Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Antiviral Res ; 192: 105122, 2021 08.
Article in English | MEDLINE | ID: mdl-34186107

ABSTRACT

There are, besides remdesivir, no approved antivirals for the treatment of SARS-CoV-2 infections. To aid in the search for antivirals against this virus, we explored the use of human tracheal airway epithelial cells (HtAEC) and human small airway epithelial cells (HsAEC) grown at the air-liquid interface (ALI). These cultures were infected at the apical side with one of two different SARS-CoV-2 isolates. Each virus was shown to replicate to high titers for extended periods of time (at least 8 days) and, in particular an isolate with the D614G in the spike (S) protein did so more efficiently at 35 °C than 37 °C. The effect of a selected panel of reference drugs that were added to the culture medium at the basolateral side of the system was explored. Remdesivir, GS-441524 (the parent nucleoside of remdesivir), EIDD-1931 (the parent nucleoside of molnupiravir) and IFN (ß1 and λ1) all resulted in dose-dependent inhibition of viral RNA and infectious virus titers collected at the apical side. However, AT-511 (the free base form of AT-527 currently in clinical testing) failed to inhibit viral replication in these in vitro primary cell models. Together, these results provide a reference for further studies aimed at selecting SARS-CoV-2 inhibitors for further preclinical and clinical development.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , SARS-CoV-2/drug effects , Virus Replication/drug effects , Animals , Cell Line , Chlorocebus aethiops , Drug Evaluation, Preclinical/methods , Epithelial Cells/virology , Humans , RNA, Viral , SARS-CoV-2/isolation & purification , Vero Cells
2.
Nat Commun ; 12(1): 3309, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083527

ABSTRACT

The ongoing pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), necessitates strategies to identify prophylactic and therapeutic drug candidates for rapid clinical deployment. Here, we describe a screening pipeline for the discovery of efficacious SARS-CoV-2 inhibitors. We screen a best-in-class drug repurposing library, ReFRAME, against two high-throughput, high-content imaging infection assays: one using HeLa cells expressing SARS-CoV-2 receptor ACE2 and the other using lung epithelial Calu-3 cells. From nearly 12,000 compounds, we identify 49 (in HeLa-ACE2) and 41 (in Calu-3) compounds capable of selectively inhibiting SARS-CoV-2 replication. Notably, most screen hits are cell-line specific, likely due to different virus entry mechanisms or host cell-specific sensitivities to modulators. Among these promising hits, the antivirals nelfinavir and the parent of prodrug MK-4482 possess desirable in vitro activity, pharmacokinetic and human safety profiles, and both reduce SARS-CoV-2 replication in an orthogonal human differentiated primary cell model. Furthermore, MK-4482 effectively blocks SARS-CoV-2 infection in a hamster model. Overall, we identify direct-acting antivirals as the most promising compounds for drug repurposing, additional compounds that may have value in combination therapies, and tool compounds for identification of viral host cell targets.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Drug Repositioning/methods , Pandemics , SARS-CoV-2 , Animals , COVID-19/prevention & control , COVID-19/virology , Cell Line , Cytidine/administration & dosage , Cytidine/analogs & derivatives , Cytidine/pharmacology , Databases, Pharmaceutical , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , HeLa Cells , High-Throughput Screening Assays/methods , Humans , Hydroxylamines/administration & dosage , Hydroxylamines/pharmacology , Mesocricetus , Nelfinavir/pharmacology , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Virus Replication/drug effects
3.
Cell Rep ; 35(1): 108940, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33784499

ABSTRACT

SARS-CoV-2 has currently precipitated the COVID-19 global health crisis. We developed a medium-throughput drug-screening system and identified a small-molecule library of 34 of 430 protein kinase inhibitors that were capable of inhibiting the SARS-CoV-2 cytopathic effect in human epithelial cells. These drug inhibitors are in various stages of clinical trials. We detected key proteins involved in cellular signaling pathways mTOR-PI3K-AKT, ABL-BCR/MAPK, and DNA-damage response that are critical for SARS-CoV-2 infection. A drug-protein interaction-based secondary screen confirmed compounds, such as the ATR kinase inhibitor berzosertib and torin2 with anti-SARS-CoV-2 activity. Berzosertib exhibited potent antiviral activity against SARS-CoV-2 in multiple cell types and blocked replication at the post-entry step. Berzosertib inhibited replication of SARS-CoV-1 and the Middle East respiratory syndrome coronavirus (MERS-CoV) as well. Our study highlights key promising kinase inhibitors to constrain coronavirus replication as a host-directed therapy in the treatment of COVID-19 and beyond as well as provides an important mechanism of host-pathogen interactions.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , DNA Damage , Isoxazoles/pharmacology , Pyrazines/pharmacology , SARS-CoV-2/physiology , Virus Replication/drug effects , A549 Cells , Animals , COVID-19/metabolism , COVID-19/pathology , Chlorocebus aethiops , Drug Evaluation, Preclinical , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Signaling System/drug effects , Middle East Respiratory Syndrome Coronavirus/metabolism , Vero Cells
4.
Nature ; 586(7827): 113-119, 2020 10.
Article in English | MEDLINE | ID: mdl-32707573

ABSTRACT

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 has triggered an ongoing global pandemic of the severe pneumonia-like disease coronavirus disease 2019 (COVID-19)1. The development of a vaccine is likely to take at least 12-18 months, and the typical timeline for approval of a new antiviral therapeutic agent can exceed 10 years. Thus, repurposing of known drugs could substantially accelerate the deployment of new therapies for COVID-19. Here we profiled a library of drugs encompassing approximately 12,000 clinical-stage or Food and Drug Administration (FDA)-approved small molecules to identify candidate therapeutic drugs for COVID-19. We report the identification of 100 molecules that inhibit viral replication of SARS-CoV-2, including 21 drugs that exhibit dose-response relationships. Of these, thirteen were found to harbour effective concentrations commensurate with probable achievable therapeutic doses in patients, including the PIKfyve kinase inhibitor apilimod2-4 and the cysteine protease inhibitors MDL-28170, Z LVG CHN2, VBY-825 and ONO 5334. Notably, MDL-28170, ONO 5334 and apilimod were found to antagonize viral replication in human pneumocyte-like cells derived from induced pluripotent stem cells, and apilimod also demonstrated antiviral efficacy in a primary human lung explant model. Since most of the molecules identified in this study have already advanced into the clinic, their known pharmacological and human safety profiles will enable accelerated preclinical and clinical evaluation of these drugs for the treatment of COVID-19.


Subject(s)
Antiviral Agents/analysis , Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Drug Evaluation, Preclinical , Drug Repositioning , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/drug effects , Betacoronavirus/growth & development , COVID-19 , Cell Line , Cysteine Proteinase Inhibitors/analysis , Cysteine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation/drug effects , Humans , Hydrazones , Induced Pluripotent Stem Cells/cytology , Models, Biological , Morpholines/analysis , Morpholines/pharmacology , Pandemics , Pyrimidines , Reproducibility of Results , SARS-CoV-2 , Small Molecule Libraries/analysis , Small Molecule Libraries/pharmacology , Triazines/analysis , Triazines/pharmacology , Virus Internalization/drug effects , Virus Replication/drug effects , COVID-19 Drug Treatment
5.
Antiviral Res ; 169: 104558, 2019 09.
Article in English | MEDLINE | ID: mdl-31302150

ABSTRACT

Several mammarenaviruses, chiefly Lassa virus (LASV) in Western Africa and Junín virus (JUNV) in the Argentine Pampas, cause severe disease in humans and pose important public health problems in their endemic regions. Moreover, mounting evidence indicates that the worldwide-distributed mammarenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen of clinical significance. The lack of licensed mammarenavirus vaccines and partial efficacy of current anti-mammarenavirus therapy limited to an off-label use of the nucleoside analog ribavirin underscore an unmet need for novel therapeutics to combat human pathogenic mammarenavirus infections. This task can be facilitated by the implementation of "drug repurposing" strategies to reduce the time and resources required to advance identified antiviral drug candidates into the clinic. We screened a drug repurposing library of 11,968 compounds (Repurposing, Focused Rescue and Accelerated Medchem [ReFRAME]) and identified several potent inhibitors of LCMV multiplication that had also strong anti-viral activity against LASV and JUNV. Our findings indicate that enzymes of the rate-limiting steps of pyrimidine and purine biosynthesis, the pro-viral MCL1 apoptosis regulator, BCL2 family member protein and the mitochondrial electron transport complex III, play critical roles in the completion of the mammarenavirus life cycle, suggesting they represent potential druggable targets to counter human pathogenic mammarenavirus infections.


Subject(s)
Antiviral Agents/pharmacology , Arenaviridae/drug effects , Databases, Pharmaceutical , Drug Evaluation, Preclinical/methods , Drug Repositioning/methods , A549 Cells , Animals , Apoptosis , Arenaviridae/physiology , Arenaviridae Infections/drug therapy , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Chlorocebus aethiops , Dose-Response Relationship, Drug , Electron Transport Complex III/metabolism , HEK293 Cells , Humans , Interferons/genetics , Junin virus/drug effects , Lassa virus/drug effects , Lymphocytic choriomeningitis virus/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Purines/biosynthesis , Pyrimidines/biosynthesis , Vero Cells , Virus Replication/drug effects
6.
Proc Natl Acad Sci U S A ; 115(42): 10750-10755, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30282735

ABSTRACT

The chemical diversity and known safety profiles of drugs previously tested in humans make them a valuable set of compounds to explore potential therapeutic utility in indications outside those originally targeted, especially neglected tropical diseases. This practice of "drug repurposing" has become commonplace in academic and other nonprofit drug-discovery efforts, with the appeal that significantly less time and resources are required to advance a candidate into the clinic. Here, we report a comprehensive open-access, drug repositioning screening set of 12,000 compounds (termed ReFRAME; Repurposing, Focused Rescue, and Accelerated Medchem) that was assembled by combining three widely used commercial drug competitive intelligence databases (Clarivate Integrity, GVK Excelra GoStar, and Citeline Pharmaprojects), together with extensive patent mining of small molecules that have been dosed in humans. To date, 12,000 compounds (∼80% of compounds identified from data mining) have been purchased or synthesized and subsequently plated for screening. To exemplify its utility, this collection was screened against Cryptosporidium spp., a major cause of childhood diarrhea in the developing world, and two active compounds previously tested in humans for other therapeutic indications were identified. Both compounds, VB-201 and a structurally related analog of ASP-7962, were subsequently shown to be efficacious in animal models of Cryptosporidium infection at clinically relevant doses, based on available human doses. In addition, an open-access data portal (https://reframedb.org) has been developed to share ReFRAME screen hits to encourage additional follow-up and maximize the impact of the ReFRAME screening collection.


Subject(s)
Antiprotozoal Agents/pharmacology , Cryptosporidiosis/drug therapy , Cryptosporidium/drug effects , Databases, Pharmaceutical , Drug Discovery , Drug Repositioning/methods , Small Molecule Libraries/pharmacology , Animals , Cryptosporidiosis/parasitology , Drug Evaluation, Preclinical/methods , Female , High-Throughput Screening Assays , Humans , Mice , Mice, Inbred C57BL
7.
Angew Chem Int Ed Engl ; 57(13): 3478-3482, 2018 03 19.
Article in English | MEDLINE | ID: mdl-29388301

ABSTRACT

The generation of ATP through oxidative phosphorylation is an essential metabolic function for Mycobaterium tuberculosis (Mtb), regardless of the growth environment. The type II NADH dehydrogenase (Ndh-2) is the conduit for electrons into the pathway, and is absent in the mammalian genome, thus making it a potential drug target. Herein, we report the identification of two types of small molecules as selective inhibitors for Ndh-2 through a multicomponent high-throughput screen. Both compounds block ATP synthesis, lead to effects consistent with loss of NADH turnover, and importantly, exert bactericidal activity against Mtb. Extensive medicinal chemistry optimization afforded the best analogue with an MIC of 90 nm against Mtb. Moreover, the two scaffolds have differential inhibitory activities against the two homologous Ndh-2 enzymes in Mtb, which will allow precise control over Ndh-2 function in Mtb to facilitate the assessment of this anti-TB drug target.


Subject(s)
Anti-Bacterial Agents/pharmacology , Indazoles/pharmacology , Mycobacterium tuberculosis/enzymology , NADH Dehydrogenase/antagonists & inhibitors , Quinazolines/pharmacology , Drug Evaluation, Preclinical , Microbial Viability/drug effects
8.
PLoS Negl Trop Dis ; 11(2): e0005373, 2017 02.
Article in English | MEDLINE | ID: mdl-28158186

ABSTRACT

Cryptosporidiosis has emerged as a leading cause of non-viral diarrhea in children under five years of age in the developing world, yet the current standard of care to treat Cryptosporidium infections, nitazoxanide, demonstrates limited and immune-dependent efficacy. Given the lack of treatments with universal efficacy, drug discovery efforts against cryptosporidiosis are necessary to find therapeutics more efficacious than the standard of care. To date, cryptosporidiosis drug discovery efforts have been limited to a few targeted mechanisms in the parasite and whole cell phenotypic screens against small, focused collections of compounds. Using a previous screen as a basis, we initiated the largest known drug discovery effort to identify novel anticryptosporidial agents. A high-content imaging assay for inhibitors of Cryptosporidium parvum proliferation within a human intestinal epithelial cell line was miniaturized and automated to enable high-throughput phenotypic screening against a large, diverse library of small molecules. A screen of 78,942 compounds identified 12 anticryptosporidial hits with sub-micromolar activity, including clofazimine, an FDA-approved drug for the treatment of leprosy, which demonstrated potent and selective in vitro activity (EC50 = 15 nM) against C. parvum. Clofazimine also displayed activity against C. hominis-the other most clinically-relevant species of Cryptosporidium. Importantly, clofazimine is known to accumulate within epithelial cells of the small intestine, the primary site of Cryptosporidium infection. In a mouse model of acute cryptosporidiosis, a once daily dosage regimen for three consecutive days or a single high dose resulted in reduction of oocyst shedding below the limit detectable by flow cytometry. Recently, a target product profile (TPP) for an anticryptosporidial compound was proposed by Huston et al. and highlights the need for a short dosing regimen (< 7 days) and formulations for children < 2 years. Clofazimine has a long history of use and has demonstrated a good safety profile for a disease that requires chronic dosing for a period of time ranging 3-36 months. These results, taken with clofazimine's status as an FDA-approved drug with over four decades of use for the treatment of leprosy, support the continued investigation of clofazimine both as a new chemical tool for understanding cryptosporidium biology and a potential new treatment of cryptosporidiosis.


Subject(s)
Antiprotozoal Agents/pharmacology , Clofazimine/pharmacology , Cryptosporidiosis/drug therapy , Cryptosporidium parvum/drug effects , Drug Repositioning , Animals , Automation, Laboratory , Cell Line , Cryptosporidiosis/parasitology , Disease Models, Animal , Drug Evaluation, Preclinical , Epithelial Cells/parasitology , High-Throughput Screening Assays , Humans , Mice , Treatment Outcome
9.
Proc Natl Acad Sci U S A ; 113(41): 11501-11506, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27663736

ABSTRACT

A variable region fusion strategy was used to generate an immunosuppressive antibody based on a novel "stalk-knob" structural motif in the ultralong complementary-determining region (CDR) of a bovine antibody. The potent Kv1.3 channel inhibitory peptides Moka1-toxin and Vm24-toxin were grafted into different CDRs of the humanized antibodies BVK and Synagis (Syn) using both ß-sheet and coiled-coil linkers. Structure-activity relationship efforts led to generation of the fusion protein Syn-Vm24-CDR3L, which demonstrated excellent selectivity and potency against effector human memory T cells (subnanomolar to picomolar EC50 values). This fusion antibody also had significantly improved plasma half-life and serum stability in rodents compared with the parent Vm24 peptide. Finally, this fusion protein showed potent in vivo efficacy in the delayed type hypersensitivity in rats. These results illustrate the utility of antibody CDR fusions as a general and effective strategy to generate long-acting functional antibodies, and may lead to a selective immunosuppressive antibody for the treatment of autoimmune diseases.


Subject(s)
Antibodies, Blocking/pharmacology , Drug Design , Immunosuppressive Agents/pharmacology , Kv1.3 Potassium Channel/antagonists & inhibitors , Amino Acid Sequence , Animals , CHO Cells , Cattle , Complementarity Determining Regions/chemistry , Cricetinae , Cricetulus , HEK293 Cells , Humans , Lymphocyte Activation/drug effects , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology
10.
J Biomol Screen ; 20(1): 101-11, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25281737

ABSTRACT

Chagas disease affects 8 million people worldwide and remains a main cause of death due to heart failure in Latin America. The number of cases in the United States is now estimated to be 300,000, but there are currently no Food and Drug Administration (FDA)-approved drugs available for patients with Chagas disease. To fill this gap, we have established a public-private partnership between the University of California, San Francisco and the Genomics Institute of the Novartis Research Foundation (GNF) with the goal of delivering clinical candidates to treat Chagas disease. The discovery phase, based on the screening of more than 160,000 compounds from the GNF Academic Collaboration Library, led to the identification of new anti-Chagas scaffolds. Part of the screening campaign used and compared two screening methods, including a colorimetric-based assay using Trypanosoma cruzi expressing ß-galactosidase and an image-based, high-content screening (HCS) assay using the CA-I/72 strain of T. cruzi. Comparing molecules tested in both assays, we found that ergosterol biosynthesis inhibitors had greater potency in the colorimetric assay than in the HCS assay. Both assays were used to inform structure-activity relationships for antiparasitic efficacy and pharmacokinetics. A new anti-T. cruzi scaffold derived from xanthine was identified, and we describe its development as lead series.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Animals , Cell Line , Chagas Disease/drug therapy , Colorimetry/methods , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Mice , Neglected Diseases/drug therapy , Small Molecule Libraries , Trypanocidal Agents/chemistry , Xanthine/chemistry , Xanthine/pharmacology
11.
Antimicrob Agents Chemother ; 58(3): 1586-95, 2014.
Article in English | MEDLINE | ID: mdl-24366744

ABSTRACT

Preventing relapses of Plasmodium vivax malaria through a radical cure depends on use of the 8-aminoquinoline primaquine, which is associated with safety and compliance issues. For future malaria eradication strategies, new, safer radical curative compounds that efficiently kill dormant liver stages (hypnozoites) will be essential. A new compound with potential radical cure activity was identified using a low-throughput assay of in vitro-cultured hypnozoite forms of Plasmodium cynomolgi (an excellent and accessible model for Plasmodium vivax). In this assay, primary rhesus hepatocytes are infected with P. cynomolgi sporozoites, and exoerythrocytic development is monitored in the presence of compounds. Liver stage cultures are fixed after 6 days and stained with anti-Hsp70 antibodies, and the relative proportions of small (hypnozoite) and large (schizont) forms relative to the untreated controls are determined. This assay was used to screen a series of 18 known antimalarials and 14 new non-8-aminoquinolines (preselected for blood and/or liver stage activity) in three-point 10-fold dilutions (0.1, 1, and 10 µM final concentrations). A novel compound, designated KAI407 showed an activity profile similar to that of primaquine (PQ), efficiently killing the earliest stages of the parasites that become either primary hepatic schizonts or hypnozoites (50% inhibitory concentration [IC50] for hypnozoites, KAI407, 0.69 µM, and PQ, 0.84 µM; for developing liver stages, KAI407, 0.64 µM, and PQ, 0.37 µM). When given as causal prophylaxis, a single oral dose of 100 mg/kg of body weight prevented blood stage parasitemia in mice. From these results, we conclude that KAI407 may represent a new compound class for P. vivax malaria prophylaxis and potentially a radical cure.


Subject(s)
Antimalarials/pharmacology , Imidazoles/pharmacology , Malaria/drug therapy , Plasmodium cynomolgi/drug effects , Pyrazines/pharmacology , Animals , Antimalarials/therapeutic use , Drug Evaluation, Preclinical/methods , Female , Hepatocytes/parasitology , Imidazoles/therapeutic use , In Vitro Techniques , Liver/parasitology , Macaca mulatta/parasitology , Malaria/parasitology , Malaria/prevention & control , Mice , Mice, Inbred ICR , Pyrazines/therapeutic use , Sporozoites/drug effects
12.
Sci Transl Med ; 5(214): 214ra168, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24307692

ABSTRACT

New chemotherapeutic compounds against multidrug-resistant Mycobacterium tuberculosis (Mtb) are urgently needed to combat drug resistance in tuberculosis (TB). We have identified and characterized the indolcarboxamides as a new class of antitubercular bactericidal agent. Genetic and lipid profiling studies identified the likely molecular target of indolcarboxamides as MmpL3, a transporter of trehalose monomycolate that is essential for mycobacterial cell wall biosynthesis. Two lead candidates, NITD-304 and NITD-349, showed potent activity against both drug-sensitive and multidrug-resistant clinical isolates of Mtb. Promising pharmacokinetic profiles of both compounds after oral dosing in several species enabled further evaluation for efficacy and safety. NITD-304 and NITD-349 were efficacious in treating both acute and chronic Mtb infections in mouse efficacy models. Furthermore, dosing of NITD-304 and NITD-349 for 2 weeks in exploratory rat toxicology studies revealed a promising safety margin. Finally, neither compound inhibited the activity of major cytochrome P-450 enzymes or the hERG (human ether-a-go-go related gene) channel. These results suggest that NITD-304 and NITD-349 should undergo further development as a potential treatment for multidrug-resistant TB.


Subject(s)
Antitubercular Agents/pharmacology , Indoles/pharmacology , Mycobacterium tuberculosis/drug effects , Tuberculosis, Multidrug-Resistant/drug therapy , Administration, Oral , Animals , Antitubercular Agents/administration & dosage , Antitubercular Agents/pharmacokinetics , Antitubercular Agents/toxicity , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Biological Availability , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Resistance, Multiple, Bacterial/genetics , Humans , Indoles/administration & dosage , Indoles/pharmacokinetics , Indoles/toxicity , Injections, Intravenous , Membrane Transport Proteins/drug effects , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Rats , Rats, Wistar , Tuberculosis, Multidrug-Resistant/diagnosis , Tuberculosis, Multidrug-Resistant/microbiology
13.
Nat Rev Microbiol ; 11(12): 849-62, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24217412

ABSTRACT

Malaria elimination has recently been reinstated as a global health priority but current therapies seem to be insufficient for the task. Elimination efforts require new drug classes that alleviate symptoms, prevent transmission and provide a radical cure. To develop these next-generation medicines, public-private partnerships are funding innovative approaches to identify compounds that target multiple parasite species at multiple stages of the parasite life cycle. In this Review, we discuss the cell-, chemistry- and target-based approaches used to discover new drug candidates that are currently in clinical trials or undergoing preclinical testing.


Subject(s)
Antimalarials/isolation & purification , Antimalarials/pharmacology , Drug Discovery/methods , Drug Discovery/trends , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans
14.
Biomed Chromatogr ; 27(12): 1720-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23881514

ABSTRACT

A simple, rapid, accurate and reproducible reverse-phase HPLC method has been developed for the identification and quantification of two alkaloids ephedrine and cryptolepine in different extracts of Sida species using photodiode array detection. Baseline separation of the two alkaloids was achieved on a Waters RP-18 X-terra column (250 × 4.6 mm, 5 µm) using a solvent system consisting of a mixture of water containing 0.1% Trifluoroacetic acid (TFA) and acetonitrile in a gradient elution mode with detection at 210 and 280 nm for ephedrine and cryptolepine, respectively. The calibration curves were linear in a concentration range of 10-250 µg/mL for both the alkaloids with correlation coefficient values >0.99. The limits of detection and quantification for ephedrine and cryptolepine were 5 and 10 µg/mL and 2.5 and 5 µg/mL, respectively. Relative standard deviation values for intra-day and inter-day precision were 1.22 and 1.04% for ephedrine and 1.71 and 2.06% for cryptolepine, respectively. Analytical recovery ranged from 92.46 to 103.95%. The developed HPLC method was applied to identify and quantify ephedrine and cryptolepine in different extracts of Sida species.


Subject(s)
Chromatography, High Pressure Liquid/methods , Ephedrine/analysis , Indole Alkaloids/analysis , Malvaceae/chemistry , Plant Extracts/chemistry , Quinolines/analysis , Ephedrine/chemistry , Indole Alkaloids/chemistry , Least-Squares Analysis , Quinolines/chemistry , Reproducibility of Results , Sensitivity and Specificity
15.
Science ; 334(6061): 1372-7, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22096101

ABSTRACT

Most malaria drug development focuses on parasite stages detected in red blood cells, even though, to achieve eradication, next-generation drugs active against both erythrocytic and exo-erythrocytic forms would be preferable. We applied a multifactorial approach to a set of >4000 commercially available compounds with previously demonstrated blood-stage activity (median inhibitory concentration < 1 micromolar) and identified chemical scaffolds with potent activity against both forms. From this screen, we identified an imidazolopiperazine scaffold series that was highly enriched among compounds active against Plasmodium liver stages. The orally bioavailable lead imidazolopiperazine confers complete causal prophylactic protection (15 milligrams/kilogram) in rodent models of malaria and shows potent in vivo blood-stage therapeutic activity. The open-source chemical tools resulting from our effort provide starting points for future drug discovery programs, as well as opportunities for researchers to investigate the biology of exo-erythrocytic forms.


Subject(s)
Antimalarials/pharmacology , Drug Discovery , Imidazoles/pharmacology , Liver/parasitology , Malaria/drug therapy , Piperazines/pharmacology , Plasmodium/drug effects , Animals , Antimalarials/chemistry , Antimalarials/pharmacokinetics , Antimalarials/therapeutic use , Cell Line, Tumor , Drug Evaluation, Preclinical , Drug Resistance , Erythrocytes/parasitology , Humans , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Imidazoles/therapeutic use , Malaria/parasitology , Malaria/prevention & control , Mice , Mice, Inbred BALB C , Molecular Structure , Piperazines/chemistry , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Plasmodium/cytology , Plasmodium/growth & development , Plasmodium/physiology , Plasmodium berghei/cytology , Plasmodium berghei/drug effects , Plasmodium berghei/growth & development , Plasmodium berghei/physiology , Plasmodium falciparum/cytology , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Plasmodium falciparum/physiology , Plasmodium yoelii/cytology , Plasmodium yoelii/drug effects , Plasmodium yoelii/growth & development , Plasmodium yoelii/physiology , Polymorphism, Single Nucleotide , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Random Allocation , Small Molecule Libraries , Sporozoites/drug effects , Sporozoites/growth & development
16.
Mol Biochem Parasitol ; 175(1): 21-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20813141

ABSTRACT

The efficacy of most marketed antimalarial drugs has been compromised by evolution of parasite resistance, underscoring an urgent need to find new drugs with new mechanisms of action. We have taken a high-throughput approach toward identifying novel antimalarial chemical inhibitors of prioritized drug targets for Plasmodium falciparum, excluding targets which are inhibited by currently used drugs. A screen of commercially available libraries identified 5655 low molecular weight compounds that inhibit growth of P. falciparum cultures with EC(50) values below 1.25µM. These compounds were then tested in 384- or 1536-well biochemical assays for activity against nine Plasmodium enzymes: adenylosuccinate synthetase (AdSS), choline kinase (CK), deoxyuridine triphosphate nucleotidohydrolase (dUTPase), glutamate dehydrogenase (GDH), guanylate kinase (GK), N-myristoyltransferase (NMT), orotidine 5'-monophosphate decarboxylase (OMPDC), farnesyl pyrophosphate synthase (FPPS) and S-adenosylhomocysteine hydrolase (SAHH). These enzymes were selected using TDRtargets.org, and are believed to have excellent potential as drug targets based on criteria such as their likely essentiality, druggability, and amenability to high-throughput biochemical screening. Six of these targets were inhibited by one or more of the antimalarial scaffolds and may have potential use in drug development, further target validation studies and exploration of P. falciparum biochemistry and biology.


Subject(s)
Antimalarials/pharmacology , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Plasmodium falciparum/drug effects , Antimalarials/isolation & purification , Enzyme Inhibitors/isolation & purification , Enzymes/metabolism , Inhibitory Concentration 50 , Plasmodium falciparum/growth & development , Protozoan Proteins/antagonists & inhibitors
17.
Proc Natl Acad Sci U S A ; 105(26): 9059-64, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18579783

ABSTRACT

The growing resistance to current first-line antimalarial drugs represents a major health challenge. To facilitate the discovery of new antimalarials, we have implemented an efficient and robust high-throughput cell-based screen (1,536-well format) based on proliferation of Plasmodium falciparum (Pf) in erythrocytes. From a screen of approximately 1.7 million compounds, we identified a diverse collection of approximately 6,000 small molecules comprised of >530 distinct scaffolds, all of which show potent antimalarial activity (<1.25 microM). Most known antimalarials were identified in this screen, thus validating our approach. In addition, we identified many novel chemical scaffolds, which likely act through both known and novel pathways. We further show that in some cases the mechanism of action of these antimalarials can be determined by in silico compound activity profiling. This method uses large datasets from unrelated cellular and biochemical screens and the guilt-by-association principle to predict which cellular pathway and/or protein target is being inhibited by select compounds. In addition, the screening method has the potential to provide the malaria community with many new starting points for the development of biological probes and drugs with novel antiparasitic activities.


Subject(s)
Antimalarials/analysis , Antimalarials/pharmacology , Computational Biology , Animals , Antimalarials/chemistry , Antimalarials/therapeutic use , Cluster Analysis , Drug Evaluation, Preclinical , Drug Resistance/drug effects , Folic Acid Antagonists/analysis , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/pharmacology , Malaria/drug therapy , Models, Molecular , Parasites/drug effects , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Reproducibility of Results , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/chemistry
18.
Nat Chem Biol ; 4(6): 347-56, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18454143

ABSTRACT

Calcium-dependent protein kinases play a crucial role in intracellular calcium signaling in plants, some algae and protozoa. In Plasmodium falciparum, calcium-dependent protein kinase 1 (PfCDPK1) is expressed during schizogony in the erythrocytic stage as well as in the sporozoite stage. It is coexpressed with genes that encode the parasite motor complex, a cellular component required for parasite invasion of host cells, parasite motility and potentially cytokinesis. A targeted gene-disruption approach demonstrated that pfcdpk1 seems to be essential for parasite viability. An in vitro biochemical screen using recombinant PfCDPK1 against a library of 20,000 compounds resulted in the identification of a series of structurally related 2,6,9-trisubstituted purines. Compound treatment caused sudden developmental arrest at the late schizont stage in P. falciparum and a large reduction in intracellular parasites in Toxoplasma gondii, which suggests a possible role for PfCDPK1 in regulation of parasite motility during egress and invasion.


Subject(s)
Adenine/analogs & derivatives , Antimalarials/pharmacology , Cyclohexylamines/pharmacology , Gene Expression Regulation, Enzymologic/genetics , Malaria/parasitology , Plasmodium falciparum/enzymology , Protein Kinases/drug effects , Protein Kinases/genetics , Protozoan Proteins/antagonists & inhibitors , Adenine/chemistry , Adenine/pharmacology , Adenine/therapeutic use , Animals , Antimalarials/chemistry , Antimalarials/therapeutic use , CHO Cells , Cell Line , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Cyclohexylamines/chemistry , Cyclohexylamines/therapeutic use , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , HeLa Cells , Humans , Life Cycle Stages/drug effects , Malaria/drug therapy , Malaria/immunology , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Structure , Molecular Weight , Movement/drug effects , Oligonucleotide Array Sequence Analysis/methods , Parasitic Sensitivity Tests , Plasmodium falciparum/growth & development , Protein Kinases/physiology , Protozoan Proteins/genetics , Protozoan Proteins/physiology , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Small Molecule Libraries , Stereoisomerism , Structure-Activity Relationship , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL