Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
J Med Chem ; 60(6): 2305-2325, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28245119

ABSTRACT

Respiratory syncytial virus (RSV) represents a threat to infants, the elderly, and the immunocompromised. RSV entry blockers are in clinical trials, but escape mutations challenge their potential. In search of RSV inhibitors, we have integrated a signature resistance mutation into a recombinant RSV virus and applied the strain to high-throughput screening. Counterscreening of candidates returned 14 confirmed hits with activities in the nano- to low-micromolar range. All blocked RSV polymerase activity in minigenome assays. Compound 1a (GRP-74915) was selected for development based on activity (EC50 = 0.21 µM, selectivity index (SI) 40) and scaffold. Resynthesis confirmed the potency of the compound, which suppressed viral RNA synthesis in infected cells. However, metabolic testing revealed a short half-life in the presence of mouse hepatocyte fractions. Metabolite tracking and chemical elaboration combined with 3D-quantitative structure-activity relationship modeling yielded analogues (i.e., 8n: EC50 = 0.06 µM, SI 500) that establish a platform for the development of a therapeutic candidate.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Drug Design , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/enzymology , Animals , Antiviral Agents/metabolism , Cell Line , DNA-Directed RNA Polymerases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Mice , Quantitative Structure-Activity Relationship , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism
2.
J Virol ; 90(16): 7368-7387, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27252534

ABSTRACT

UNLABELLED: Influenza A virus (IAV) infections cause major morbidity and mortality, generating an urgent need for novel antiviral therapeutics. We recently established a dual myxovirus high-throughput screening protocol that combines a fully replication-competent IAV-WSN strain and a respiratory syncytial virus reporter strain for the simultaneous identification of IAV-specific, paramyxovirus-specific, and broad-spectrum inhibitors. In the present study, this protocol was applied to a screening campaign to assess a diverse chemical library with over 142,000 entries. Focusing on IAV-specific hits, we obtained a hit rate of 0.03% after cytotoxicity testing and counterscreening. Three chemically distinct hit classes with nanomolar potency and favorable cytotoxicity profiles were selected. Time-of-addition, minigenome, and viral entry studies demonstrated that these classes block hemagglutinin (HA)-mediated membrane fusion. Antiviral activity extends to an isolate from the 2009 pandemic and, in one case, another group 1 subtype. Target identification through biolayer interferometry confirmed binding of all hit compounds to HA. Resistance profiling revealed two distinct escape mechanisms: primary resistance, associated with reduced compound binding, and secondary resistance, associated with unaltered binding. Secondary resistance was mediated, unusually, through two different pairs of cooperative mutations, each combining a mutation eliminating the membrane-proximal stalk N-glycan with a membrane-distal change in HA1 or HA2. Chemical synthesis of an analog library combined with in silico docking extracted a docking pose for the hit classes. Chemical interrogation spotlights IAV HA as a major druggable target for small-molecule inhibition. Our study identifies novel chemical scaffolds with high developmental potential, outlines diverse routes of IAV escape from entry inhibition, and establishes a path toward structure-aided lead development. IMPORTANCE: This study is one of the first to apply a fully replication-competent third-generation IAV reporter strain to a large-scale high-throughput screen (HTS) drug discovery campaign, allowing multicycle infection and screening in physiologically relevant human respiratory cells. A large number of potential druggable targets was thus chemically interrogated, but mechanistic characterization, positive target identification, and resistance profiling demonstrated that three chemically promising and structurally distinct hit classes selected for further analysis all block HA-mediated membrane fusion. Viral escape from inhibition could be achieved through primary and secondary resistance mechanisms. In silico docking predicted compound binding to a microdomain located at the membrane-distal site of the prefusion HA stalk that was also previously suggested as a target site for chemically unrelated HA inhibitors. This study identifies an unexpected chemodominance of the HA stalk microdomain for small-molecule inhibitors in IAV inhibitor screening campaigns and highlights a novel mechanism of cooperative resistance to IAV entry blockers.


Subject(s)
Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Influenza A virus/drug effects , Influenza A virus/physiology , Virus Internalization/drug effects , Antiviral Agents/chemistry , Antiviral Agents/toxicity , Drug Resistance, Viral , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Mutation , Protein Binding
3.
Biochemistry ; 54(36): 5589-604, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26307636

ABSTRACT

Myxoviruses such as influenza A virus (IAV) and respiratory syncytial virus (RSV) are major human pathogens, mandating the development of novel therapeutics. To establish a high-throughput screening protocol for the simultaneous identification of pathogen- and host-targeted hit candidates against either pathogen or both, we have attempted co-infection of cells with IAV and RSV. However, viral replication kinetics were incompatible, RSV signal window was low, and an IAV-driven minireplicon reporter assay used in initial screens narrowed the host cell range and restricted the assay to single-cycle infections. To overcome these limitations, we developed an RSV strain carrying firefly luciferase fused to an innovative universal small-molecule assisted shut-off domain, which boosted assay signal window, and a hyperactive fusion protein that synchronized IAV and RSV reporter expression kinetics and suppressed the identification of RSV entry inhibitors sensitive to a recently reported RSV pan-resistance mechanism. Combined with a replication-competent recombinant IAV strain harboring nanoluciferase, the assay performed well on a human respiratory cell line and supports multicycle infections. Miniaturized to 384-well format, the protocol was validated through screening of a set of the National Institutes of Health Clinical Collection (NCC) in quadruplicate. These test screens demonstrated favorable assay parameters and reproducibility. Application to a LOPAC library of bioactive compounds in a proof-of-concept campaign detected licensed antimyxovirus therapeutics, ribavirin and the neuraminidase inhibitor zanamivir, and identified two unexpected RSV-specific hit candidates, Fenretinide and the opioid receptor antagonist BNTX-7. Hits were evaluated in direct and orthogonal dose-response counterscreens using a standard recRSV reporter strain expressing Renilla luciferase.


Subject(s)
Antiviral Agents/chemistry , Influenza A virus/genetics , Respiratory Syncytial Viruses/genetics , Animals , Antiviral Agents/pharmacology , Benzylidene Compounds/pharmacology , Cell Line , Coinfection , Dogs , Dose-Response Relationship, Drug , Feasibility Studies , Fenretinide/chemistry , Fenretinide/pharmacology , Genes, Reporter , High-Throughput Screening Assays , Humans , Influenza A virus/drug effects , Influenza A virus/physiology , Luciferases, Firefly/genetics , Luciferases, Renilla/genetics , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/chemistry , Narcotic Antagonists/pharmacology , Neuraminidase/antagonists & inhibitors , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/physiology , Ribavirin/chemistry , Ribavirin/pharmacology , Virus Internalization/drug effects , Virus Replication , Zanamivir/chemistry , Zanamivir/pharmacology
4.
DNA Cell Biol ; 34(8): 505-10, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25961744

ABSTRACT

Respiratory syncytial virus (RSV) is responsible for majority of infant hospitalizations due to viral infections. Despite its clinical importance, no vaccine against RSV or effective antiviral therapy is available. Several structural classes of small-molecule RSV entry inhibitor have been described and one compound has advanced to clinical testing. Mutations in either one of two resistance hot spots in the F protein mediate unusual pan-resistance to all of these inhibitor classes. Based on the biochemical characterization of resistant viruses and structural insight into the RSV F trimer, we propose a kinetic escape model as the origin of pan-resistance. Since a resistant RSV remained pathogenic in the mouse model, pan-resistance mutations could emerge rapidly in circulating RSV strains. We evaluate clinical implications and discuss consequences for the design of future RSV drug discovery campaigns.


Subject(s)
Antiviral Agents/chemistry , Drug Discovery/methods , Drug Resistance, Viral/genetics , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/drug effects , Virus Internalization/drug effects , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Humans , Mice , Models, Molecular , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/physiology , Small Molecule Libraries/analysis , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...