Network


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

Hotspot


Dive into the research topics where Ahmed Aman is active.

Publication


Featured researches published by Ahmed Aman.


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.


Cancer Research | 2011

Wnt Inhibitor Screen Reveals Iron Dependence of β-Catenin Signaling in Cancers

Siyuan Song; Tania Christova; Stephen Perusini; Solmaz Alizadeh; Renyue Bao; Bryan W. Miller; Rose Hurren; Yulia Jitkova; Marcela Gronda; Methvin Isaac; Babu Joseph; Ratheesh Subramaniam; Ahmed Aman; Anh Chau; Donna E. Hogge; Scott Weir; James Kasper; Aaron D. Schimmer; Rima Al-awar; Jeffery L. Wrana; Liliana Attisano

Excessive signaling from the Wnt pathway is associated with numerous human cancers. Using a high throughput screen designed to detect inhibitors of Wnt/β-catenin signaling, we identified a series of acyl hydrazones that act downstream of the β-catenin destruction complex to inhibit both Wnt-induced and cancer-associated constitutive Wnt signaling via destabilization of β-catenin. We found that these acyl hydrazones bind iron in vitro and in intact cells and that chelating activity is required to abrogate Wnt signaling and block the growth of colorectal cancer cell lines with constitutive Wnt signaling. In addition, we found that multiple iron chelators, desferrioxamine, deferasirox, and ciclopirox olamine similarly blocked Wnt signaling and cell growth. Moreover, in patients with AML administered ciclopirox olamine, we observed decreased expression of the Wnt target gene AXIN2 in leukemic cells. The novel class of acyl hydrazones would thus be prime candidates for further development as chemotherapeutic agents. Taken together, our results reveal a critical requirement for iron in Wnt signaling and they show that iron chelation serves as an effective mechanism to inhibit Wnt signaling in humans.


Journal of Controlled Release | 2012

A docetaxel-carboxymethylcellulose nanoparticle outperforms the approved taxane nanoformulation, Abraxane, in mouse tumor models with significant control of metastases

Mark J. Ernsting; Mami Murakami; Elijus Undzys; Ahmed Aman; Barry Press; Shyh-Dar Li

Cellax is a PEGylated carboxymethylcellulose conjugate of docetaxel (DTX) which condenses into a 120-nm nanoparticle, and was compared against the approved clinical taxane nanoformulation (Abraxane®) in mouse models. Cellax increased the systemic exposure of taxanes by 37× compared to Abraxane, and improved the delivery specificity: Cellax uptake was selective to the tumor, liver and spleen, with a 203× increase in tumor accumulation compared to Abraxane. The concentration of released DTX in Cellax treated tumors was well above the IC50 for at least 10 d, while paclitaxel released from Abraxane was undetectable after 24h. In s.c. PC3 (prostate) and B16F10 (melanoma) models, Cellax exhibited enhanced efficacy and was better tolerated compared to Abraxane. In an orthotopic 4T1 breast tumor model, Cellax reduced the incidence of lung metastasis to 40% with no metastasic incidence in other tissues. Mice treated with Abraxane displayed increased lung metastasic incidence (>85%) with metastases detected in the bone, liver, spleen and kidney. These results confirm that Cellax is a more effective drug delivery strategy compared to the approved taxane nanomedicine.


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.


Cancer Cell | 2015

Inhibition of the Mitochondrial Protease ClpP as a Therapeutic Strategy for Human Acute Myeloid Leukemia

Alicia Cole; Zezhou Wang; Etienne Coyaud; Veronique Voisin; Marcela Gronda; Yulia Jitkova; Rachel Mattson; Rose Hurren; Sonja Babovic; Neil MacLean; Ian Restall; Xiaoming Wang; Danny V. Jeyaraju; Mahadeo A. Sukhai; Swayam Prabha; Shaheena Bashir; Ashwin Ramakrishnan; Elisa Leung; Yi Hua Qia; Nianxian Zhang; Kevin R. Combes; Troy Ketela; Fengshu Lin; Walid A. Houry; Ahmed Aman; Rima Al-awar; Wei Zheng; Erno Wienholds; Chang Jiang Xu; John E. Dick

From an shRNA screen, we identified ClpP as a member of the mitochondrial proteome whose knockdown reduced the viability of K562 leukemic cells. Expression of this mitochondrial protease that has structural similarity to the cytoplasmic proteosome is increased in leukemic cells from approximately half of all patients with AML. Genetic or chemical inhibition of ClpP killed cells from both human AML cell lines and primary samples in which the cells showed elevated ClpP expression but did not affect their normal counterparts. Importantly, Clpp knockout mice were viable with normal hematopoiesis. Mechanistically, we found that ClpP interacts with mitochondrial respiratory chain proteins and metabolic enzymes, and knockdown of ClpP in leukemic cells inhibited oxidative phosphorylation and mitochondrial metabolism.


ACS Medicinal Chemistry Letters | 2013

Potent Targeting of the STAT3 Protein in Brain Cancer Stem Cells: A Promising Route for Treating Glioblastoma

Sina Haftchenary; H. Artee Luchman; Andriana O. Jouk; Anthony J. Veloso; Brent D. G. Page; Xin Ran Cheng; Sean S. Dawson; Natalie Grinshtein; Vijay M. Shahani; Kagan Kerman; David R. Kaplan; Carly Griffin; Ahmed Aman; Rima Al-awar; Samuel Weiss; Patrick T. Gunning

The STAT3 gene is abnormally active in glioblastoma (GBM) and is a critically important mediator of tumor growth and therapeutic resistance in GBM. Thus, for poorly treated brain cancers such as gliomas, astrocytomas, and glioblastomas, which harbor constitutively activated STAT3, a STAT3-targeting therapeutic will be of significant importance. Herein, we report a most potent, small molecule, nonphosphorylated STAT3 inhibitor, 31 (SH-4-54) that strongly binds to STAT3 protein (K D = 300 nM). Inhibitor 31 potently kills glioblastoma brain cancer stem cells (BTSCs) and effectively suppresses STAT3 phosphorylation and its downstream transcriptional targets at low nM concentrations. Moreover, in vivo, 31 exhibited blood-brain barrier permeability, potently controlled glioma tumor growth, and inhibited pSTAT3 in vivo. This work, for the first time, demonstrates the power of STAT3 inhibitors for the treatment of BTSCs and validates the therapeutic efficacy of a STAT3 inhibitor for GBM clinical application.


Clinical Cancer Research | 2016

Disulfiram when Combined with Copper Enhances the Therapeutic Effects of Temozolomide for the Treatment of Glioblastoma.

Xueqing Lun; Wells Jc; Natalie Grinshtein; King Jc; Hao X; Dang Nh; Xin Wang; Ahmed Aman; David Uehling; Alessandro Datti; Jeff Wrana; Jay Easaw; Artee Luchman; Samuel Weiss; J. G. Cairncross; David R. Kaplan; Steve Robbins; Donna L. Senger

Purpose: Glioblastoma is one of the most lethal cancers in humans, and with existing therapy, survival remains at 14.6 months. Current barriers to successful treatment include their infiltrative behavior, extensive tumor heterogeneity, and the presence of a stem-like population of cells, termed brain tumor–initiating cells (BTIC) that confer resistance to conventional therapies. Experimental Design: To develop therapeutic strategies that target BTICs, we focused on a repurposing approach that explored already-marketed (clinically approved) drugs for therapeutic potential against patient-derived BTICs that encompass the genetic and phenotypic heterogeneity of glioblastoma observed clinically. Results: Using a high-throughput in vitro drug screen, we found that montelukast, clioquinol, and disulfiram (DSF) were cytotoxic against a large panel of patient-derived BTICs. Of these compounds, disulfiram, an off-patent drug previously used to treat alcoholism, in the presence of a copper supplement, showed low nanomolar efficacy in BTICs including those resistant to temozolomide and the highly infiltrative quiescent stem-like population. Low dose DSF-Cu significantly augmented temozolomide activity in vitro, and importantly, prolonged in vivo survival in patient-derived BTIC models established from both newly diagnosed and recurrent tumors. Moreover, we found that in addition to acting as a potent proteasome inhibitor, DSF-Cu functionally impairs DNA repair pathways and enhances the effects of DNA alkylating agents and radiation. These observations suggest that DSF-Cu inhibits proteasome activity and augments the therapeutic effects of DNA-damaging agents (temozolomide and radiation). Conclusions: DSF-Cu should be considered as an adjuvant therapy for the treatment of patients with glioblastoma in both newly diagnosed and recurrent settings. Clin Cancer Res; 22(15); 3860–75. ©2016 AACR.


Cell Metabolism | 2016

Metformin Pharmacokinetics in Mouse Tumors: Implications for Human Therapy

Ryan J.O. Dowling; Sonya Lam; Christian Bassi; Samar Mouaaz; Ahmed Aman; Taira Kiyota; Rima Al-awar; Pamela J. Goodwin; Vuk Stambolic

Metformin exhibits anticancer properties and is currently being explored as a therapeutic option for a variety of cancer types. Epidemiological studies demonstrate associations between metformin use in patients with type 2 diabetes and decreased cancer incidence and cancer-related mortality (Evans et al., 2005). Metformin also exhibits inhibitory effects on cancer cells in vitro and in mouse models (Anisimov et al., 2005; Zakikhani et al., 2006). Its mechanism of action is believed to involve both indirect (insulin-dependent) and direct (insulin-independent) effects.


Biomaterials | 2012

Amphiphilic micelles of poly(2-methyl-2-carboxytrimethylene carbonate-co-D,L- lactide)-graft-poly(ethylene glycol) for anti-cancer drug delivery to solid tumours

Karyn S Ho; Ahmed Aman; Rima Al-awar; Molly S. Shoichet

Drug delivery to solid tumours remains a challenge because both tumour physiology and drug solubility are unfavourable. Engineered materials can provide the basis for drug reformulation, incorporating active compounds and modulating their pharmacokinetic and biodistribution behaviour. To this end, we encapsulated docetaxel, a poorly soluble taxane drug, in a self-assembled polymeric nanoparticle micelle of poly(2-methyl-2-carboxytrimethylene carbonate-co-D,L-lactide)-graft-poly(ethylene glycol) (poly(TMCC-co-LA)-g-PEG). This formulation was compared with its conventional ethanolic polysorbate 80 formulation in terms of plasma circulation and biodistribution in an orthotopic mouse model of breast cancer. Notably, the polymeric nanoparticle formulation achieved greater tumour retention, resulting in prolonged exposure of cancer cells to the active drug. This behaviour was unique to the tumour tissue. The active drug was eliminated at equal or greater rates in all other tissues assayed when delivered in the polymeric nanoparticles vs. the free drug formulation. Thus, these polymeric nanoparticles are promising vehicles for solid tumour drug delivery applications, offering greater tumour exposure while eliminating the need for toxic solvents and surfactants in the dosing formulation.

Collaboration


Dive into the Ahmed Aman's collaboration.

Top Co-Authors

Avatar

Rima Al-awar

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Aaron D. Schimmer

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Rose Hurren

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Richard Marcellus

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

Marcela Gronda

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaoming Wang

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Carly Griffin

Ontario Institute for Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge