Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Virology ; 540: 195-206, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31929001

ABSTRACT

Respiratory syncytial virus (RSV) infection can cause mucus overproduction and bronchiolitis in infants leading to severe disease and hospitalization. As a therapeutic strategy, immune modulatory agents may help prevent RSV-driven immune responses that cause severe airway disease. We developed a high throughput screen to identify compounds that reduced RSV-driven mucin 5AC (Muc5AC) expression and identified dexamethasone. Despite leading to a pronounced reduction in RSV-driven Muc5AC, dexamethasone increased RSV infection in vitro and delayed viral clearance in mice. This correlated with reduced expression of a subset of immune response genes and reduced lymphocyte infiltration in vivo. Interestingly, dexamethasone increased RSV infection levels without altering antiviral interferon signaling. In summary, the immunosuppressive activities of dexamethasone had favorable inhibitory effects on RSV-driven mucus production yet prevented immune defense activities that limit RSV infection in vitro and in vivo. These findings offer an explanation for the lack of efficacy of glucocorticoids in RSV-infected patients.


Subject(s)
Dexamethasone/pharmacology , Interferons/metabolism , Mucus/metabolism , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/drug effects , Signal Transduction/drug effects , Virus Replication/drug effects , Animals , Cell Line , Cytokines/metabolism , Gene Regulatory Networks , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Mice , Mucin 5AC/genetics , Mucin 5AC/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Respiratory Syncytial Virus Infections/genetics
2.
Sci Transl Med ; 11(491)2019 05 08.
Article in English | MEDLINE | ID: mdl-31068442

ABSTRACT

Parasitic filarial nematodes cause debilitating infections in people in resource-limited countries. A clinically validated approach to eliminating worms uses a 4- to 6-week course of doxycycline that targets Wolbachia, a bacterial endosymbiont required for worm viability and reproduction. However, the prolonged length of therapy and contraindication in children and pregnant women have slowed adoption of this treatment. Here, we describe discovery and optimization of quinazolines CBR417 and CBR490 that, with a single dose, achieve >99% elimination of Wolbachia in the in vivo Litomosoides sigmodontis filarial infection model. The efficacious quinazoline series was identified by pairing a primary cell-based high-content imaging screen with an orthogonal ex vivo validation assay to rapidly quantify Wolbachia elimination in Brugia pahangi filarial ovaries. We screened 300,368 small molecules in the primary assay and identified 288 potent and selective hits. Of 134 primary hits tested, only 23.9% were active in the worm-based validation assay, 8 of which contained a quinazoline heterocycle core. Medicinal chemistry optimization generated quinazolines with excellent pharmacokinetic profiles in mice. Potent antiwolbachial activity was confirmed in L. sigmodontis, Brugia malayi, and Onchocerca ochengi in vivo preclinical models of filarial disease and in vitro selectivity against Loa loa (a safety concern in endemic areas). The favorable efficacy and in vitro safety profiles of CBR490 and CBR417 further support these as clinical candidates for treatment of filarial infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Drug Discovery , Filariasis/drug therapy , Filariasis/parasitology , Filarioidea/physiology , Quinazolines/therapeutic use , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Female , Filarioidea/drug effects , Filarioidea/microbiology , High-Throughput Screening Assays , Mice , Phenotype , Quinazolines/chemistry , Quinazolines/pharmacology , Small Molecule Libraries , Wolbachia/drug effects
3.
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
4.
ACS Med Chem Lett ; 5(8): 947-50, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25147620

ABSTRACT

Imidazopyridine 1 was identified from a phenotypic screen against P. falciparum (Pf) blood stages and subsequently optimized for activity on liver-stage schizonts of the rodent parasite P. yoelii (Py) as well as hypnozoites of the simian parasite P. cynomolgi (Pc). We applied these various assays to the cell-based lead optimization of the imidazopyrazines, exemplified by 3 (KAI407), and show that optimized compounds within the series with improved pharmacokinetic properties achieve causal prophylactic activity in vivo and may have the potential to target the dormant stages of P. vivax malaria.

5.
Antimicrob Agents Chemother ; 58(9): 5060-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24913172

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.


