling, via modulation of the transcriptional coactivator Yes-associated protein (YAP) and regulation of YAP target genes. In confluent MCF7 cells, (R)-PFI-2 rapidly altered YAP localization, suggesting continuous and dynamic regulation of YAP by the methyltransferase activity of SETD7. These data establish (R)-PFI-2 and related compounds as a valuable tool-kit for the study of the diverse roles of SETD7 in cells and further validate protein methyltransferases as a druggable target class.


Structure | 2012

An Allosteric Inhibitor of Protein Arginine Methyltransferase 3

Alena Siarheyeva; Guillermo Senisterra; Abdellah Allali-Hassani; Aiping Dong; Elena Dobrovetsky; Gregory A. Wasney; Irene Chau; Richard Marcellus; Taraneh Hajian; Feng Liu; Ilia Korboukh; David Smil; Yuri Bolshan; Jinrong Min; Hong Wu; Hong Zeng; Peter Loppnau; Gennadiy Poda; Carly Griffin; Ahmed Aman; Peter J. Brown; Jian Jin; Rima Al-awar; C.H. Arrowsmith; Matthieu Schapira; Masoud Vedadi

PRMT3, a protein arginine methyltransferase, has been shown to influence ribosomal biosynthesis by catalyzing the dimethylation of the 40S ribosomal protein S2. Although PRMT3 has been reported to be a cytosolic protein, it has been shown to methylate histone H4 peptide (H4 1-24) in vitro. Here, we report the identification of a PRMT3 inhibitor (1-(benzo[d][1,2,3]thiadiazol-6-yl)-3-(2-cyclohexenylethyl)urea; compound 1) with IC50 value of 2.5 μM by screening a library of 16,000 compounds using H4 (1-24) peptide as a substrate. The crystal structure of PRMT3 in complex with compound 1 as well as kinetic analysis reveals an allosteric mechanism of inhibition. Mutating PRMT3 residues within the allosteric site or using compound 1 analogs that disrupt interactions with allosteric site residues both abrogated binding and inhibitory activity. These data demonstrate an allosteric mechanism for inhibition of protein arginine methyltransferases, an emerging class of therapeutic targets.


Journal of Medicinal Chemistry | 2016

Structure-Based Optimization of a Small Molecule Antagonist of the Interaction Between WD Repeat-Containing Protein 5 (WDR5) and Mixed-Lineage Leukemia 1 (MLL1).

Matthaeus Getlik; David Smil; Carlos Zepeda-Velazquez; Yuri Bolshan; Gennady Poda; Hong Wu; Aiping Dong; Ekaterina Kuznetsova; Richard Marcellus; Guillermo Senisterra; Ludmila Dombrovski; Taraneh Hajian; Taira Kiyota; Matthieu Schapira; C.H. Arrowsmith; Peter J. Brown; Masoud Vedadi; Rima Al-awar

WD repeat-containing protein 5 (WDR5) is an important component of the multiprotein complex essential for activating mixed-lineage leukemia 1 (MLL1). Rearrangement of the MLL1 gene is associated with onset and progression of acute myeloid and lymphoblastic leukemias, and targeting the WDR5-MLL1 interaction may result in new cancer therapeutics. Our previous work showed that binding of small molecule ligands to WDR5 can modulate its interaction with MLL1, suppressing MLL1 methyltransferase activity. Initial structure-activity relationship studies identified N-(2-(4-methylpiperazin-1-yl)-5-substituted-phenyl) benzamides as potent and selective antagonists of this protein-protein interaction. Guided by crystal structure data and supported by in silico library design, we optimized the scaffold by varying the C-1 benzamide and C-5 substituents. This allowed us to develop the first highly potent (Kdisp < 100 nM) small molecule antagonists of the WDR5-MLL1 interaction and demonstrate that N-(4-(4-methylpiperazin-1-yl)-3-(morpholinomethyl)-[1,1-biphenyl]-3-yl)-6-oxo-4-(trifluoromethyl)-1,6-dihydropyridine-3-carboxamide 16d (OICR-9429) is a potent and selective chemical probe suitable to help dissect the biological role of WDR5.


Oncotarget | 2016

Small molecule epigenetic screen identifies novel EZH2 and HDAC inhibitors that target glioblastoma brain tumor-initiating cells

Natalie Grinshtein; Constanza Rioseco; Richard Marcellus; David Uehling; Ahmed Aman; Xueqing Lun; Osamu Muto; Lauren Podmore; Jake Lever; Yaoqing Shen; Michael D. Blough; Greg Cairncross; Stephen M. Robbins; Steven J.M. Jones; Marco A. Marra; Rima Al-awar; Donna L. Senger; David R. Kaplan

Glioblastoma (GBM) is the most lethal and aggressive adult brain tumor, requiring the development of efficacious therapeutics. Towards this goal, we screened five genetically distinct patient-derived brain-tumor initiating cell lines (BTIC) with a unique collection of small molecule epigenetic modulators from the Structural Genomics Consortium (SGC). We identified multiple hits that inhibited the growth of BTICs in vitro, and further evaluated the therapeutic potential of EZH2 and HDAC inhibitors due to the high relevance of these targets for GBM. We found that the novel SAM-competitive EZH2 inhibitor UNC1999 exhibited low micromolar cytotoxicity in vitro on a diverse collection of BTIC lines, synergized with dexamethasone (DEX) and suppressed tumor growth in vivo in combination with DEX. In addition, a unique brain-penetrant class I HDAC inhibitor exhibited cytotoxicity in vitro on a panel of BTIC lines and extended survival in combination with TMZ in an orthotopic BTIC model in vivo. Finally, a combination of EZH2 and HDAC inhibitors demonstrated synergy in vitro by augmenting apoptosis and increasing DNA damage. Our findings identify key epigenetic modulators in GBM that regulate BTIC growth and survival and highlight promising combination therapies.


Breast Cancer Research | 2016

Downregulation of histone H2A and H2B pathways is associated with anthracycline sensitivity in breast cancer

Marsela Braunstein; Linda Liao; Nicola Lyttle; Nazleen Lobo; Karen Taylor; Paul M. Krzyzanowski; Irina Kalatskaya; Cindy Q. Yao; Lincoln Stein; Paul C. Boutros; Chris Twelves; Richard Marcellus; John M.S. Bartlett; Melanie Spears

BackgroundDrug resistance in breast cancer is the major obstacle to effective treatment with chemotherapy. While upregulation of multidrug resistance genes is an important component of drug resistance mechanisms in vitro, their clinical relevance remains to be determined. Therefore, identifying pathways that could be targeted in the clinic to eliminate anthracycline-resistant breast cancer remains a major challenge.MethodsWe generated paired native and epirubicin-resistant MDA-MB-231, MCF7, SKBR3 and ZR-75-1 epirubicin-resistant breast cancer cell lines to identify pathways contributing to anthracycline resistance. Native cell lines were exposed to increasing concentrations of epirubicin until resistant cells were generated. To identify mechanisms driving epirubicin resistance, we used a complementary approach including gene expression analyses to identify molecular pathways involved in resistance, and small-molecule inhibitors to reverse resistance. In addition, we tested its clinical relevance in a BR9601 adjuvant clinical trial.ResultsCharacterisation of epirubicin-resistant cells revealed that they were cross-resistant to doxorubicin and SN-38 and had alterations in apoptosis and cell-cycle profiles. Gene expression analysis identified deregulation of histone H2A and H2B genes in all four cell lines. Histone deacetylase small-molecule inhibitors reversed resistance and were cytotoxic for epirubicin-resistant cell lines, confirming that histone pathways are associated with epirubicin resistance. Gene expression of a novel 18-gene histone pathway module analysis of the BR9601 adjuvant clinical trial revealed that patients with low expression of the 18-gene histone module benefited from anthracycline treatment more than those with high expression (hazard ratio 0.35, 95xa0% confidence interval 0.13–0.96, pu2009=u20090.042).ConclusionsThis study revealed a key pathway that contributes to anthracycline resistance and established model systems for investigating drug resistance in all four major breast cancer subtypes. As the histone modification can be targeted with small-molecule inhibitors, it represents a possible means of reversing clinical anthracycline resistance.Trial registrationClinicalTrials.gov identifier NCT00003012. Registered on 1 November 1999.


Stem cell reports | 2016

Identification of RSK and TTK as Modulators of Blood Vessel Morphogenesis Using an Embryonic Stem Cell-Based Vascular Differentiation Assay

Lamis Hammoud; Jessica R. Adams; Amanda J. Loch; Richard Marcellus; David Uehling; Ahmed Aman; Christopher Fladd; Trevor D. McKee; Christine E.B. Jo; Rima Al-awar; Sean E. Egan; Janet Rossant

Summary Blood vessels are formed through vasculogenesis, followed by remodeling of the endothelial network through angiogenesis. Many events that occur during embryonic vascular development are recapitulated during adult neoangiogenesis, which is critical to tumor growth and metastasis. Current antiangiogenic tumor therapies, based largely on targeting the vascular endothelial growth factor pathway, show limited clinical benefits, thus necessitating the discovery of alternative targets. Here we report the development of a robust embryonic stem cell-based vascular differentiation assay amenable to small-molecule screens to identify novel modulators of angiogenesis. In this context, RSK and TTK were identified as angiogenic modulators. Inhibition of these pathways inhibited angiogenesis in embryoid bodies and human umbilical vein endothelial cells. Furthermore, inhibition of RSK and TTK reduced tumor growth, vascular density, and improved survival in an in vivo Lewis lung carcinoma mouse model. Our study suggests that RSK and TTK are potential targets for antiangiogenic therapy, and provides an assay system for further pathway screens.


Cancer Research | 2012

Abstract 156: Screening and characterization of inhibitors of G-protein coupled receptor kinase GRK6 as potential therapeutics for multiple myeloma

Carly Griffin; Ratheesh Subramanian; Hassan S. Zaidi; Richard Marcellus; Babu Joseph; Methvin Isaac; David Uehling; Chungyee Leung-Hagesteijn; Rodger Tiedemann; Rima Al-awar

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, ILnnMultiple myeloma (MM) is one of the most common hematological malignancies, but current therapy options are limited to high-dose chemotherapy or high-risk stem-cell transplantation. It is clear that the development of more selective and less toxic treatments for MM is greatly needed. In a recent kinome-wide RNAi study by Tiedemann and colleagues (2010), the G-protein coupled receptor kinase-6 (GRK6) was identified as a critical kinase required for survival of MM cells. This study also suggests that MM cells, but not other cell types, are dependent on GRK6; and that gene silencing by shRNA or siRNA of GRK6, but not other GRKs, results in decreased survival. At present, the G-protein coupled receptor (GPCR) signaling mediated by GRK6 in MM cells is not well understood. Our current research aims to identify the important GPCRs phosphorylated by GRK6 and the signaling proteins/pathways implicated in survival of multiple myeloma cells. Through gene silencing techniques and expression of either the wild-type or kinase-dead form of GRK6 protein, we have determined that a functional GRK6 kinase domain is required for survival of MM cells. We have also demonstrated that the signaling pathway downstream of CXCR4 phosphorylation by GRK6 is defective in cells expressing the kinase-dead GRK6-mutant protein. This loss of signaling by the GRK6-mutant protein was observed for other GPCRs found to be important for survival of MM cells. These findings helped to validate that the kinase domain of GRK6 is a potential target for MM, and spurred the identification of small molecule kinase inhibitors of GRK6. Compounds with high potency in preliminary biochemical assays were screened against MM and non-MM cell lines to evaluate their activity and specificity in vivo. Small molecule inhibition of GRK6 kinase activity is a novel approach to the treatment of MM, as it specifically targets a protein found to be critical for survival of these cells through an unbiased RNAi study. Treatment options for patients with MM are limited and based on our preliminary findings small molecule inhibitors of GRK6 offer an alternative therapeutic approach to the treatment of MM.nnCitation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 156. doi:1538-7445.AM2012-156


Cancer Research | 2012

Abstract 4989: Selective inhibitors of the inositol-requiring enzyme 1 kinase domain

Carly Griffin; Ayome Abibi; Ratheesh Subramaniam; Hassan S. Zaidi; Richard Marcellus; Gennadiy Poda; Michael Prakesch; David Uehling; Marella D. Canny; David Chiovitti; Daniel Durocher; Frank Sicheri; Rima Al-awar

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, ILnnInositol-requiring enzyme 1 (IRE1) is a key player in endoplasmic reticulum (ER) stress conditions. IRE1 is a highly conserved ER-membrane protein activated by the unfolded protein response (UPR) or other ER-stressors, such as hypoxia and glucose deprivation. Stress causes IRE1 to undergo oligomerization and autophosphorylation, which triggers nonconventional splicing of XBP-1 mRNA by its cytosolic endonuclease domain. The resulting spliced XBP-1 protein (XBP-1s) is a transcription factor that serves to increase the protein folding capacity and ultimately restore homeostasis of the ER. Thus, sustained IRE1 activity promotes cell survival and inhibition of IRE1 may be a potential therapeutic target for diseases associated with chronic ER-stress, such as neurodegenerative disorders, diabetes, and cancer. Proper RNase function of IRE1 is dependent upon autophosphorylation of the kinase domain. We therefore screened a library of 380 known kinase inhibitors, consisting of tool compounds and compounds already in clinical use, for those with activity against the human IRE1 kinase domain. As a result, a number of compounds were found that potently inhibit phosphorylation of a biotin-STK peptide substrate in the presence of human IRE1 (IC50 < 1 μM), as determined by HTRF (homogeneous time-resolved fluorescence). The lead compounds were then screened in cell-based assays. Several ATP-mimetic compounds with diverse chemotypes were found to inhibit expression of XBP-1s in human cancer cells under pharmacologically-induced acute ER-stress. Furthermore, transcriptional targets of XBP-1s and phosphorylation of IRE1 were also negatively affected by these compounds. Interestingly one compound in particular, a known ROCK1 (Rho-associated coiled-coil containing protein kinase 1) inhibitor (OICR000287A), was significantly more toxic to cells under acute ER-stress than to unstressed cells. This study suggests that development of ATP-competitive inhibitors of human IRE1 is a promising therapeutic strategy for ER-stress related diseases including myeloma, pancreatic and other secretory cancers.nnCitation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4989. doi:1538-7445.AM2012-4989


Nature Chemical Biology | 2015

Pharmacological targeting of the Wdr5-MLL interaction in C/EBPα N-terminal leukemia

Florian Grebien; Masoud Vedadi; Matthäus Getlik; Roberto Giambruno; Amit Grover; Roberto Avellino; Anna Skucha; Sarah Vittori; Ekaterina Kuznetsova; David Smil; Dalia Barsyte-Lovejoy; Fengling Li; Gennadiy Poda; Matthieu Schapira; Hong Wu; Aiping Dong; Guillermo Senisterra; Alexey Stukalov; Kilian Huber; Andreas Schönegger; Richard Marcellus; Martin Bilban; Christoph Bock; Peter J. Brown; Johannes Zuber; Keiryn L. Bennett; Rima Al-awar; Ruud Delwel; Claus Nerlov; C.H. Arrowsmith

Collaboration


Dive into the Richard Marcellus's collaboration.

Top Co-Authors

Avatar

Rima Al-awar

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Ahmed Aman

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carly Griffin

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

David Uehling

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gennadiy Poda

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge