Network


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

Hotspot


Dive into the research topics where Suzanne Norval is active.

Publication


Featured researches published by Suzanne Norval.


Nature | 2012

Automated design of ligands to polypharmacological profiles

Jérémy Besnard; Gian Filippo Ruda; Vincent Setola; Keren Abecassis; Ramona M. Rodriguiz; Xi Ping Huang; Suzanne Norval; Maria F. Sassano; Antony I. Shin; Lauren A. Webster; Frederick R. C. Simeons; Laste Stojanovski; Annik Prat; Nabil G. Seidah; Daniel B. Constam; G. Richard Bickerton; Kevin D. Read; William C. Wetsel; Ian H. Gilbert; Bryan L. Roth; Andrew L. Hopkins

The clinical efficacy and safety of a drug is determined by its activity profile across many proteins in the proteome. However, designing drugs with a specific multi-target profile is both complex and difficult. Therefore methods to design drugs rationally a priori against profiles of several proteins would have immense value in drug discovery. Here we describe a new approach for the automated design of ligands against profiles of multiple drug targets. The method is demonstrated by the evolution of an approved acetylcholinesterase inhibitor drug into brain-penetrable ligands with either specific polypharmacology or exquisite selectivity profiles for G-protein-coupled receptors. Overall, 800 ligand–target predictions of prospectively designed ligands were tested experimentally, of which 75% were confirmed to be correct. We also demonstrate target engagement in vivo. The approach can be a useful source of drug leads when multi-target profiles are required to achieve either selectivity over other drug targets or a desired polypharmacology.


Nature | 2015

A novel multiple-stage antimalarial agent that inhibits protein synthesis

Beatriz Baragaña; Irene Hallyburton; Marcus C. S. Lee; Neil R. Norcross; Raffaella Grimaldi; Thomas D. Otto; William R. Proto; Andrew M. Blagborough; Stephan Meister; Grennady Wirjanata; Andrea Ruecker; Leanna M. Upton; Tara S. Abraham; Mariana Justino de Almeida; Anupam Pradhan; Achim Porzelle; María Santos Martínez; Judith M. Bolscher; Andrew Woodland; Suzanne Norval; Fabio Zuccotto; John Thomas; Frederick R. C. Simeons; Laste Stojanovski; Maria Osuna-Cabello; Patrick M. Brock; Thomas S. Churcher; Katarzyna A. Sala; Sara E. Zakutansky; María Belén Jiménez-Díaz

There is an urgent need for new drugs to treat malaria, with broad therapeutic potential and novel modes of action, to widen the scope of treatment and to overcome emerging drug resistance. Here we describe the discovery of DDD107498, a compound with a potent and novel spectrum of antimalarial activity against multiple life-cycle stages of the Plasmodium parasite, with good pharmacokinetic properties and an acceptable safety profile. DDD107498 demonstrates potential to address a variety of clinical needs, including single-dose treatment, transmission blocking and chemoprotection. DDD107498 was developed from a screening programme against blood-stage malaria parasites; its molecular target has been identified as translation elongation factor 2 (eEF2), which is responsible for the GTP-dependent translocation of the ribosome along messenger RNA, and is essential for protein synthesis. This discovery of eEF2 as a viable antimalarial drug target opens up new possibilities for drug discovery.


Science Translational Medicine | 2012

The Anti-Trypanosome Drug Fexinidazole Shows Potential for Treating Visceral Leishmaniasis

Susan Wyllie; Stephen Patterson; Laste Stojanovski; Frederick R. C. Simeons; Suzanne Norval; Robert Kime; Kevin D. Read; Alan H. Fairlamb

Fexinidazole, a drug in clinical testing for African sleeping sickness, shows potential as an oral treatment for another neglected tropical disease. A New Job for an Old Drug Fever, fatigue, weight loss, and swelling of the spleen and liver are all symptoms of visceral leishmaniasis—a tropical disease that is also known as kala-azar or black fever. Caused by the protozoan parasite Leishmania donovani, which is transmitted to people through the bite of a sand fly, the disease is almost always fatal if untreated. Although several drugs exist, they are costly and not always safe, effective, or easy to administer. To address the need for better drugs, Wyllie et al. investigated the possibility of using fexinidazole to treat visceral leishmaniasis. This antiparasitic compound, developed decades ago, is now undergoing early clinical trials as an oral therapy for African sleeping sickness, a disease that is caused by a related protozoan parasite called Trypanosoma brucei. Fexinidazole’s mode of action is thought to involve a trypanosome nitroreductase; the finding that a closely related enzyme is encoded by the leishmania genome inspired Wyllie et al. to pursue fexinidazole as a therapy for visceral leishmaniasis. They found that the compound and two of its metabolites (which rapidly form in vivo) showed activity against both developmental stages of L. donovani in vitro. The metabolites were cytotoxic, killing all the parasites within 30 hours. For unclear reasons, only the metabolites were active against L. donovani grown in macrophages (the cells in which the parasite reproduces during infection). In a mouse model of visceral leishmaniasis, a daily oral dose of fexinidazole for 5 days almost completely suppressed infection—an activity that is comparable to that of drugs currently in clinical use against this deadly tropical disease. Visceral leishmaniasis kills more people than any other parasitic disease except malaria. The clinical trials of fexinidazole for African sleeping sickness have already shown that the drug is extremely safe. The discovery that it may also be a viable oral treatment for visceral leishmaniasis bodes well for those afflicted with this disease. Safer and more effective oral drugs are required to treat visceral leishmaniasis, a parasitic disease that kills 50,000 to 60,000 people each year in parts of Asia, Africa, and Latin America. Here, we report that fexinidazole, a drug currently in phase 1 clinical trials for treating African trypanosomiasis, shows promise for treating visceral leishmaniasis. This 2-substituted 5-nitroimidazole drug is rapidly oxidized in vivo in mice, dogs, and humans to sulfoxide and sulfone metabolites. Both metabolites of fexinidazole were active against Leishmania donovani amastigotes grown in macrophages, whereas the parent compound was inactive. Pharmacokinetic studies with fexinidazole (200 mg/kg) showed that fexinidazole sulfone achieves blood concentrations in mice above the EC99 (effective concentration inhibiting growth by 99%) value for at least 24 hours after a single oral dose. A once-daily regimen for 5 days at this dose resulted in a 98.4% suppression of infection in a mouse model of visceral leishmaniasis, equivalent to that seen with the drugs miltefosine and Pentostam, which are currently used clinically to treat this tropical disease. In African trypanosomes, the mode of action of nitro drugs involves reductive activation via a NADH (reduced form of nicotinamide adenine dinucleotide)–dependent bacterial-like nitroreductase. Overexpression of the leishmanial homolog of this nitroreductase in L. donovani increased sensitivity to fexinidazole by 19-fold, indicating that a similar mechanism is involved in both parasites. These findings illustrate the potential of fexinidazole as an oral drug therapy for treating visceral leishmaniasis.


Journal of Medicinal Chemistry | 2012

Discovery of a novel class of orally active trypanocidal N-myristoyltransferase inhibitors.

Stephen Brand; Laura A. T. Cleghorn; Stuart P. McElroy; David A. Robinson; Victoria Smith; Irene Hallyburton; Justin R. Harrison; Neil R. Norcross; Daniel Spinks; Tracy Bayliss; Suzanne Norval; Laste Stojanovski; Leah S. Torrie; Julie A. Frearson; Ruth Brenk; Alan H. Fairlamb; Michael A. J. Ferguson; Kevin D. Read; Paul G. Wyatt; Ian H. Gilbert

N-Myristoyltransferase (NMT) represents a promising drug target for human African trypanosomiasis (HAT), which is caused by the parasitic protozoa Trypanosoma brucei. We report the optimization of a high throughput screening hit (1) to give a lead molecule DDD85646 (63), which has potent activity against the enzyme (IC50 = 2 nM) and T. brucei (EC50 = 2 nM) in culture. The compound has good oral pharmacokinetics and cures rodent models of peripheral HAT infection. This compound provides an excellent tool for validation of T. brucei NMT as a drug target for HAT as well as a valuable lead for further optimization.


Journal of Medicinal Chemistry | 2010

Aryl Phosphoramidates of 5-Phospho Erythronohydroxamic Acid, A New Class of Potent Trypanocidal Compounds

Gian Filippo Ruda; Pui Ee Wong; Vincent P. Alibu; Suzanne Norval; Kevin D. Read; Michael P. Barrett; Ian H. Gilbert

RNAi and enzymatic studies have shown the importance of 6-phosphogluconate dehydrogenase (6-PGDH) in Trypanosoma brucei for the parasite survival and make it an attractive drug target for the development of new treatments against human African trypanosomiasis. 2,3-O-Isopropylidene-4-erythrono hydroxamate is a potent inhibitor of parasite Trypanosoma brucei 6-phosphogluconate dehydrogenase (6-PGDH), the third enzyme of the pentose phosphate pathway. However, this compound does not have trypanocidal activity due to its poor membrane permeability. Consequently, we have previously reported a prodrug approach to improve the antiparasitic activity of this inhibitor by converting the phosphate group into a less charged phosphate prodrug. The activity of prodrugs appeared to be dependent on their stability in phosphate buffer. Here we have successfully further extended the development of the aryl phosphoramidate prodrugs of 2,3-O-isopropylidene-4-erythrono hydroxamate by synthesizing a small library of phosphoramidates and evaluating their biological activity and stability in a variety of assays. Some of the compounds showed high trypanocidal activity and good correlation of activity with their stability in fresh mouse blood.


Antimicrobial Agents and Chemotherapy | 2013

The R Enantiomer of the Antitubercular Drug PA-824 as a Potential Oral Treatment for Visceral Leishmaniasis

Stephen Patterson; Susan Wyllie; Laste Stojanovski; Meghan R. Perry; Frederick R. C. Simeons; Suzanne Norval; Maria Osuna-Cabello; Manu De Rycker; Kevin D. Read; Alan H. Fairlamb

ABSTRACT The novel nitroimidazopyran agent (S)-PA-824 has potent antibacterial activity against Mycobacterium tuberculosis in vitro and in vivo and is currently in phase II clinical trials for tuberculosis (TB). In contrast to M. tuberculosis, where (R)-PA-824 is inactive, we report here that both enantiomers of PA-824 show potent parasiticidal activity against Leishmania donovani, the causative agent of visceral leishmaniasis (VL). In leishmania-infected macrophages, (R)-PA-824 is 6-fold more active than (S)-PA-824. Both des-nitro analogues are inactive, underlining the importance of the nitro group in the mechanism of action. Although the in vitro and in vivo pharmacological profiles of the two enantiomers are similar, (R)-PA-824 is more efficacious in the murine model of VL, with >99% suppression of parasite burden when administered orally at 100 mg kg of body weight−1, twice daily for 5 days. In M. tuberculosis, (S)-PA-824 is a prodrug that is activated by a deazaflavin-dependent nitroreductase (Ddn), an enzyme which is absent in Leishmania spp. Unlike the case with nifurtimox and fexinidazole, transgenic parasites overexpressing the leishmania nitroreductase are not hypersensitive to either (R)-PA-824 or (S)-PA-824, indicating that this enzyme is not the primary target of these compounds. Drug combination studies in vitro indicate that fexinidazole and (R)-PA-824 are additive whereas (S)-PA-824 and (R)-PA-824 show mild antagonistic behavior. Thus, (R)-PA-824 is a promising candidate for late lead optimization for VL and may have potential for future use in combination therapy with fexinidazole, currently in phase II clinical trials against VL.


Cell | 2017

Development of a Novel Lead that Targets M. tuberculosis Polyketide Synthase 13.

Anup Aggarwal; Maloy K. Parai; Nishant D. Shetty; Deeann Wallis; Lisa K. Woolhiser; Courtney Hastings; Noton K. Dutta; Stacy Galaviz; Ramesh C. Dhakal; Rupesh Shrestha; Shoko Wakabayashi; Chris Walpole; David A. Matthews; David M. Floyd; Paul Scullion; Jennifer Riley; Ola Epemolu; Suzanne Norval; Thomas Snavely; Gregory T. Robertson; Eric J. Rubin; Thomas R. Ioerger; Frik A. Sirgel; Ruben Gerhard van der Merwe; Paul D. van Helden; Peter M. Keller; Erik C. Böttger; Petros C. Karakousis; Anne J. Lenaerts; James C. Sacchettini

Summary Widespread resistance to first-line TB drugs is a major problem that will likely only be resolved through the development of new drugs with novel mechanisms of action. We have used structure-guided methods to develop a lead molecule that targets the thioesterase activity of polyketide synthase Pks13, an essential enzyme that forms mycolic acids, required for the cell wall of Mycobacterium tuberculosis. Our lead, TAM16, is a benzofuran class inhibitor of Pks13 with highly potent in vitro bactericidal activity against drug-susceptible and drug-resistant clinical isolates of M. tuberculosis. In multiple mouse models of TB infection, TAM16 showed in vivo efficacy equal to the first-line TB drug isoniazid, both as a monotherapy and in combination therapy with rifampicin. TAM16 has excellent pharmacological and safety profiles, and the frequency of resistance for TAM16 is ∼100-fold lower than INH, suggesting that it can be developed as a new antitubercular aimed at the acute infection. PaperClip


Journal of Medicinal Chemistry | 2014

Lead optimization of a pyrazole sulfonamide series of Trypanosoma brucei N-myristoyltransferase inhibitors: identification and evaluation of CNS penetrant compounds as potential treatments for stage 2 human African trypanosomiasis.

Stephen Brand; Neil R. Norcross; Stephen Thompson; Justin R. Harrison; Victoria Smith; David A. Robinson; Leah S. Torrie; Stuart P. McElroy; Irene Hallyburton; Suzanne Norval; Paul Scullion; Laste Stojanovski; Frederick R. C. Simeons; Daan M. F. van Aalten; Julie A. Frearson; Ruth Brenk; Alan H. Fairlamb; Michael A. J. Ferguson; Paul G. Wyatt; Ian H. Gilbert; Kevin D. Read

Trypanosoma bruceiN-myristoyltransferase (TbNMT) is an attractive therapeutic target for the treatment of human African trypanosomiasis (HAT). From previous studies, we identified pyrazole sulfonamide, DDD85646 (1), a potent inhibitor of TbNMT. Although this compound represents an excellent lead, poor central nervous system (CNS) exposure restricts its use to the hemolymphatic form (stage 1) of the disease. With a clear clinical need for new drug treatments for HAT that address both the hemolymphatic and CNS stages of the disease, a chemistry campaign was initiated to address the shortfalls of this series. This paper describes modifications to the pyrazole sulfonamides which markedly improved blood–brain barrier permeability, achieved by reducing polar surface area and capping the sulfonamide. Moreover, replacing the core aromatic with a flexible linker significantly improved selectivity. This led to the discovery of DDD100097 (40) which demonstrated partial efficacy in a stage 2 (CNS) mouse model of HAT.


PLOS ONE | 2012

Skin-Targeted Inhibition of PPAR β/δ by Selective Antagonists to Treat PPAR β/δ – Mediated Psoriasis-Like Skin Disease In Vivo

Katrin Hack; Louise Reilly; Colin N. A. Palmer; Kevin D. Read; Suzanne Norval; Robert Kime; Kally Booth; John Foerster

We have previously shown that peroxisome proliferator activating receptor ß/δ (PPAR β/δ is overexpressed in psoriasis. PPAR β/δ is not present in adult epidermis of mice. Targeted expression of PPAR β/δ and activation by a selective synthetic agonist is sufficient to induce an inflammatory skin disease resembling psoriasis. Several signalling pathways dysregulated in psoriasis are replicated in this model, suggesting that PPAR β/δ activation contributes to psoriasis pathogenesis. Thus, inhibition of PPAR β/δ might harbour therapeutical potential. Since PPAR β/δ has pleiotropic functions in metabolism, skin-targeted inhibition offer the potential of reducing systemic adverse effects. Here, we report that three selective PPAR β/δ antagonists, GSK0660, compound 3 h, and GSK3787 can be formulated for topical application to the skin and that their skin concentration can be accurately quantified using ultra-high performance liquid chromatography (UPLC)/mass spectrometry. These antagonists show efficacy in our transgenic mouse model in reducing psoriasis – like changes triggered by activation of PPAR β/δ. PPAR β/δ antagonists GSK0660 and compound 3 do not exhibit systemic drug accumulation after prolonged application to the skin, nor do they induce inflammatory or irritant changes. Significantly, the irreversible PPAR β/δ antagonist (GSK3787) retains efficacy when applied topically only three times per week which could be of practical clinical usefulness. Our data suggest that topical inhibition of PPAR β/δ to treat psoriasis may warrant further exploration.


eLife | 2016

The anti-tubercular drug delamanid as a potential oral treatment for visceral leishmaniasis

Stephen Patterson; Susan Wyllie; Suzanne Norval; Laste Stojanovski; Frederick R. C. Simeons; Jennifer L Auer; Maria Osuna-Cabello; Kevin D. Read; Alan H. Fairlamb

There is an urgent requirement for safe, oral and cost-effective drugs for the treatment of visceral leishmaniasis (VL). We report that delamanid (OPC-67683), an approved drug for multi-drug resistant tuberculosis, is a potent inhibitor of Leishmania donovani both in vitro and in vivo. Twice-daily oral dosing of delamanid at 30 mg kg-1 for 5 days resulted in sterile cures in a mouse model of VL. Treatment with lower doses revealed a U-shaped (hormetic) dose-response curve with greater parasite suppression at 1 mg kg-1 than at 3 mg kg-1 (5 or 10 day dosing). Dosing delamanid for 10 days confirmed the hormetic dose-response and improved the efficacy at all doses investigated. Mechanistic studies reveal that delamanid is rapidly metabolised by parasites via an enzyme, distinct from the nitroreductase that activates fexinidazole. Delamanid has the potential to be repurposed as a much-needed oral therapy for VL. DOI: http://dx.doi.org/10.7554/eLife.09744.001

Collaboration


Dive into the Suzanne Norval'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