Subject(s)
Antimalarials/pharmacology , Imidazoles/pharmacology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/transmission , Piperazines/pharmacology , Animals , Inhibitory Concentration 50 , Mice , Mice, Inbred ICR , Plasmodium falciparum/drug effects , Sporozoites/drug effects
6.
Nature ; 504(7479): 248-253, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24284631

ABSTRACT

Achieving the goal of malaria elimination will depend on targeting Plasmodium pathways essential across all life stages. Here we identify a lipid kinase, phosphatidylinositol-4-OH kinase (PI(4)K), as the target of imidazopyrazines, a new antimalarial compound class that inhibits the intracellular development of multiple Plasmodium species at each stage of infection in the vertebrate host. Imidazopyrazines demonstrate potent preventive, therapeutic, and transmission-blocking activity in rodent malaria models, are active against blood-stage field isolates of the major human pathogens P. falciparum and P. vivax, and inhibit liver-stage hypnozoites in the simian parasite P. cynomolgi. We show that imidazopyrazines exert their effect through inhibitory interaction with the ATP-binding pocket of PI(4)K, altering the intracellular distribution of phosphatidylinositol-4-phosphate. Collectively, our data define PI(4)K as a key Plasmodium vulnerability, opening up new avenues of target-based discovery to identify drugs with an ideal activity profile for the prevention, treatment and elimination of malaria.


Subject(s)
1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Malaria/drug therapy , Malaria/parasitology , Plasmodium/drug effects , Plasmodium/enzymology , 1-Phosphatidylinositol 4-Kinase/chemistry , 1-Phosphatidylinositol 4-Kinase/genetics , 1-Phosphatidylinositol 4-Kinase/metabolism , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Cytokinesis/drug effects , Drug Resistance/drug effects , Drug Resistance/genetics , Fatty Acids/metabolism , Female , Hepatocytes/parasitology , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , Life Cycle Stages/drug effects , Macaca mulatta , Male , Models, Biological , Models, Molecular , Phosphatidylinositol Phosphates/metabolism , Plasmodium/classification , Plasmodium/growth & development , Pyrazoles/metabolism , Pyrazoles/pharmacology , Quinoxalines/metabolism , Quinoxalines/pharmacology , Reproducibility of Results , Schizonts/cytology , Schizonts/drug effects , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
7.
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
8.
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
9.
Cell ; 135(1): 49-60, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18854154

ABSTRACT

Human Immunodeficiency Viruses (HIV-1 and HIV-2) rely upon host-encoded proteins to facilitate their replication. Here, we combined genome-wide siRNA analyses with interrogation of human interactome databases to assemble a host-pathogen biochemical network containing 213 confirmed host cellular factors and 11 HIV-1-encoded proteins. Protein complexes that regulate ubiquitin conjugation, proteolysis, DNA-damage response, and RNA splicing were identified as important modulators of early-stage HIV-1 infection. Additionally, over 40 new factors were shown to specifically influence the initiation and/or kinetics of HIV-1 DNA synthesis, including cytoskeletal regulatory proteins, modulators of posttranslational modification, and nucleic acid-binding proteins. Finally, 15 proteins with diverse functional roles, including nuclear transport, prostaglandin synthesis, ubiquitination, and transcription, were found to influence nuclear import or viral DNA integration. Taken together, the multiscale approach described here has uncovered multiprotein virus-host interactions that likely act in concert to facilitate the early steps of HIV-1 infection.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , Proteins/metabolism , Virus Replication , Cell Line , Humans , RNA Interference , Two-Hybrid System Techniques
10.
Virology ; 362(1): 16-25, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17257639

ABSTRACT

Despite decades of research, HIV remains a global health threat. Issues of multi-drug resistance and lack of an effective vaccine have recently led to the targeting of host factors for anti-viral drug development. While a few genome-wide screens for novel HIV co-factors have been reported, the promise of finding a therapeutic target has yet to be realized. Here, we report a screen of a cDNA library representing 15,000 unique genes in an infectious HIV system, and show that genomic screening can lead to the identification of novel proviral host factors. Mixed lineage kinase 3 (MLK3/MAP3K11) was identified as one of the strongest enhancers of infection and mutant studies show that its activity is dependent on its kinase function. Consistent with its known role in the activation of the AP-1 pathway through JNK kinase, MLK3 was able to enhance Tat-dependent HIV transcription in vitro thus leading to an increase in infection signal. RNA interference studies confirm the involvement of endogenous MLK3 in HIV infection, further implicating this kinase as a potential therapeutic target.


Subject(s)
DNA, Complementary/analysis , HIV Infections/genetics , HIV/growth & development , MAP Kinase Kinase Kinases/physiology , Anti-HIV Agents/pharmacology , Gene Library , Gene Products, tat/physiology , Genome , HIV Infections/drug therapy , HeLa Cells , Humans , Jurkat Cells , MAP Kinase Kinase Kinases/genetics , Mutation , Proviruses/growth & development , RNA Interference , Transcription, Genetic , tat Gene Products, Human Immunodeficiency Virus , Mitogen-Activated Protein Kinase Kinase Kinase 11
11.
Bioorg Med Chem Lett ; 17(1): 260-5, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17035019

ABSTRACT

A series of 4-aminopyrimidines (1) was identified as novel HIV inhibitors of unknown molecular target. Structural modifications were carried out to establish its SAR and identify the linking site for target identification. A number of analogs were found to possess single digit inhibitory activity for HIV replication. Several analogs with various potential linkers, including a biotinated analog, also exhibited excellent potency, and could serve as tools for the identification of novel anti-HIV targets.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV-1/drug effects , Pyrimidines/chemistry , Anti-HIV Agents/chemical synthesis , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 16(16): 4174-7, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16781149

ABSTRACT

A novel sulfanyltriazole was discovered as an HIV-1 non-nucleoside reverse transcriptase inhibitor via HTS using a cell-based assay. Chemical modifications and molecular modeling studies were carried out to establish its SAR and understand its interactions with the enzyme. These modifications led to the identification of sulfanyltriazoles with low nanomolar potency for inhibiting HIV-1 replication and promising activities against selected NNRTI resistant mutants. These novel and potent sulfanyltriazoles could serve as advanced leads for further optimization.


Subject(s)
Anti-HIV Agents/pharmacology , Chemistry, Pharmaceutical/methods , HIV-1/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Triazoles/chemistry , Drug Design , Drug Resistance, Viral , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Models, Chemical , Models, Molecular , Mutation , Sulfur Compounds/chemistry , Temperature , Virus Replication
13.
Bioorg Med Chem Lett ; 16(16): 4246-51, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16782337

ABSTRACT

A novel series of quinolones was discovered as HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTIs) using a structure-based approach. The lead quinolones exhibited single digit nanomolar potency in the HIV-1 replication assays. The preliminary SAR of these quinolones was also established via systematic structural modifications. These novel and potent quinolones could serve as advanced leads for further optimization.


Subject(s)
Reverse Transcriptase Inhibitors/pharmacology , Binding Sites , Drug Resistance, Viral , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/metabolism , Models, Chemical , Models, Molecular , Mutation , Quinolones/chemistry , Structure-Activity Relationship , Virus Replication
14.
Bioorg Med Chem Lett ; 16(8): 2105-8, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16480865

ABSTRACT

A novel oxindole was discovered as an HIV non-nucleoside reverse transcriptase inhibitor via HTS using a cell-based assay. Systematic structural modifications were carried out to establish its SAR. These modifications led to the identification of oxindoles with low nanomolar potency for inhibiting HIV replication. These novel and potent oxindoles could serve as advanced leads for further optimizations.


Subject(s)
HIV-1/drug effects , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/pharmacology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacology , Drug Resistance, Viral , Indoles/chemical synthesis , Indoles/pharmacology , Stereoisomerism , Structure-Activity Relationship , Virus Replication
15.
Bioorg Med Chem Lett ; 16(8): 2109-12, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16464578

ABSTRACT

A series of heterocycle-containing oxindoles was synthesized and their HIV antiviral activities were assessed. Some of these analogs exhibited potent inhibitory activities against both wild-type virus and a number of drug-resistant mutant viruses. In addition, oxindole 9z also showed promising pharmacokinetics.


Subject(s)
HIV-1/drug effects , Indoles/chemical synthesis , Indoles/pharmacokinetics , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/pharmacokinetics , Virus Replication/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacology , Drug Design , Drug Resistance, Viral , Stereoisomerism , Structure-Activity Relationship
16.
J Virol ; 80(1): 130-7, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16352537

ABSTRACT

In order to identify novel proviral host factors involved in human immunodeficiency virus (HIV) infection, we performed a screen of a small interfering RNA (siRNA) library targeting 5,000 genes with the highest potential for being targets for therapeutics. Many siRNAs in the library against known host factors, such as TSG101, furin, and CXCR4, were identified as inhibitors by the screen and thus served as internal validation. In addition, many novel factors whose knockdown inhibited infection were identified, including Pak3, a member of the serine/threonine group I PAK kinases. The HIV accessory factor Nef has been shown to associate with a PAK kinase, leading to enhanced viral production; however, the exact identity of the kinase has remained controversial. Prompted by the Pak3 screen hit, we further investigated the involvement of group I PAK kinases in HIV using siRNA. Contrary to the current literature, Pak1 depletion strongly inhibited HIV infection in multiple cell systems and decreased levels of integrated provirus, while Pak2 depletion showed no effect. Overexpression of a constitutively active Pak1 mutant also enhanced HIV infection, further supporting its role as the dominant PAK involved.


Subject(s)
Gene Expression Regulation, Viral/drug effects , HIV Infections , Protein Serine-Threonine Kinases/physiology , RNA, Small Interfering/pharmacology , Virus Replication/drug effects , HIV-1/physiology , HeLa Cells , Humans , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Receptors, Cell Surface/genetics , Receptors, Virus/genetics , Virus Replication/genetics , p21-Activated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...