scholarly journals UCT943, a Next-GenerationPlasmodium falciparumPI4K Inhibitor Preclinical Candidate for the Treatment of Malaria

2018 ◽  
Vol 62 (9) ◽  
Author(s):  
Christel Brunschwig ◽  
Nina Lawrence ◽  
Dale Taylor ◽  
Efrem Abay ◽  
Mathew Njoroge ◽  
...  

ABSTRACTThe 2-aminopyridine MMV048 was the first drug candidate inhibitingPlasmodiumphosphatidylinositol 4-kinase (PI4K), a novel drug target for malaria, to enter clinical development. In an effort to identify the next generation of PI4K inhibitors, the series was optimized to improve properties such as solubility and antiplasmodial potency across the parasite life cycle, leading to the 2-aminopyrazine UCT943. The compound displayed higher asexual blood stage, transmission-blocking, and liver stage activities than MMV048 and was more potent against resistantPlasmodium falciparumandPlasmodium vivaxclinical isolates. Excellentin vitroantiplasmodial activity translated into high efficacy inPlasmodium bergheiand humanizedP. falciparumNOD-scid IL-2Rγnullmouse models. The high passive permeability and high aqueous solubility of UCT943, combined with low to moderatein vivointrinsic clearance, resulted in sustained exposure and high bioavailability in preclinical species. In addition, the predicted human dose for a curative single administration using monkey and dog pharmacokinetics was low, ranging from 50 to 80 mg. As a next-generationPlasmodiumPI4K inhibitor, UCT943, based on the combined preclinical data, has the potential to form part of a single-exposure radical cure and prophylaxis (SERCaP) to treat, prevent, and block the transmission of malaria.

2014 ◽  
Vol 58 (9) ◽  
pp. 5325-5331 ◽  
Author(s):  
Monalisa Chatterji ◽  
Radha Shandil ◽  
M. R. Manjunatha ◽  
Suresh Solapure ◽  
Vasanthi Ramachandran ◽  
...  

ABSTRACTNew therapeutic strategies against multidrug-resistant (MDR) and extensively drug-resistant (XDR)Mycobacterium tuberculosisare urgently required to combat the global tuberculosis (TB) threat. Toward this end, we previously reported the identification of 1,4-azaindoles, a promising class of compounds with potent antitubercular activity through noncovalent inhibition of decaprenylphosphoryl-β-d-ribose 2′-epimerase (DprE1). Further, this series was optimized to improve its physicochemical properties and pharmacokinetics in mice. Here, we describe the short-listing of a potential clinical candidate, compound 2, that has potent cellular activity, drug-like properties, efficacy in mouse and rat chronic TB infection models, and minimalin vitrosafety risks. We also demonstrate that the compounds, including compound 2, have no antagonistic activity with other anti-TB drugs. Moreover, compound 2 shows synergy with PA824 and TMC207in vitro, and the synergy effect is translatedin vivowith TMC207. The series is predicted to have a low clearance in humans, and the predicted human dose for compound 2 is ≤1 g/day. Altogether, our data suggest that a 1,4-azaindole (compound 2) is a promising candidate for the development of a novel anti-TB drug.


2011 ◽  
Vol 56 (2) ◽  
pp. 703-707 ◽  
Author(s):  
Sergio Wittlin ◽  
Eric Ekland ◽  
J Carl Craft ◽  
Julie Lotharius ◽  
Ian Bathurst ◽  
...  

ABSTRACTWith the emergence ofPlasmodium falciparuminfections exhibiting increased parasite clearance times in response to treatment with artemisinin-based combination therapies, the need for new therapeutic agents is urgent. Solithromycin, a potent new fluoroketolide currently in development, has been shown to be an effective, broad-spectrum antimicrobial agent. Malarial parasites possess an unusual organelle, termed the apicoplast, which carries a cryptic genome of prokaryotic origin that encodes its own translation and transcription machinery. Given the similarity of apicoplast and bacterial ribosomes, we have examined solithromycin for antimalarial activity. Other antibiotics known to target the apicoplast, such as the macrolide azithromycin, demonstrate a delayed-death effect, whereby treated asexual blood-stage parasites die in the second generation of drug exposure. Solithromycin demonstrated potentin vitroactivity against the NF54 strain ofP. falciparum, as well as against two multidrug-resistant strains, Dd2 and 7G8. The dramatic increase in potency observed after two generations of exposure suggests that it targets the apicoplast. Solithromycin also retained potency against azithromycin-resistant parasites derived from Dd2 and 7G8, although these lines did demonstrate a degree of cross-resistance. In anin vivomodel ofP. bergheiinfection in mice, solithromycin demonstrated a 100% cure rate when administered as a dosage regimen of four doses of 100 mg/kg of body weight, the same dose required for artesunate or chloroquine to achieve 100% cure rates in this rodent malaria model. These promisingin vitroandin vivodata support further investigations into the development of solithromycin as an antimalarial agent.


2012 ◽  
Vol 56 (7) ◽  
pp. 3849-3856 ◽  
Author(s):  
Subathdrage D. M. Sumanadasa ◽  
Christopher D. Goodman ◽  
Andrew J. Lucke ◽  
Tina Skinner-Adams ◽  
Ishani Sahama ◽  
...  

ABSTRACTHistone deacetylase (HDAC) enzymes posttranslationally modify lysines on histone and nonhistone proteins and play crucial roles in epigenetic regulation and other important cellular processes. HDAC inhibitors (e.g., suberoylanilide hydroxamic acid [SAHA; also known as vorinostat]) are used clinically to treat some cancers and are under investigation for use against many other diseases. Development of new HDAC inhibitors for noncancer indications has the potential to be accelerated by piggybacking onto cancer studies, as several HDAC inhibitors have undergone or are undergoing clinical trials. One such compound, SB939, is a new orally active hydroxamate-based HDAC inhibitor with an improved pharmacokinetic profile compared to that of SAHA. In this study, thein vitroandin vivoantiplasmodial activities of SB939 were investigated. SB939 was found to be a potent inhibitor of the growth ofPlasmodium falciparumasexual-stage parasitesin vitro(50% inhibitory concentration [IC50], 100 to 200 nM), causing hyperacetylation of parasite histone and nonhistone proteins. In combination with the aspartic protease inhibitor lopinavir, SB939 displayed additive activity. SB939 also potently inhibited thein vitrogrowth of exoerythrocytic-stagePlasmodiumparasites in liver cells (IC50, ∼150 nM), suggesting that inhibitor targeting to multiple malaria parasite life cycle stages may be possible. In an experimentalin vivomurine model of cerebral malaria, orally administered SB939 significantly inhibitedP. bergheiANKA parasite growth, preventing development of cerebral malaria-like symptoms. These results identify SB939 as a potent new antimalarial HDAC inhibitor and underscore the potential of investigating next-generation anticancer HDAC inhibitors as prospective new drug leads for treatment of malaria.


2014 ◽  
Vol 59 (1) ◽  
pp. 136-144 ◽  
Author(s):  
A. M. Upton ◽  
S. Cho ◽  
T. J. Yang ◽  
Y. Kim ◽  
Y. Wang ◽  
...  

ABSTRACTNitroimidazoles are a promising new class of antitubercular agents. The nitroimidazo-oxazole delamanid (OPC-67683, Deltyba) is in phase III trials for the treatment of multidrug-resistant tuberculosis, while the nitroimidazo-oxazine PA-824 is entering phase III for drug-sensitive and drug-resistant tuberculosis. TBA-354 (SN31354[(S)-2-nitro-6-((6-(4-trifluoromethoxy)phenyl)pyridine-3-yl)methoxy)-6,7-dihydro-5H-imidazo[2,1-b][1,3]oxazine]) is a pyridine-containing biaryl compound with exceptional efficacy against chronic murine tuberculosis and favorable bioavailability in preliminary rodent studies. It was selected as a potential next-generation antituberculosis nitroimidazole following an extensive medicinal chemistry effort. Here, we further evaluate the pharmacokinetic properties and activity of TBA-354 againstMycobacterium tuberculosis. TBA-354 is narrow spectrum and bactericidalin vitroagainst replicating and nonreplicatingMycobacterium tuberculosis, with potency similar to that of delamanid and greater than that of PA-824. The addition of serum protein or albumin does not significantly alter this activity. TBA-354 maintains activity againstMycobacterium tuberculosisH37Rv isogenic monoresistant strains and clinical drug-sensitive and drug-resistant isolates. Spontaneous resistant mutants appear at a frequency of 3 × 10−7.In vitrostudies andin vivostudies in mice confirm that TBA-354 has high bioavailability and a long elimination half-life.In vitrostudies suggest a low risk of drug-drug interactions. Low-dose aerosol infection models of acute and chronic murine tuberculosis reveal time- and dose-dependentin vivobactericidal activity that is at least as potent as that of delamanid and more potent than that of PA-824. Its superior potency and pharmacokinetic profile that predicts suitability for once-daily oral dosing suggest that TBA-354 be studied further for its potential as a next-generation nitroimidazole.


2017 ◽  
Vol 61 (12) ◽  
Author(s):  
Gordana Panic ◽  
Marie-Thérèse Ruf ◽  
Jennifer Keiser

ABSTRACT To date, there is only one drug in use, praziquantel, to treat more than 250 million people afflicted with schistosomiasis, a debilitating parasitic disease. The aryl hydantoin Ro 13-3978 is a promising drug candidate with in vivo activity superior to that of praziquantel against both adult and juvenile Schistosoma mansoni organisms. Given the drug's contrasting low activity in vitro and the timing of its onset of action in vivo, it was postulated that immune-assisted parasite clearance could contribute to the drug's in vivo activity. We undertook histopathological studies to investigate this hypothesis. Infected mice were treated with an effective dose of Ro 13-3978 (100 mg/kg of body weight) and were dissected before and after the drug's in vivo onset of action. The veins and livers were excised, paraffin-embedded, and sectioned, and macrophages (IBA-1), neutrophils (Neutro), B cells (CD45R), and T cells (CD3) were stained by immunohistochemistry. For comparison, samples from infected untreated mice and mice treated with effective doses of praziquantel (400 mg/kg), oxamniquine (200 mg/kg), and mefloquine (200 mg/kg) were examined. At 24 h after treatment with Ro 13-3978, significant macrophage recruitment to the veins was observed, along with a modest increase in circulating B cells, and at 48 h, neutrophils and T cells are also present. Treatment with praziquantel and oxamniquine showed similar patterns of recruitment but with comparatively higher cellular levels, whereas mefloquine treatment resulted in minimal cell recruitment until 3 days posttreatment. Our study sheds light on the immediate immune responses to antischistosomal treatment in mice and provides further insight into immune effector mechanisms of schistosome clearance.


2016 ◽  
Vol 60 (5) ◽  
pp. 2858-2863 ◽  
Author(s):  
Anne-Marie Zeeman ◽  
Suresh B. Lakshminarayana ◽  
Nicole van der Werff ◽  
Els J. Klooster ◽  
Annemarie Voorberg-van der Wel ◽  
...  

ABSTRACTTwoPlasmodiumPI4 kinase (PI4K) inhibitors, KDU691 and LMV599, were selected forin vivotesting as causal prophylactic and radical-cure agents forPlasmodium cynomolgisporozoite-infected rhesus macaques, based on theirin vitroactivity against liver stages. Animals were infected withP. cynomolgisporozoites, and compounds were dosed orally. Both the KDU691 and LMV599 compounds were fully protective when administered prophylactically, and the more potent compound LMV599 achieved protection as a single oral dose of 25 mg/kg of body weight. In contrast, when tested for radical cure, five daily doses of 20 mg/kg of KDU691 or 25 mg/kg of LMV599 did not prevent relapse, as all animals experienced a secondary infection due to the reactivation of hypnozoites in the liver. Pharmacokinetic data show that LMV599 achieved plasma exposure that was sufficient to achieve efficacy based on ourin vitrodata. These findings indicate thatPlasmodiumPI4K is a potential drug target for malaria prophylaxis but not radical cure. Longerin vitroculture systems will be required to assess these compounds' activity on established hypnozoites and predict radical curein vivo.


2012 ◽  
Vol 57 (1) ◽  
pp. 317-325 ◽  
Author(s):  
Neil R. Stokes ◽  
Nicola Baker ◽  
James M. Bennett ◽  
Joanne Berry ◽  
Ian Collins ◽  
...  

ABSTRACTThe bacterial cell division protein FtsZ is an attractive target for small-molecule antibacterial drug discovery. Derivatives of 3-methoxybenzamide, including compound PC190723, have been reported to be potent and selective antistaphylococcal agents which exert their effects through the disruption of intracellular FtsZ function. Here, we report the further optimization of 3-methoxybenzamide derivatives towards a drug candidate. Thein vitroandin vivocharacterization of a more advanced lead compound, designated compound 1, is described. Compound 1 was potently antibacterial, with an average MIC of 0.12 μg/ml against all staphylococcal species, including methicillin- and multidrug-resistantStaphylococcus aureusandStaphylococcus epidermidis. Compound 1 inhibited anS. aureusstrain carrying the G196A mutation in FtsZ, which confers resistance to PC190723. Like PC190723, compound 1 acted on whole bacterial cells by blocking cytokinesis. No interactions between compound 1 and a diverse panel of antibiotics were measured in checkerboard experiments. Compound 1 displayed suitablein vitropharmaceutical properties and a favorablein vivopharmacokinetic profile following intravenous and oral administration, with a calculated bioavailability of 82.0% in mice. Compound 1 demonstrated efficacy in a murine model of systemicS. aureusinfection and caused a significant decrease in the bacterial load in the thigh infection model. A greater reduction in the number ofS. aureuscells recovered from infected thighs, equivalent to 3.68 log units, than in those recovered from controls was achieved using a succinate prodrug of compound 1, which was designated compound 2. In summary, optimized derivatives of 3-methoxybenzamide may yield a first-in-class FtsZ inhibitor for the treatment of antibiotic-resistant staphylococcal infections.


2011 ◽  
Vol 55 (12) ◽  
pp. 5602-5608 ◽  
Author(s):  
Marcel Kaiser ◽  
Michael A. Bray ◽  
Monica Cal ◽  
Bernadette Bourdin Trunz ◽  
Els Torreele ◽  
...  

ABSTRACTFexinidazole is a 5-nitroimidazole drug currently in clinical development for the treatment of human sleeping sickness (human African trypanosomiasis [HAT]), caused by infection with species of the protozoan parasiteTrypanosoma brucei. The compound and its two principal metabolites, sulfoxide and sulfone, have been assessed for their ability to kill a range ofT. bruceiparasite strainsin vitroand to cure both acute and chronic HAT disease models in the mouse. The parent molecule and both metabolites have shown trypanocidal activityin vitroin the 0.7-to-3.3 μM (0.2-to-0.9 μg/ml) range against all parasite strains tested.In vivo, fexinidazole is orally effective in curing both acute and chronic diseases in the mouse at doses of 100 mg/kg of body weight/day for 4 days and 200 mg/kg/day for 5 days, respectively. Pharmacokinetic data indicate that it is likely that the sulfoxide and sulfone metabolites provide most, if not all, of thein vivokilling activity. Fexinidazole and its metabolites require up to 48 h exposure in order to induce maximal trypanocidal efficacyin vitro. The parent drug and its metabolites show noin vitrocross-reactivity in terms of trypanocidal activity with either themselves or other known trypanocidal drugs in use in humans. Thein vitroandin vivoantitrypanosomal activities of fexinidazole and its two principal metabolites provide evidence that the compound has the potential to be an effective oral treatment for both theT. b. gambienseandT. b. rhodesienseforms of human sleeping sickness and both stages of the disease.


2014 ◽  
Vol 59 (3) ◽  
pp. 1398-1404 ◽  
Author(s):  
Guzmán Álvarez ◽  
Javier Varela ◽  
Eugenia Cruces ◽  
Marcelo Fernández ◽  
Martín Gabay ◽  
...  

ABSTRACTAlthough the parasitic infection Chagas' disease was described over 100 years ago, even now there are not suitable drugs. The available drugs nifurtimox and benznidazole have limited efficacies and tolerances, with proven mutagenic effects. Attempting to find appropriate drugs to deal with this problem, here we report on the development and pharmacological characterization of new amide-containing thiazoles. In the present study, we evaluated thein vitroandin vivoeffects of new candidates againstTrypanosoma cruzi, the etiological agent of Chagas' disease. The lead amide-containing thiazole derivative had potentin vitroactivity, an absence of bothin vitromutagenic andin vivoclastogenic effects, and excellentin vitroselectivity andin vivotolerance. The compound suppressed parasitemia in mice, modifying the anti-T. cruziantibodies like the reference drug, benznidazole, and displayed the lowest mortality among the tested drugs. The present evidence suggests that this compound is a promising anti-T. cruziagent surpassing the lead optimization stage in drug development and leading to a candidate for preclinical study.


2013 ◽  
Vol 57 (10) ◽  
pp. 5045-5052 ◽  
Author(s):  
Zheng Hou ◽  
Fei Da ◽  
Baohui Liu ◽  
Xiaoyan Xue ◽  
Xiuli Xu ◽  
...  

ABSTRACTStaphylococcus epidermidisis one of the most frequent causes of device-associated infections, because it is known to cause biofilms that grow on catheters or other surgical implants. The persistent increasing resistance ofS. epidermidisand other coagulase-negative staphylococci (CoNS) has driven the need for newer antibacterial agents with innovative therapeutic strategies. Thanatin is reported to display potent antibiotic activities, especially against extended-spectrum-beta-lactamase-producingEscherichia coli. The present study aimed to investigate whether a shorter derivative peptide (R-thanatin) could be used as a novel antibacterial agent. We found that R-thanatin was highly potentin vitroagainst coagulase-negative staphylococci, such asS. epidermidis,S. haemolyticus, andS. hominis, and inhibited biofilm formation at subinhibitory concentrations. Properties of little toxicity to human red blood cells (hRBCs) and human umbilical vein endothelial cells, a low incidence of resistance, and relatively high stability in plasma were confirmed. Excellentin vivoprotective effects were also observed using a methicillin-resistantS. epidermidis(MRSE)-induced urinary tract infection rat model. Electron microscopy and confocal laser-scanning microscopy analyses suggested that R-thanatin disturbed cell division of MRSE severely, which might be the reason for inhibition of MRSE growth. These findings indicate that R-thanatin is active against the growth and biofilm formation of MRSEin vitroandin vivo. R-thanatin might be considered as a specific drug candidate for treating CoNS infections.


Sign in / Sign up

Export Citation Format

Share Document