Network


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

Hotspot


Dive into the research topics where Bruno Simoneau is active.

Publication


Featured researches published by Bruno Simoneau.


Nature | 2003

An NS3 protease inhibitor with antiviral effects in humans infected with hepatitis C virus

Daniel Lamarre; Paul C. Anderson; Murray D. Bailey; Pierre L. Beaulieu; Gordon Bolger; Pierre R. Bonneau; Michael Bös; Dale R. Cameron; Mireille Cartier; Michael G. Cordingley; Anne-Marie Faucher; Nathalie Goudreau; Stephen H. Kawai; George Kukolj; Lisette Lagacé; LaPlante; Narjes H; Poupart Ma; Jean Rancourt; Sentjens Re; St George R; Bruno Simoneau; Gerhard Steinmann; Diane Thibeault; Youla S. Tsantrizos; Weldon Sm; Chan-Loi Yong; Montse Llinas-Brunet

Hepatitis C virus (HCV) infection is a serious cause of chronic liver disease worldwide with more than 170 million infected individuals at risk of developing significant morbidity and mortality. Current interferon-based therapies are suboptimal especially in patients infected with HCV genotype 1, and they are poorly tolerated, highlighting the unmet medical need for new therapeutics. The HCV-encoded NS3 protease is essential for viral replication and has long been considered an attractive target for therapeutic intervention in HCV-infected patients. Here we identify a class of specific and potent NS3 protease inhibitors and report the evaluation of BILN 2061, a small molecule inhibitor biologically available through oral ingestion and the first of its class in human trials. Administration of BILN 2061 to patients infected with HCV genotype 1 for 2 days resulted in an impressive reduction of HCV RNA plasma levels, and established proof-of-concept in humans for an HCV NS3 protease inhibitor. Our results further illustrate the potential of the viral-enzyme-targeted drug discovery approach for the development of new HCV therapeutics.


Nature Medicine | 2002

Herpes simplex virus helicase-primase inhibitors are active in animal models of human disease

James J. Crute; Christine A. Grygon; Karl D. Hargrave; Bruno Simoneau; Anne-Marie Faucher; Gordon Bolger; Philip Kibler; Michel Liuzzi; Michael G. Cordingley

Herpes simplex virus infections are the cause of significant morbidity, and currently used therapeutics are largely based on modified nucleoside analogs that inhibit viral DNA polymerase function. To target this disease in a new way, we have identified and optimized selective thiazolylphenyl-containing inhibitors of the herpes simplex virus (HSV) helicase-primase enzyme. The most potent compounds inhibited the helicase, the primase and the DNA-dependent ATPase activities of the enzyme with IC50 (50% inhibitory concentration) values less than 100 nM. Inhibition of the enzymatic activities was through stabilization of the interaction between the helicase-primase and DNA substrates, preventing the progression through helicase or primase catalytic cycles. Helicase-primase inhibitors also prevented viral replication as demonstrated in viral growth assays. One compound, BILS 179 BS, displayed an EC50 (effective concentration inhibiting viral growth by 50%) of 27 nM against viral growth with a selectivity index greater than 2,000. Antiviral activity was also demonstrated for multiple strains of HSV, including strains resistant to nucleoside-based therapies. Most importantly, BILS 179 BS was orally active against HSV infections in murine models of HSV-1 and HSV-2 disease and more effective than acyclovir when the treatment frequency per day was reduced or when initiation of treatment was delayed up to 65 hours after infection. These studies validate the use of helicase-primase inhibitors for the treatment of acute herpesvirus infections and provide new lead compounds for optimization and design of superior anti-HSV agents.


Journal of Virology | 2005

Inhibitors of Respiratory Syncytial Virus Replication Target Cotranscriptional mRNA Guanylylation by Viral RNA-Dependent RNA Polymerase

Michel Liuzzi; Stephen W. Mason; Mireille Cartier; Carole Lawetz; Robert S. McCollum; Nathalie Dansereau; Gordon Bolger; Nicole Lapeyre; Yvon Gaudette; Lisette Lagacé; Marie-Josée Massariol; Florence Dô; Paul Whitehead; Lynne Lamarre; Erika Scouten; Josée Bordeleau; Serge Landry; Jean Rancourt; Gulrez Fazal; Bruno Simoneau

ABSTRACT Respiratory syncytial virus (RSV) is a major cause of respiratory illness in infants, immunocompromised patients, and the elderly. New antiviral agents would be important tools in the treatment of acute RSV disease. RSV encodes its own RNA-dependent RNA polymerase that is responsible for the synthesis of both genomic RNA and subgenomic mRNAs. The viral polymerase also cotranscriptionally caps and polyadenylates the RSV mRNAs at their 5′ and 3′ ends, respectively. We have previously reported the discovery of the first nonnucleoside transcriptase inhibitor of RSV polymerase through high-throughput screening. Here we report the design of inhibitors that have improved potency both in vitro and in antiviral assays and that also exhibit activity in a mouse model of RSV infection. We have isolated virus with reduced susceptibility to this class of inhibitors. The mutations conferring resistance mapped to a novel motif within the RSV L gene, which encodes the catalytic subunit of RSV polymerase. This motif is distinct from the catalytic region of the L protein and bears some similarity to the nucleotide binding domain within nucleoside diphosphate kinases. These findings lead to the hypothesis that this class of inhibitors may block synthesis of RSV mRNAs by inhibiting guanylylation of viral transcripts. We show that short transcripts produced in the presence of inhibitor in vitro do not contain a 5′ cap but, instead, are triphosphorylated, confirming this hypothesis. These inhibitors constitute useful tools for elucidating the molecular mechanism of RSV capping and represent valid leads for the development of novel anti-RSV therapeutics.


Journal of Virology | 2012

Distinct effects of two HIV-1 capsid assembly inhibitor families that bind the same site within the N-terminal domain of the viral CA protein

Christopher T. Lemke; Steve Titolo; Uta K. von Schwedler; Nathalie Goudreau; Jean‐François Mercier; Elizabeth Wardrop; Anne Marie Faucher; René Coulombe; Soma S.R. Banik; Lee Fader; Alexandre Gagnon; Stephen H. Kawai; Jean Rancourt; Martin Tremblay; Christiane Yoakim; Bruno Simoneau; Jacques Archambault; Wesley I. Sundquist; Stephen W. Mason

ABSTRACT The emergence of resistance to existing classes of antiretroviral drugs necessitates finding new HIV-1 targets for drug discovery. The viral capsid (CA) protein represents one such potential new target. CA is sufficient to form mature HIV-1 capsids in vitro, and extensive structure-function and mutational analyses of CA have shown that the proper assembly, morphology, and stability of the mature capsid core are essential for the infectivity of HIV-1 virions. Here we describe the development of an in vitro capsid assembly assay based on the association of CA-NC subunits on immobilized oligonucleotides. This assay was used to screen a compound library, yielding several different families of compounds that inhibited capsid assembly. Optimization of two chemical series, termed the benzodiazepines (BD) and the benzimidazoles (BM), resulted in compounds with potent antiviral activity against wild-type and drug-resistant HIV-1. Nuclear magnetic resonance (NMR) spectroscopic and X-ray crystallographic analyses showed that both series of inhibitors bound to the N-terminal domain of CA. These inhibitors induce the formation of a pocket that overlaps with the binding site for the previously reported CAP inhibitors but is expanded significantly by these new, more potent CA inhibitors. Virus release and electron microscopic (EM) studies showed that the BD compounds prevented virion release, whereas the BM compounds inhibited the formation of the mature capsid. Passage of virus in the presence of the inhibitors selected for resistance mutations that mapped to highly conserved residues surrounding the inhibitor binding pocket, but also to the C-terminal domain of CA. The resistance mutations selected by the two series differed, consistent with differences in their interactions within the pocket, and most also impaired virus replicative capacity. Resistance mutations had two modes of action, either directly impacting inhibitor binding affinity or apparently increasing the overall stability of the viral capsid without affecting inhibitor binding. These studies demonstrate that CA is a viable antiviral target and demonstrate that inhibitors that bind within the same site on CA can have distinct binding modes and mechanisms of action.


Bioorganic & Medicinal Chemistry Letters | 2011

Discovery of a 1,5-dihydrobenzo[b][1,4]diazepine-2,4-dione series of inhibitors of HIV-1 capsid assembly.

Lee Fader; Richard C. Bethell; Pierre R. Bonneau; Michael Bös; Yves Bousquet; Michael G. Cordingley; René Coulombe; Patrick Deroy; Anne-Marie Faucher; Alexandre Gagnon; Nathalie Goudreau; Chantal Grand-Maitre; Ingrid Guse; Oliver Hucke; Stephen H. Kawai; Jean-Eric Lacoste; Serge Landry; Christopher T. Lemke; Eric Malenfant; Stephen W. Mason; Sébastien Morin; Jeff O’Meara; Bruno Simoneau; Steve Titolo; Christiane Yoakim

The discovery of a 1,5-dihydrobenzo[b][1,4]diazepine-2,4-dione series of inhibitors of HIV-1 capsid assembly is described. Synthesis of analogs of the 1,5-dihydrobenzo[b][1,4]diazepine-2,4-dione hit established structure-activity relationships. Replacement of the enamine functionality of the hit series with either an imidazole or a pyrazole ring led to compounds that inhibited both capsid assembly and reverse transcriptase. Optimization of the bicyclic benzodiazepine scaffold to include a 3-phenyl substituent led to lead compound 48, a pure capsid assembly inhibitor with improved antiviral activity.


Bioorganic & Medicinal Chemistry Letters | 2009

Investigation on the role of the tetrazole in the binding of thiotetrazolylacetanilides with HIV-1 wild type and K103N/Y181C double mutant reverse transcriptases.

Alexandre Gagnon; Serge Landry; René Coulombe; Araz Jakalian; Ingrid Guse; Bounkham Thavonekham; Pierre R. Bonneau; Christiane Yoakim; Bruno Simoneau

The role of the tetrazole moiety in the binding of aryl thiotetrazolylacetanilides with HIV-1 wild type and K103N/Y181C double mutant reverse transcriptases was explored. Different acyclic, cyclic and heterocyclic replacements were investigated in order to evaluate the conformational and electronic contribution of the tetrazole ring to the binding of the inhibitors in the NNRTI pocket. The replacement of the tetrazole by a pyrazolyl group led to reversal of selectivity, providing inhibitors with excellent potency against the double mutant reverse transcriptase.


Tetrahedron Letters | 1991

Stereoselective rardical-mediated reduction and alkylation of α-halo esters

Yvan Guindon; J.-F. Lavallée; L. Boisvert; C. Chabot; D. Delorme; Christiane Yoakim; D. Hall; R. Lemieux; Bruno Simoneau

Abstract Radical-mediated reduction and alkylation of β-alkoxy-α-halo esters could be achieved with good stereoselection at low temperature.


Antimicrobial Agents and Chemotherapy | 2014

Preclinical Profile of BI 224436, a Novel HIV-1 Non-Catalytic Site Integrase Inhibitor

Craig Fenwick; Ma’an Amad; Murray D. Bailey; Richard C. Bethell; Michael Bös; Pierre R. Bonneau; Michael G. Cordingley; René Coulombe; Jianmin Duan; Paul Edwards; Lee Fader; Anne-Marie Faucher; Michel Garneau; Araz Jakalian; Stephen H. Kawai; Louie Lamorte; Steven R. LaPlante; Laibin Luo; Steve Mason; Marc-André Poupart; Nathalie Rioux; Patricia Schroeder; Bruno Simoneau; Sonia Tremblay; Youla S. Tsantrizos; Myriam Witvrouw; Christiane Yoakim

ABSTRACT BI 224436 is an HIV-1 integrase inhibitor with effective antiviral activity that acts through a mechanism that is distinct from that of integrase strand transfer inhibitors (INSTIs). This 3-quinolineacetic acid derivative series was identified using an enzymatic integrase long terminal repeat (LTR) DNA 3′-processing assay. A combination of medicinal chemistry, parallel synthesis, and structure-guided drug design led to the identification of BI 224436 as a candidate for preclinical profiling. It has antiviral 50% effective concentrations (EC50s) of <15 nM against different HIV-1 laboratory strains and cellular cytotoxicity of >90 μM. BI 224436 also has a low, ∼2.1-fold decrease in antiviral potency in the presence of 50% human serum and, by virtue of a steep dose-response curve slope, exhibits serum-shifted EC95 values ranging between 22 and 75 nM. Passage of virus in the presence of inhibitor selected for either A128T, A128N, or L102F primary resistance substitutions, all mapping to a conserved allosteric pocket on the catalytic core of integrase. BI 224436 also retains full antiviral activity against recombinant viruses encoding INSTI resistance substitutions N155S, Q148H, and E92Q. In drug combination studies performed in cellular antiviral assays, BI 224436 displays an additive effect in combination with most approved antiretrovirals, including INSTIs. BI 224436 has drug-like in vitro absorption, distribution, metabolism, and excretion (ADME) properties, including Caco-2 cell permeability, solubility, and low cytochrome P450 inhibition. It exhibited excellent pharmacokinetic profiles in rat (clearance as a percentage of hepatic flow [CL], 0.7%; bioavailability [F], 54%), monkey (CL, 23%; F, 82%), and dog (CL, 8%; F, 81%). Based on the excellent biological and pharmacokinetic profile, BI 224436 was advanced into phase 1 clinical trials.


Bioorganic & Medicinal Chemistry | 1999

Discovery of non-peptidic P2-P3 butanediamide renin inhibitors with high oral efficacy.

Bruno Simoneau; Pierre Lavallee; Paul C. Anderson; Murray D. Bailey; Gary W. Bantle; Sylvie Berthiaume; Catherine Chabot; Gulrez Fazal; Ted Halmos; William W. Ogilvie; Marc-André Poupart; Bounkham Thavonekham; Zhili Xin; Diane Thibeault; Gordon Bolger; Maret Panzenbeck; Raymond J. Winquist; Grace Jung

A new series of non-peptidic renin inhibitors having a 2-substituted butanediamide moiety at the P2 and P3 positions has been identified. The optimized inhibitors have IC50 values of 0.8 to 1.4 nM and 2.5 to 7.6 nM in plasma renin assays at pH 6.0 and 7.4, respectively. When evaluated in the normotensive cynomolgus monkey model, two of the most potent inhibitors were orally active at a dose as low as 3 mg/kg. These potent renin inhibitors are characterized by oral bioavailabilities of 40 and 89% in the cynomolgus monkey. Inhibitor 3z (BILA 2157 BS) was selected as candidate for pre-development.


Antimicrobial Agents and Chemotherapy | 2003

Oral Bioavailability and In Vivo Efficacy of the Helicase-Primase Inhibitor BILS 45 BS against Acyclovir-Resistant Herpes Simplex Virus Type 1

Jianmin Duan; Michel Liuzzi; William Paris; Francine Liard; Abigail Browne; Nathalie Dansereau; Bruno Simoneau; Anne-Marie Faucher; Michael G. Cordingley

ABSTRACT This study investigated the oral bioavailability and efficacy of BILS 45 BS, a selective herpes simplex virus (HSV) helicase-primase inhibitor, against acyclovir (ACV)-resistant (ACVr) infections mediated by the HSV type 1 (HSV-1) dlsptk and PAAr5 mutant strains. In vitro, the compound was more potent than ACV against wild-type clinical and laboratory HSV-1 strains and ACVr HSV isolates, as determined by a standard plaque reduction assay, with a mean 50% effective concentration of about 0.15 μM. The oral bioavailability of BILS 45 BS in hairless mice was 49%, with a peak concentration in plasma of 31.5 μM after administration of a single dose of 25 mg/kg. Following cutaneous infection of nude mice, both the HSV-1 dlsptk and PAAr5 mutant strains induced significant, reproducible, and persistent cutaneous lesions that lasted for more than 2 weeks. Oral treatment with ACV (100 or 125 mg/kg/day, three times a day by gavage) did not affect either mutant-induced infection. In contrast, BILS 45 BS at an oral dose of 100 mg/kg/day almost completely abolished cutaneous lesions mediated by both ACVr HSV-1 mutants. The 50% effective doses of BILS 45 BS were 56.7 and 61 mg/kg/day against dlsptk- and PAAr5-induced infections, respectively. Taken together, our results demonstrate very effective oral therapy of experimental ACVr HSV-1 infections in nude mice and support the potential use of HSV helicase-primase inhibitors for the treatment of nucleoside-resistant HSV disease in humans.

Collaboration


Dive into the Bruno Simoneau'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge