Network


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

Hotspot


Dive into the research topics where Frederick R. C. Simeons is active.

Publication


Featured researches published by Frederick R. C. Simeons.


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.


The EMBO Journal | 2013

PDK1 regulates VDJ recombination, cell-cycle exit and survival during B-cell development

Ram K C Venigalla; Victoria A. McGuire; Rosemary G. Clarke; Janet C. Patterson-Kane; Ayaz Najafov; Rachel Toth; Pierre C McCarthy; Frederick R. C. Simeons; Laste Stojanovski; J. Simon C. Arthur

Phosphoinositide‐dependent kinase‐1 (PDK1) controls the activation of a subset of AGC kinases. Using a conditional knockout of PDK1 in haematopoietic cells, we demonstrate that PDK1 is essential for B cell development. B‐cell progenitors lacking PDK1 arrested at the transition of pro‐B to pre‐B cells, due to a cell autonomous defect. Loss of PDK1 decreased the expression of the IgH chain in pro‐B cells due to impaired recombination of the IgH distal variable segments, a process coordinated by the transcription factor Pax5. The expression of Pax5 in pre‐B cells was decreased in PDK1 knockouts, which correlated with reduced expression of the Pax5 target genes IRF4, IRF8 and Aiolos. As a result, Ccnd3 is upregulated in PDK1 knockout pre‐B cells and they have an impaired ability to undergo cell‐cycle arrest, a necessary event for Ig light chain rearrangement. Instead, these cells underwent apoptosis that correlated with diminished expression of the pro‐survival gene Bcl2A1. Reintroduction of both Pax5 and Bcl2A1 together into PDK1 knockout pro‐B cells restored their ability to differentiate in vitro into mature B cells.


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.


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.


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


Journal of Medicinal Chemistry | 2016

Trisubstituted Pyrimidines as Efficacious and Fast-Acting Antimalarials

Neil R. Norcross; Beatriz Baragaña; Caroline Wilson; Irene Hallyburton; Maria Osuna-Cabello; Suzanne Norval; Jennifer Riley; Laste Stojanovski; Frederick R. C. Simeons; Achim Porzelle; Raffaella Grimaldi; Sergio Wittlin; Sandra Duffy; Vicky M. Avery; Stephan Meister; Laura Sanz; Belén Jiménez-Díaz; Iñigo Angulo-Barturen; Santiago Ferrer; María Santos Martínez; Francisco Javier Gamo; Julie A. Frearson; David W. Gray; Alan H. Fairlamb; Elizabeth A. Winzeler; David Waterson; Simon F. Campbell; Paul Willis; Kevin D. Read; Ian H. Gilbert

In this paper we describe the optimization of a phenotypic hit against Plasmodium falciparum, based on a trisubstituted pyrimidine scaffold. This led to compounds with good pharmacokinetics and oral activity in a P. berghei mouse model of malaria. The most promising compound (13) showed a reduction in parasitemia of 96% when dosed at 30 mg/kg orally once a day for 4 days in the P. berghei mouse model of malaria. It also demonstrated a rapid rate of clearance of the erythrocytic stage of P. falciparum in the SCID mouse model with an ED90 of 11.7 mg/kg when dosed orally. Unfortunately, the compound is a potent inhibitor of cytochrome P450 enzymes, probably due to a 4-pyridyl substituent. Nevertheless, this is a lead molecule with a potentially useful antimalarial profile, which could either be further optimized or be used for target hunting.


Nature | 2018

Cyclin-dependent kinase 12 is a drug target for visceral leishmaniasis

Susan Wyllie; Michael George Thomas; Stephen Patterson; Sabrinia Crouch; Manu De Rycker; Rhiannon Lowe; Stephanie Gresham; Michael D. Urbaniak; Thomas D. Otto; Laste Stojanovski; Frederick R. C. Simeons; Sujatha Manthri; Lorna MacLean; Fabio Zuccotto; Nadine Homeyer; Hannah Pflaumer; Markus Boesche; Lalitha Sastry; Paul Connolly; Sebastian Albrecht; Matthew Berriman; Gerard Drewes; David W. Gray; Sonja Ghidelli-Disse; Susan Dixon; Jose M. Fiandor; Paul G. Wyatt; Michael A. J. Ferguson; Alan H. Fairlamb; Timothy James Miles

Visceral leishmaniasis causes considerable mortality and morbidity in many parts of the world. There is an urgent need for the development of new, effective treatments for this disease. Here we describe the development of an anti-leishmanial drug-like chemical series based on a pyrazolopyrimidine scaffold. The leading compound from this series (7, DDD853651/GSK3186899) is efficacious in a mouse model of visceral leishmaniasis, has suitable physicochemical, pharmacokinetic and toxicological properties for further development, and has been declared a preclinical candidate. Detailed mode-of-action studies indicate that compounds from this series act principally by inhibiting the parasite cdc-2-related kinase 12 (CRK12), thus defining a druggable target for visceral leishmaniasis.A series of compounds are discovered for the treatment of visceral leishmaniasis, and cdc2-related kinase 12 (CRK12) is identified as the probable primary drug target.


ACS Infectious Diseases | 2017

Chemical Validation of Methionyl-tRNA Synthetase as a Druggable Target in Leishmania donovani

Leah S. Torrie; Stephen Brand; David A. Robinson; Eun Jung Ko; Laste Stojanovski; Frederick R. C. Simeons; Susan Wyllie; John Thomas; Lucy Ellis; Maria Osuna-Cabello; Ola Epemolu; Andrea Nühs; Jennifer Riley; Lorna MacLean; Sujatha Manthri; Kevin D. Read; Ian H. Gilbert; Alan H. Fairlamb; Manu De Rycker

Methionyl-tRNA synthetase (MetRS) has been chemically validated as a drug target in the kinetoplastid parasite Trypanosoma brucei. In the present study, we investigate the validity of this target in the related trypanosomatid Leishmania donovani. Following development of a robust high-throughput compatible biochemical assay, a compound screen identified DDD806905 as a highly potent inhibitor of LdMetRS (Ki of 18 nM). Crystallography revealed this compound binds to the methionine pocket of MetRS with enzymatic studies confirming DDD806905 displays competitive inhibition with respect to methionine and mixed inhibition with respect to ATP binding. DDD806905 showed activity, albeit with different levels of potency, in various Leishmania cell-based viability assays, with on-target activity observed in both Leishmania promastigote cell assays and a Leishmania tarentolae in vitro translation assay. Unfortunately, this compound failed to show efficacy in an animal model of leishmaniasis. We investigated the potential causes for the discrepancies in activity observed in different Leishmania cell assays and the lack of efficacy in the animal model and found that high protein binding as well as sequestration of this dibasic compound into acidic compartments may play a role. Despite medicinal chemistry efforts to address the dibasic nature of DDD806905 and analogues, no progress could be achieved with the current chemical series. Although DDD806905 is not a developable antileishmanial compound, MetRS remains an attractive antileishmanial drug target.

Collaboration


Dive into the Frederick R. C. Simeons'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