Janette Reader
University of Pretoria
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Janette Reader.
Science Translational Medicine | 2017
Tanya Paquet; Claire Le Manach; Diego Gonzàlez Cabrera; Yassir Younis; Philipp P. Henrich; Tara S. Abraham; Marcus C. S. Lee; Rajshekhar Basak; Sonja Ghidelli-Disse; Maria Jose Lafuente-Monasterio; Marcus Bantscheff; Andrea Ruecker; Andrew M. Blagborough; Sara E. Zakutansky; Anne-Marie Zeeman; Karen L. White; David M. Shackleford; Janne Mannila; Julia Morizzi; Christian Scheurer; Iñigo Angulo-Barturen; María Santos Martínez; Santiago Ferrer; Laura Sanz; Francisco Javier Gamo; Janette Reader; Mariette Botha; Koen J. Dechering; Robert W. Sauerwein; Anchalee Tungtaeng
MMV390048, a member of a new class of inhibitors of the Plasmodium phosphatidylinositol 4-kinase, shows potential for both treatment and prophylaxis. A new antimalarial in the armamentarium Paquet et al. screened a small-molecule library against the human malaria parasite, Plasmodium falciparum, and identified the 2-aminopyridine chemical class with potent activity. The optimized compound from this class, MMV390048, was active against multiple parasite life cycle stages, in both the mammalian host and the mosquito vector, and also killed drug-resistant parasites. MMV390048 killed the malaria parasite by blocking the parasite’s phosphatidylinositol 4-kinase (PI4K) and was able to protect monkeys from malaria infection. MMV390048 has potential as a new antimalarial drug that may contribute to global malaria eradication efforts. As part of the global effort toward malaria eradication, phenotypic whole-cell screening revealed the 2-aminopyridine class of small molecules as a good starting point to develop new antimalarial drugs. Stemming from this series, we found that the derivative, MMV390048, lacked cross-resistance with current drugs used to treat malaria. This compound was efficacious against all Plasmodium life cycle stages, apart from late hypnozoites in the liver. Efficacy was shown in the humanized Plasmodium falciparum mouse model, and modest reductions in mouse-to-mouse transmission were achieved in the Plasmodium berghei mouse model. Experiments in monkeys revealed the ability of MMV390048 to be used for full chemoprotection. Although MMV390048 was not able to eliminate liver hypnozoites, it delayed relapse in a Plasmodium cynomolgi monkey model. Both genomic and chemoproteomic studies identified a kinase of the Plasmodium parasite, phosphatidylinositol 4-kinase, as the molecular target of MMV390048. The ability of MMV390048 to block all life cycle stages of the malaria parasite suggests that this compound should be further developed and may contribute to malaria control and eradication as part of a single-dose combination treatment.
Journal of Medicinal Chemistry | 2015
Claire Le Manach; Tanya Paquet; Christel Brunschwig; Mathew Njoroge; Ze Han; Diego Gonzàlez Cabrera; Sridevi Bashyam; Rajkumar Dhinakaran; Dale Taylor; Janette Reader; Mariette Botha; Alisje Churchyard; Sonja B. Lauterbach; Theresa L. Coetzer; Lyn-Marie Birkholtz; Stephan Meister; Elizabeth A. Winzeler; David Waterson; Michael J. Witty; Sergio Wittlin; María-Belén Jiménez-Díaz; María Santos Martínez; Santiago Ferrer; Iñigo Angulo-Barturen; Leslie J. Street; Kelly Chibale
Toward improving pharmacokinetics, in vivo efficacy, and selectivity over hERG, structure-activity relationship studies around the central core of antimalarial imidazopyridazines were conducted. This study led to the identification of potent pyrazolopyridines, which showed good in vivo efficacy and pharmacokinetics profiles. The lead compounds also proved to be very potent in the parasite liver and gametocyte stages, which makes them of high interest.
Bioorganic & Medicinal Chemistry | 2015
Bianca K. Verlinden; Marna de Beer; Boobalan Pachaiyappan; Ethan Besaans; Warren A. Andayi; Janette Reader; Jandeli Niemand; Riëtte van Biljon; Kiplin R. Guy; Timothy J. Egan; Patrick M. Woster; Lyn-Marie Birkholtz
A new series of potent potent aryl/alkylated (bis)urea- and (bis)thiourea polyamine analogues were synthesized and evaluated in vitro for their antiplasmodial activity. Altering the carbon backbone and terminal substituents increased the potency of analogues in the compound library 3-fold, with the most active compounds, 15 and 16, showing half-maximal inhibitory concentrations (IC50 values) of 28 and 30 nM, respectively, against various Plasmodium falciparum parasite strains without any cross-resistance. In vitro evaluation of the cytotoxicity of these analogues revealed marked selectivity towards targeting malaria parasites compared to mammalian HepG2 cells (>5000-fold lower IC50 against the parasite). Preliminary biological evaluation of the polyamine analogue antiplasmodial phenotype revealed that (bis)urea compounds target parasite asexual proliferation, whereas (bis)thiourea compounds of the same series have the unique ability to block transmissible gametocyte forms of the parasite, indicating pluripharmacology against proliferative and non-proliferative forms of the parasite. In this manuscript, we describe these results and postulate a refined structure-activity relationship (SAR) model for antiplasmodial polyamine analogues. The terminally aryl/alkylated (bis)urea- and (bis)thiourea-polyamine analogues featuring a 3-5-3 or 3-6-3 carbon backbone represent a structurally novel and distinct class of potential antiplasmodials with activities in the low nanomolar range, and high selectivity against various lifecycle forms of P. falciparum parasites.
Antimicrobial Agents and Chemotherapy | 2014
Nadia Saidani; Cyrille Y. Botté; Michael Deligny; Anne-Laure Bonneau; Janette Reader; Ronald Lasselin; Goulven Merer; Alisson Niepceron; Fabien Brossier; Jean-Christophe Cintrat; Bernard Rousseau; Lyn-Marie Birkholtz; Marie-France Cesbron-Delauw; Jean-François Dubremetz; Corinne Mercier; Henri Vial; Roman Lopez; Eric Maréchal
ABSTRACT A piperidinyl-benzimidazolone scaffold has been found in the structure of different inhibitors of membrane glycerolipid metabolism, acting on enzymes manipulating diacylglycerol and phosphatidic acid. Screening a focus library of piperidinyl-benzimidazolone analogs might therefore identify compounds acting against infectious parasites. We first evaluated the in vitro effects of (S)-2-(dibenzylamino)-3-phenylpropyl 4-(1,2-dihydro-2-oxobenzo[d]imidazol-3-yl)piperidine-1-carboxylate (compound 1) on Toxoplasma gondii and Plasmodium falciparum. In T. gondii, motility and apical complex integrity appeared to be unaffected, whereas cell division was inhibited at compound 1 concentrations in the micromolar range. In P. falciparum, the proliferation of erythrocytic stages was inhibited, without any delayed death phenotype. We then explored a library of 250 analogs in two steps. We selected 114 compounds with a 50% inhibitory concentration (IC50) cutoff of 2 μM for at least one species and determined in vitro selectivity indexes (SI) based on toxicity against K-562 human cells. We identified compounds with high gains in the IC50 (in the 100 nM range) and SI (up to 1,000 to 2,000) values. Isobole analyses of two of the most active compounds against P. falciparum indicated that their interactions with artemisinin were additive. Here, we propose the use of structure-activity relationship (SAR) models, which will be useful for designing probes to identify the target compound(s) and optimizations for monotherapy or combined-therapy strategies.
Antimicrobial Agents and Chemotherapy | 2013
Jandeli Niemand; Pieter Buys Burger; Bianca K. Verlinden; Janette Reader; Annie M. Joubert; Annette Kaiser; Abraham I. Louw; Kiaran Kirk; Otto Phanstiel; Lyn-Marie Birkholtz
ABSTRACT Anthracene-polyamine conjugates inhibit the in vitro proliferation of the intraerythrocytic human malaria parasite Plasmodium falciparum, with 50% inhibitory concentrations (IC50s) in the nM to μM range. The compounds are taken up into the intraerythrocytic parasite, where they arrest the parasite cell cycle. Both the anthracene and polyamine components of the conjugates play a role in their antiplasmodial effect.
Journal of Medicinal Chemistry | 2017
Kawaljit Singh; John Okombo; Christel Brunschwig; Ferdinand Ndubi; Linley Barnard; Chad Wilkinson; Peter M. Njogu; Mathew Njoroge; Lizahn Laing; Marta Machado; Miguel Prudêncio; Janette Reader; Mariette Botha; Sindisiwe Nondaba; Lyn-Marie Birkholtz; Sonja B. Lauterbach; Alisje Churchyard; Theresa L. Coetzer; Jeremy N. Burrows; Clive Yeates; Paolo Denti; Lubbe Wiesner; Timothy J. Egan; Sergio Wittlin; Kelly Chibale
Further structure-activity relationship (SAR) studies on the recently identified pyrido[1,2-a]benzimidazole (PBI) antimalarials have led to the identification of potent, metabolically stable compounds with improved in vivo oral efficacy in the P. berghei mouse model and additional activity against parasite liver and gametocyte stages, making them potential candidates for preclinical development. Inhibition of hemozoin formation possibly contributes to the mechanism of action.
ChemMedChem | 2017
Rozanne Harmse; Dina Coertzen; Ho Ning Wong; Frans J. Smit; Mariëtte van der Watt; Janette Reader; Sindiswe H. Nondaba; Lyn-Marie Birkholtz; Richard K. Haynes; David D. N'Da
Dihydroartemisinin (DHA), either used in its own right or as the active drug generated in vivo from the other artemisinins in current clinical use—artemether and artesunate—induces quiescence in ring‐stage parasites of Plasmodium falciparum (Pf). This induction of quiescence is linked to artemisinin resistance. Thus, we have turned to structurally disparate artemisinins that are incapable of providing DHA on metabolism. Accordingly, 11‐azaartemisinin 5 and selected N‐sulfonyl derivatives were screened against intraerythrocytic asexual stages of drug‐sensitive Pf NF54 and drug‐resistant K1 and W2 parasites. Most displayed appreciable activities against all three strains, with IC50 values <10.5 nm. The p‐trifluoromethylbenzenesulfonyl‐11‐azaartemisinin derivative 11 [(4′‐trifluoromethyl)benzenesulfonylazaartemisinin] was the most active, with IC50 values between 2 and 3 nm. The compounds were screened against Pf NF54 early and transmissible late intraerythrocytic‐stage gametocytes using luciferase and parasite lactate dehydrogenase (pLDH) assays. The 2′‐thienylsulfonyl derivative 16 (2′‐thiophenesulfonylazaartemisinin) was notably active against late‐stage (IV–V) gametocytes with an IC50 value of 8.7 nm. All compounds were relatively nontoxic to human fetal lung WI‐38 fibroblasts, showing selectivity indices of >2000 toward asexual parasites. Overall, the readily accessible 11‐azaartemisinin 5 and the sulfonyl derivatives 11 and 16 represent potential candidates for further development, in particular for transmission blocking of artemisinin‐resistant parasites.
Journal of Antimicrobial Chemotherapy | 2018
Mariëtte E. van der Watt; Janette Reader; Alisje Churchyard; Sindisiwe Nondaba; Sonja B. Lauterbach; Jandeli Niemand; Sijuade Abayomi; Riette Van Biljon; Jessica I. Connacher; R.D.J. Van Wyk; Claire Le Manach; Tanya Paquet; Diego Gonzàlez Cabrera; Christel Brunschwig; Anjo Theron; Sonia Lozano-Arias; Janneth Rodrigues; Esperanza Herreros; Didier Leroy; James Duffy; Leslie J. Street; Kelly Chibale; Dalu Mancama; Theresa L. Coetzer; Lyn-Marie Birkholtz
Objectives Novel chemical tools to eliminate malaria should ideally target both the asexual parasites and transmissible gametocytes. Several imidazopyridazines (IMPs) and 2-aminopyridines (2-APs) have been described as potent antimalarial candidates targeting lipid kinases. However, these have not been extensively explored for stage-specific inhibition of gametocytes in Plasmodium falciparum parasites. Here we provide an in-depth evaluation of the gametocytocidal activity of compounds from these chemotypes and identify novel starting points for dual-acting antimalarials. Methods We evaluated compounds against P. falciparum gametocytes using several assay platforms for cross-validation and stringently identified hits that were further profiled for stage specificity, speed of action and ex vivo efficacy. Physicochemical feature extraction and chemogenomic fingerprinting were applied to explore the kinase inhibition susceptibility profile. Results We identified 34 compounds with submicromolar activity against late stage gametocytes, validated across several assay platforms. Of these, 12 were potent at <100 nM (8 were IMPs and 4 were 2-APs) and were also active against early stage gametocytes and asexual parasites, with >1000-fold selectivity towards the parasite over mammalian cells. Front-runner compounds targeted mature gametocytes within 48 h and blocked transmission to mosquitoes. The resultant chemogenomic fingerprint of parasites treated with the lead compounds revealed the importance of targeting kinases in asexual parasites and gametocytes. Conclusions This study encompasses an in-depth evaluation of the kinase inhibitor space for gametocytocidal activity. Potent lead compounds have enticing dual activities and highlight the importance of targeting the kinase superfamily in malaria elimination strategies.
Antimicrobial Agents and Chemotherapy | 2018
Dina Coertzen; Janette Reader; Mariëtte van der Watt; Sindisiwe Nondaba; Liezl Gibhard; Lubbe Wiesner; Peter J. Smith; Sarah D'Alessandro; Donatella Taramelli; Ho Ning Wong; Jan L. Du Preez; Ronald Wai Keung Wu; Lyn-Marie Birkholtz; Richard K. Haynes
The emergence of resistance toward artemisinin combination therapies (ACTs) by the malaria parasite Plasmodium falciparum has the potential to severely compromise malaria control. Therefore, the development of new artemisinins in combination with new drugs that impart activities toward both intraerythrocytic proliferative asexual and transmissible gametocyte stages, in particular, those of resistant parasites, is urgently required. ABSTRACT The emergence of resistance toward artemisinin combination therapies (ACTs) by the malaria parasite Plasmodium falciparum has the potential to severely compromise malaria control. Therefore, the development of new artemisinins in combination with new drugs that impart activities toward both intraerythrocytic proliferative asexual and transmissible gametocyte stages, in particular, those of resistant parasites, is urgently required. We define artemisinins as oxidant drugs through their ability to oxidize reduced flavin cofactors of flavin disulfide reductases critical for maintaining redox homeostasis in the malaria parasite. Here we compare the activities of 10-amino artemisinin derivatives toward the asexual and gametocyte stages of P. falciparum parasites. Of these, artemisone and artemiside inhibited asexual and gametocyte stages, particularly stage V gametocytes, in the low-nanomolar range. Further, treatment of both early and late gametocyte stages with artemisone or artemiside combined with the pro-oxidant redox partner methylene blue displayed notable synergism. These data suggest that modulation of redox homeostasis is likely an important druggable process, particularly in gametocytes, and this finding thereby enhances the prospect of using combinations of oxidant and redox drugs for malaria control.
Malaria Journal | 2014
Lyn-Marie Birkholtz; Didier Leroy; Janette Reader; Mariette Botha; Dalu Mancama; Theresa L. Coetzer
As per the 2007 Global Malaria Eradication Plan, malaria transmission blocking is seen as key to malaria elimination strategies. A highlight that has emerged from gametocytocidal assays to identify novel compounds with malaria transmission blocking ability is that, unlike asexual-based assays, greater variability in end-point readout may exist between these assays that interrogate different parasite biological functions. Drug mode-of-action is likely to be an important factor on this outcome. Such variability may be mitigated by screening compounds based on similar pharmacophores in series. One of the major concerns with the current assay platforms is their inability to be robustly used to screen variant pharmacophores accurately as the different assay principles may interrogate different biological functions. As such, compounds targeting a specific biological pathway may in extreme cases either fail in a certain assay, or by contrast, may be flagged as false positives. Taking assay platform differences into account, and relying on good intra-assay variability for each assay optimized in our laboratories, the ATP, pLDH, luciferase reporter and PrestoBlue™ assays were compared in context of a blinded MMV 10-compound set. All the assays were performed in parallel on the same gametocyte population (except for the luciferase reporter lines). The remaining parameters for each assay were all comparable. In each case, the assay was performed for 48 h of continuous drug pressure for at least three replicates. Although direct comparison of absolute inhibition values are difficult between assay platforms, similar trends were observed including comparative performance of the luciferase marker assay and the PrestoBlue™ assay for e.g. DHA and Methylene blue. Interestingly, the ATP assay could not detect any inhibitory activity for some quinoline family members and may therefore be more sensitive in indicating the inability of these compounds to inhibit gametocytes. Data from the luciferase reporter assays for these compounds indicate that the compounds are indeed more active against early stage gametocytes. The signal obtained for these compounds in the PrestoBlue™ and pLDH assays may therefore rather reflect the inability of these assays in discriminating activity of compounds against earlier stages of gametocytes, whereas the ATP assay more accurately reflect these compounds’ activity. Whilst this data is informative from a biological perspective and may provide indications of the drug mode of action, it does highlight the care that has to be taken in screening platforms where compounds may be falsely assigned activity (or lack thereof) based on a single assay.