Network


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

Hotspot


Dive into the research topics where Boni F. Sebayang is active.

Publication


Featured researches published by Boni F. Sebayang.


Science Translational Medicine | 2013

Quinolone-3-Diarylethers: A New Class of Antimalarial Drug

Aaron Nilsen; Alexis N. LaCrue; Karen L. White; Isaac P. Forquer; R. Matthew Cross; Jutta Marfurt; Michael W. Mather; Michael J. Delves; David M. Shackleford; Fabián E. Sáenz; Joanne M. Morrisey; Jessica Steuten; Tina Mutka; Yuexin Li; Grennady Wirjanata; Eileen Ryan; Sandra Duffy; Jane Xu Kelly; Boni F. Sebayang; Anne-Marie Zeeman; Rintis Noviyanti; Robert E. Sinden; Clemens H. M. Kocken; Ric N. Price; Vicky M. Avery; Iñigo Angulo-Barturen; María Belén Jiménez-Díaz; Santiago Ferrer; Esperanza Herreros; Laura Sanz

ELQ-300, an investigational drug for treating and preventing malaria, shows potent transmission-blocking activity in rodent models of malaria. Taking the Bite Out of Malaria Malaria is spread from person to person by mosquitoes that inject 8 to 10 sporozoite forms of the parasite in a single bite. The sporozoites reproduce in the liver to produce 10,000 to 30,000 merozoites before the liver schizont ruptures and parasites flood into the bloodstream where the absolute parasite burden may increase to a thousand billion (1012) circulating parasites. Some of these parasites develop into gametocytes that may be ingested by another mosquito where they progress through ookinete, oocyst, and sporozoite stages to complete the cycle. Like quinine, most antimalarial drugs in use today target only the symptomatic blood stage. The efficacy of these drugs has been compromised by resistance, and so there is a pressing need for new drugs that target multiple stages of the parasite life cycle for use in malaria treatment and prevention. Clearly, it is advantageous to strike at the liver stage where parasite numbers are low, to diminish the likelihood of selecting for a resistant mutant and before the infection has a chance to weaken the defenses of the human host. In a new study, Nilsen and colleagues describe ELQ-300, a 4(1H)-quinolone-3-diarylether, which targets the liver and blood stages, including the forms that are crucial to disease transmission (gametocytes, zygotes, and ookinetes). In mouse models of malaria, a single oral dose of 0.03 mg/kg prevented sporozoite-induced infections, whereas four daily doses of 1 mg/kg achieved complete cures of patent infections. ELQ-300 is a preclinical candidate that may be coformulated with other antimalarials to prevent and treat malaria, with the potential to aid in eradication of the disease. The goal for developing new antimalarial drugs is to find a molecule that can target multiple stages of the parasite’s life cycle, thus impacting prevention, treatment, and transmission of the disease. The 4(1H)-quinolone-3-diarylethers are selective potent inhibitors of the parasite’s mitochondrial cytochrome bc1 complex. These compounds are highly active against the human malaria parasites Plasmodium falciparum and Plasmodium vivax. They target both the liver and blood stages of the parasite as well as the forms that are crucial for disease transmission, that is, the gametocytes, the zygote, the ookinete, and the oocyst. Selected as a preclinical candidate, ELQ-300 has good oral bioavailability at efficacious doses in mice, is metabolically stable, and is highly active in blocking transmission in rodent models of malaria. Given its predicted low dose in patients and its predicted long half-life, ELQ-300 has potential as a new drug for the treatment, prevention, and, ultimately, eradication of human malaria.


Antimicrobial Agents and Chemotherapy | 2014

KAF156 Is an Antimalarial Clinical Candidate with Potential for Use in Prophylaxis, Treatment, and Prevention of Disease Transmission

Kelli Kuhen; Arnab K. Chatterjee; Matthias Rottmann; Kerstin Gagaring; Rachel Borboa; Jennifer Buenviaje; Zhong Chen; Carolyn Francek; Tao Wu; Advait Nagle; S. Whitney Barnes; David Plouffe; Marcus C. S. Lee; David A. Fidock; Wouter Graumans; Marga van de Vegte-Bolmer; Geert Jan van Gemert; Grennady Wirjanata; Boni F. Sebayang; Jutta Marfurt; Bruce Russell; Rossarin Suwanarusk; Ric N. Price; François Nosten; Anchalee Tungtaeng; Montip Gettayacamin; Jetsumon Sattabongkot; Jennifer Taylor; John R. Walker; David C. Tully

ABSTRACT Renewed global efforts toward malaria eradication have highlighted the need for novel antimalarial agents with activity against multiple stages of the parasite life cycle. We have previously reported the discovery of a novel class of antimalarial compounds in the imidazolopiperazine series that have activity in the prevention and treatment of blood stage infection in a mouse model of malaria. Consistent with the previously reported activity profile of this series, the clinical candidate KAF156 shows blood schizonticidal activity with 50% inhibitory concentrations of 6 to 17.4 nM against P. falciparum drug-sensitive and drug-resistant strains, as well as potent therapeutic activity in a mouse models of malaria with 50, 90, and 99% effective doses of 0.6, 0.9, and 1.4 mg/kg, respectively. When administered prophylactically in a sporozoite challenge mouse model, KAF156 is completely protective as a single oral dose of 10 mg/kg. Finally, KAF156 displays potent Plasmodium transmission blocking activities both in vitro and in vivo. Collectively, our data suggest that KAF156, currently under evaluation in clinical trials, has the potential to treat, prevent, and block the transmission of malaria.


Nature Communications | 2014

Pyrazoleamide compounds are potent antimalarials that target Na+ homeostasis in intraerythrocytic Plasmodium falciparum

Akhil B. Vaidya; Joanne M. Morrisey; Zhongsheng Zhang; Sudipta Das; Thomas M. Daly; Thomas D. Otto; Natalie J. Spillman; Matthew Wyvratt; Peter Siegl; Jutta Marfurt; Grennady Wirjanata; Boni F. Sebayang; Ric N. Price; Arnab K. Chatterjee; Advait Nagle; Marcin Stasiak; Susan A. Charman; Iñigo Angulo-Barturen; Santiago Ferrer; María Belén Jiménez-Díaz; María Santos Martínez; Francisco Javier Gamo; Vicky M. Avery; Andrea Ruecker; Michael J. Delves; Kiaran Kirk; Matthew Berriman; Jeremy N. Burrows; Erkang Fan; Lawrence W. Bergman

The quest for new antimalarial drugs, especially those with novel modes of action, is essential in the face of emerging drug-resistant parasites. Here we describe a new chemical class of molecules, pyrazoleamides, with potent activity against human malaria parasites and showing remarkably rapid parasite clearance in an in vivo model. Investigations involving pyrazoleamide-resistant parasites, whole-genome sequencing and gene transfers reveal that mutations in two proteins, a calcium-dependent protein kinase (PfCDPK5) and a P-type cation-ATPase (PfATP4), are necessary to impart full resistance to these compounds. A pyrazoleamide compound causes a rapid disruption of Na+ regulation in blood-stage Plasmodium falciparum parasites. Similar effect on Na+ homeostasis was recently reported for spiroindolones, which are antimalarials of a chemical class quite distinct from pyrazoleamides. Our results reveal that disruption of Na+ homeostasis in malaria parasites is a promising mode of antimalarial action mediated by at least two distinct chemical classes.


PLOS ONE | 2013

Effective Preparation of Plasmodium vivax Field Isolates for High-Throughput Whole Genome Sequencing

Sarah Auburn; Jutta Marfurt; Gareth Maslen; Susana Campino; Valentin Ruano Rubio; Magnus Manske; Barbara Machunter; Rintis Noviyanti; Leily Trianty; Boni F. Sebayang; Grennady Wirjanata; Kanlaya Sriprawat; Daniel Alcock; Bronwyn MacInnis; Olivo Miotto; Taane G. Clark; Bruce Russell; Nicholas M. Anstey; François Nosten; Dominic P. Kwiatkowski; Ric N. Price

Whole genome sequencing (WGS) of Plasmodium vivax is problematic due to the reliance on clinical isolates which are generally low in parasitaemia and sample volume. Furthermore, clinical isolates contain a significant contaminating background of host DNA which confounds efforts to map short read sequence of the target P. vivax DNA. Here, we discuss a methodology to significantly improve the success of P. vivax WGS on natural (non-adapted) patient isolates. Using 37 patient isolates from Indonesia, Thailand, and travellers, we assessed the application of CF11-based white blood cell filtration alone and in combination with short term ex vivo schizont maturation. Although CF11 filtration reduced human DNA contamination in 8 Indonesian isolates tested, additional short-term culture increased the P. vivax DNA yield from a median of 0.15 to 6.2 ng µl−1 packed red blood cells (pRBCs) (p = 0.001) and reduced the human DNA percentage from a median of 33.9% to 6.22% (p = 0.008). Furthermore, post-CF11 and culture samples from Thailand gave a median P. vivax DNA yield of 2.34 ng µl−1 pRBCs, and 2.65% human DNA. In 22 P. vivax patient isolates prepared with the 2-step method, we demonstrate high depth (median 654X coverage) and breadth (≥89%) of coverage on the Illumina GAII and HiSeq platforms. In contrast to the A+T-rich P. falciparum genome, negligible bias was observed in coverage depth between coding and non-coding regions of the P. vivax genome. This uniform coverage will greatly facilitate the detection of SNPs and copy number variants across the genome, enabling unbiased exploration of the natural diversity in P. vivax populations.


Antimicrobial Agents and Chemotherapy | 2012

Comparative Ex Vivo Activity of Novel Endoperoxides in Multidrug-Resistant Plasmodium falciparum and P. vivax

Jutta Marfurt; Ferryanto Chalfein; Pak Prayoga; Frans Wabiser; Grennady Wirjanata; Boni F. Sebayang; Kim A. Piera; Sergio Wittlin; Richard K. Haynes; Jörg J. Möhrle; Nicholas M. Anstey; Ric N. Price

ABSTRACT The declining efficacy of artemisinin derivatives against Plasmodium falciparum highlights the urgent need to identify alternative highly potent compounds for the treatment of malaria. In Papua Indonesia, where multidrug resistance has been documented against both P. falciparum and P. vivax malaria, comparative ex vivo antimalarial activity against Plasmodium isolates was assessed for the artemisinin derivatives artesunate (AS) and dihydroartemisinin (DHA), the synthetic peroxides OZ277 and OZ439, the semisynthetic 10-alkylaminoartemisinin derivatives artemisone and artemiside, and the conventional antimalarial drugs chloroquine (CQ), amodiaquine (AQ), and piperaquine (PIP). Ex vivo drug susceptibility was assessed in 46 field isolates (25 P. falciparum and 21 P. vivax). The novel endoperoxide compounds exhibited potent ex vivo activity against both species, but significant differences in intrinsic activity were observed. Compared to AS and its active metabolite DHA, all the novel compounds showed lower or equal 50% inhibitory concentrations (IC50s) in both species (median IC50s between 1.9 and 3.6 nM in P. falciparum and 0.7 and 4.6 nM in P. vivax). The antiplasmodial activity of novel endoperoxides showed different cross-susceptibility patterns in the two Plasmodium species: whereas their ex vivo activity correlated positively with CQ, PIP, AS, and DHA in P. falciparum, the same was not apparent in P. vivax. The current study demonstrates for the first time potent activity of novel endoperoxides against drug-resistant P. vivax. The high activity against drug-resistant strains of both Plasmodium species confirms these compounds to be promising candidates for future artemisinin-based combination therapy (ACT) regimens in regions of coendemicity.


Antimicrobial Agents and Chemotherapy | 2015

Potent Ex Vivo Activity of Naphthoquine and Methylene Blue against Drug-Resistant Clinical Isolates of Plasmodium falciparum and Plasmodium vivax

Grennady Wirjanata; Boni F. Sebayang; Ferryanto Chalfein; Prayoga; Irene Handayuni; Leily Trianty; Rintis Noviyanti; Brice Campo; Jeanne Rini Poespoprodjo; Jörg J. Möhrle; Ric N. Price; Jutta Marfurt

ABSTRACT The 4-aminoquinoline naphthoquine (NQ) and the thiazine dye methylene blue (MB) have potent in vitro efficacies against Plasmodium falciparum, but susceptibility data for P. vivax are limited. The species- and stage-specific ex vivo activities of NQ and MB were assessed using a modified schizont maturation assay on clinical field isolates from Papua, Indonesia, where multidrug-resistant P. falciparum and P. vivax are prevalent. Both compounds were highly active against P. falciparum (median [range] 50% inhibitory concentration [IC50]: NQ, 8.0 nM [2.6 to 71.8 nM]; and MB, 1.6 nM [0.2 to 7.0 nM]) and P. vivax (NQ, 7.8 nM [1.5 to 34.2 nM]; and MB, 1.2 nM [0.4 to 4.3 nM]). Stage-specific drug susceptibility assays revealed significantly greater IC50s in parasites exposed at the trophozoite stage than at the ring stage for NQ in P. falciparum (26.5 versus 5.1 nM, P = 0.021) and P. vivax (341.6 versus 6.5 nM, P = 0.021) and for MB in P. vivax (10.1 versus 1.6 nM, P = 0.010). The excellent ex vivo activities of NQ and MB against both P. falciparum and P. vivax highlight their potential utility for the treatment of multidrug-resistant malaria in areas where both species are endemic.


Antimicrobial Agents and Chemotherapy | 2015

Contrasting Ex Vivo Efficacies of “Reversed Chloroquine” Compounds in Chloroquine-Resistant Plasmodium falciparum and P. vivax Isolates

Grennady Wirjanata; Boni F. Sebayang; Ferryanto Chalfein; Prayoga; Irene Handayuni; Rintis Noviyanti; Jeanne Rini Poespoprodjo; Steven J. Burgess; David H. Peyton; Ric N. Price; Jutta Marfurt

ABSTRACT Chloroquine (CQ) has been the mainstay of malaria treatment for more than 60 years. However, the emergence and spread of CQ resistance now restrict its use to only a few areas where malaria is endemic. The aim of the present study was to investigate whether a novel combination of a CQ-like moiety and an imipramine-like pharmacophore can reverse CQ resistance ex vivo. Between March to October 2011 and January to September 2013, two “reversed chloroquine” (RCQ) compounds (PL69 and PL106) were tested against multidrug-resistant field isolates of Plasmodium falciparum (n = 41) and Plasmodium vivax (n = 45) in Papua, Indonesia, using a modified ex vivo schizont maturation assay. The RCQ compounds showed high efficacy against both CQ-resistant P. falciparum and P. vivax field isolates. For P. falciparum, the median 50% inhibitory concentrations (IC50s) were 23.2 nM for PL69 and 26.6 nM for PL106, compared to 79.4 nM for unmodified CQ (P < 0.001 and P = 0.036, respectively). The corresponding values for P. vivax were 19.0, 60.0, and 60.9 nM (P < 0.001 and P = 0.018, respectively). There was a significant correlation between IC50s of CQ and PL69 (Spearmans rank correlation coefficient [rs] = 0.727, P < 0.001) and PL106 (rs = 0.830, P < 0.001) in P. vivax but not in P. falciparum. Both RCQs were equally active against the ring and trophozoite stages of P. falciparum, but in P. vivax, PL69 and PL106 showed less potent activity against trophozoite stages (median IC50s, 130.2 and 172.5 nM) compared to ring stages (median IC50s, 17.6 and 91.3 nM). RCQ compounds have enhanced ex vivo activity against CQ-resistant clinical isolates of P. falciparum and P. vivax, suggesting the potential use of reversal agents in antimalarial drug development. Interspecies differences in RCQ compound activity may indicate differences in CQ pharmacokinetics between the two Plasmodium species.


PLOS Biology | 2018

The Plasmodium falciparum transcriptome in severe malaria reveals altered expression of genes involved in important processes including surface antigen–encoding var genes

Gerry Tonkin-Hill; Leily Trianty; Rintis Noviyanti; Hanh H. T. Nguyen; Boni F. Sebayang; Daniel A. Lampah; Jutta Marfurt; Simon A. Cobbold; Janavi S. Rambhatla; Malcolm J. McConville; Stephen J. Rogerson; Graham V. Brown; Karen P. Day; Ric N. Price; Nicholas M. Anstey; Anthony T. Papenfuss; Michael F. Duffy

Within the human host, the malaria parasite Plasmodium falciparum is exposed to multiple selection pressures. The host environment changes dramatically in severe malaria, but the extent to which the parasite responds to—or is selected by—this environment remains unclear. From previous studies, the parasites that cause severe malaria appear to increase expression of a restricted but poorly defined subset of the PfEMP1 variant, surface antigens. PfEMP1s are major targets of protective immunity. Here, we used RNA sequencing (RNAseq) to analyse gene expression in 44 parasite isolates that caused severe and uncomplicated malaria in Papuan patients. The transcriptomes of 19 parasite isolates associated with severe malaria indicated that these parasites had decreased glycolysis without activation of compensatory pathways; altered chromatin structure and probably transcriptional regulation through decreased histone methylation; reduced surface expression of PfEMP1; and down-regulated expression of multiple chaperone proteins. Our RNAseq also identified novel associations between disease severity and PfEMP1 transcripts, domains, and smaller sequence segments and also confirmed all previously reported associations between expressed PfEMP1 sequences and severe disease. These findings will inform efforts to identify vaccine targets for severe malaria and also indicate how parasites adapt to—or are selected by—the host environment in severe malaria.


Malaria Journal | 2016

Differences in PfEMP1s recognized by antibodies from patients with uncomplicated or severe malaria

Michael F. Duffy; Rintis Noviyanti; Takafumi Tsuboi; Zhi-Ping Feng; Leily Trianty; Boni F. Sebayang; Eizo Takashima; Fransisca Sumardy; Daniel A. Lampah; Louise Turner; Thomas Lavstsen; Freya J. I. Fowkes; Peter Siba; Stephen J. Rogerson; Thor G. Theander; Jutta Marfurt; Ric N. Price; Nicholas M. Anstey; Graham V. Brown; Anthony T. Papenfuss

BackgroundPlasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) variants are encoded by var genes and mediate pathogenic cytoadhesion and antigenic variation in malaria. PfEMP1s can be broadly divided into three principal groups (A, B and C) and they contain conserved arrangements of functional domains called domain cassettes. Despite their tremendous diversity there is compelling evidence that a restricted subset of PfEMP1s is expressed in severe disease. In this study antibodies from patients with severe and uncomplicated malaria were compared for differences in reactivity with a range of PfEMP1s to determine whether antibodies to particular PfEMP1 domains were associated with severe or uncomplicated malaria.MethodsParts of expressed var genes in a severe malaria patient were identified by RNAseq and several of these partial PfEMP1 domains were expressed together with others from laboratory isolates. Antibodies from Papuan patients to these parts of multiple PfEMP1 proteins were measured.ResultsPatients with uncomplicated malaria were more likely to have antibodies that recognized PfEMP1 of Group C type and recognized a broader repertoire of group A and B PfEMP1s than patients with severe malaria.ConclusionThese data suggest that exposure to a broad range of group A and B PfEMP1s is associated with protection from severe disease in Papua, Indonesia.


Malaria Journal | 2015

Quantification of Plasmodium ex vivo drug susceptibility by flow cytometry

Grennady Wirjanata; Irene Handayuni; Pak Prayoga; Dwi Apriyanti; Ferryanto Chalfein; Boni F. Sebayang; Steven Kho; Rintis Noviyanti; Brice Campo; Jeanne Rini Poespoprodjo; Ric N. Price; Jutta Marfurt

Collaboration


Dive into the Boni F. Sebayang's collaboration.

Top Co-Authors

Avatar

Jutta Marfurt

Charles Darwin University

View shared research outputs
Top Co-Authors

Avatar

Ric N. Price

Charles Darwin University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rintis Noviyanti

Eijkman Institute for Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Leily Trianty

Eijkman Institute for Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Irene Handayuni

Charles Darwin University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Arnab K. Chatterjee

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge