Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters








Publication year range
1.
Eur J Med Chem ; 229: 114087, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34998056

ABSTRACT

STING mediates innate immune responses that are triggered by the presence of cytosolic DNA. Activation of STING to boost antigen recognition is a therapeutic modality that is currently being tested in cancer patients using nucleic-acid based macrocyclic STING ligands. We describe here the discovery of 3,4-dihydroquinazolin-2(1H)-one based 6,6-bicyclic heterocyclic agonists of human STING that activate all known human variants of STING with high potency.


Subject(s)
Antineoplastic Agents/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Immunity, Innate/drug effects , Membrane Proteins/metabolism , Neoplasms/drug therapy , Small Molecule Libraries/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cytosol/chemistry , DNA/chemistry , Haplorhini , Humans , Male , Membrane Proteins/genetics , Mice, Inbred BALB C , Protein Binding , Signal Transduction , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
2.
Eur J Med Chem ; 209: 112869, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33038794

ABSTRACT

The adaptor protein STING plays a major role in innate immune sensing of cytosolic nucleic acids, by triggering a robust interferon response. Despite the importance of this protein as a potential therapeutic target for serious unmet medical conditions including cancer and infectious disease there remains a paucity of STING ligands. Starting with a benzothiazinone series of weak STING activators (human EC50 ∼10 µM) we identified several chemotypes with sub-micromolar STING activity across all the major protein polymorphs. An example compound 53 based on an oxindole core structure demonstrated robust on-target functional activation of STING (human EC50 185 nM) in immortalised and primary cells and a cytokine induction fingerprint consistent with STING activation. Our study has identified several related series of potent small molecule human STING activators with potential to be developed as immunomodulatory therapeutics.


Subject(s)
Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Membrane Proteins/agonists , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Cells, Cultured , Cytokines/metabolism , Drug Discovery , HEK293 Cells , Humans , Membrane Proteins/metabolism , Oxindoles/chemistry , Oxindoles/pharmacology , Thiazines/chemistry , Thiazines/pharmacology
3.
PLoS One ; 15(9): e0237743, 2020.
Article in English | MEDLINE | ID: mdl-32911484

ABSTRACT

The cGAS/STING pathway initiates an innate immune response when DNA is detected in the cytosol. DNA bound cGAS synthesizes cyclic dinucleotides which bind and activate the adaptor STING, leading to downstream secretion of Type I interferons and other pro-inflammatory NFκB pathway cytokines. In the mouse, the STING driven innate immune response is central to immune based clearance of various tumors and this has triggered a significant effort focused on the discovery of human STING agonists for the treatment of cancer. This report uses an in vitro kinase assay to show that G10, a previously identified STING pathway activator is actually a weak but direct STING agonist and identifies other more potent leads.


Subject(s)
Membrane Proteins/metabolism , Animals , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Interferon Regulatory Factor-3/metabolism , Membrane Proteins/chemistry , Mice , Phosphorylation , Protein Domains , Protein Stability , Signal Transduction , THP-1 Cells
4.
Proc Natl Acad Sci U S A ; 116(52): 26549-26554, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31822620

ABSTRACT

Valproic acid (VPA) is an anticonvulsant drug that is also used to treat migraines and bipolar disorder. Its proposed biological targets include human voltage-gated sodium channels, among other membrane proteins. We used the prokaryotic NavMs sodium channel, which has been shown to be a good exemplar for drug binding to human sodium channels, to examine the structural and functional interactions of VPA. Thermal melt synchrotron radiation circular dichroism spectroscopic binding studies of the full-length NavMs channel (which includes both pore and voltage sensor domains), and a pore-only construct, undertaken in the presence and absence of VPA, indicated that the drug binds to and destabilizes the channel, but not the pore-only construct. This is in contrast to other antiepileptic compounds that have previously been shown to bind in the central hydrophobic core of the pore region of the channel, and that tend to increase the thermal stability of both pore-only constructs and full-length channels. Molecular docking studies also indicated that the VPA binding site is associated with the voltage sensor, rather than the hydrophobic cavity of the pore domain. Electrophysiological studies show that VPA influences the block and inactivation rates of the NavMs channel, although with lower efficacy than classical channel-blocking compounds. It thus appears that, while VPA is capable of binding to these voltage-gated sodium channels, it has a very different mode and site of action than other anticonvulsant compounds.

5.
J Med Chem ; 62(12): 5773-5796, 2019 06 27.
Article in English | MEDLINE | ID: mdl-30964988

ABSTRACT

The design, optimization, and evaluation of a series of novel imidazopyridazine-based subtype-selective positive allosteric modulators (PAMs) for the GABAA ligand-gated ion channel are described. From a set of initial hits multiple subseries were designed and evaluated based on binding affinity and functional activity. As designing in the desired level of functional selectivity proved difficult, a probability-based assessment was performed to focus the project's efforts on a single subseries that had the greatest odds of delivering the target profile. These efforts ultimately led to the identification of two precandidates from this subseries, which were advanced to preclinical safety studies and subsequently to the identification of the clinical candidate PF-06372865.


Subject(s)
Drug Design , Imidazoles/pharmacology , Pyridazines/pharmacology , Receptors, GABA-A/metabolism , Allosteric Regulation/drug effects , Humans , Imidazoles/chemistry , Pyridazines/chemistry
6.
Science ; 363(6429): 875-880, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30792303

ABSTRACT

Potassium (K+) channels have been evolutionarily tuned for activation by diverse biological stimuli, and pharmacological activation is thought to target these specific gating mechanisms. Here we report a class of negatively charged activators (NCAs) that bypass the specific mechanisms but act as master keys to open K+ channels gated at their selectivity filter (SF), including many two-pore domain K+ (K2P) channels, voltage-gated hERG (human ether-à-go-go-related gene) channels and calcium (Ca2+)-activated big-conductance potassium (BK)-type channels. Functional analysis, x-ray crystallography, and molecular dynamics simulations revealed that the NCAs bind to similar sites below the SF, increase pore and SF K+ occupancy, and open the filter gate. These results uncover an unrecognized polypharmacology among K+ channel activators and highlight a filter gating machinery that is conserved across different families of K+ channels with implications for rational drug design.


Subject(s)
Chlorobenzenes/pharmacology , ERG1 Potassium Channel/agonists , ERG1 Potassium Channel/chemistry , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channels/agonists , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Tetrahydronaphthalenes/pharmacology , Tetrazoles/pharmacology , Thiourea/analogs & derivatives , ortho-Aminobenzoates/pharmacology , Animals , CHO Cells , Chlorobenzenes/chemistry , Cricetulus , Crystallography, X-Ray , Drug Design , HEK293 Cells , Humans , Molecular Dynamics Simulation , Protein Domains , Tetrahydronaphthalenes/chemistry , Tetrazoles/chemistry , Thiourea/chemistry , Thiourea/pharmacology , Xenopus , ortho-Aminobenzoates/chemistry
7.
Proc Natl Acad Sci U S A ; 115(48): 12301-12306, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30429323

ABSTRACT

TRPA1, a member of the transient receptor potential channel (TRP) family, is genetically linked to pain in humans, and small molecule inhibitors are efficacious in preclinical animal models of inflammatory pain. These findings have driven significant interest in development of selective TRPA1 inhibitors as potential analgesics. The majority of TRPA1 inhibitors characterized to date have been reported to interact with the S5 transmembrane helices forming part of the pore region of the channel. However, the development of many of these inhibitors as clinical drug candidates has been prevented by high lipophilicity, low solubility, and poor pharmacokinetic profiles. Identification of alternate compound interacting sites on TRPA1 provides the opportunity to develop structurally distinct modulators with novel structure-activity relationships and more desirable physiochemical properties. In this paper, we have identified a previously undescribed potent and selective small molecule thiadiazole structural class of TRPA1 inhibitor. Using species ortholog chimeric and mutagenesis strategies, we narrowed down the site of interaction to ankyrinR #6 within the distal N-terminal region of TRPA1. To identify the individual amino acid residues involved, we generated a computational model of the ankyrinR domain. This model was used predictively to identify three critical amino acids in human TRPA1, G238, N249, and K270, which were confirmed by mutagenesis to account for compound activity. These findings establish a small molecule interaction region on TRPA1, expanding potential avenues for developing TRPA1 inhibitor analgesics and for probing the mechanism of channel gating.


Subject(s)
Small Molecule Libraries/chemistry , TRPA1 Cation Channel/chemistry , TRPA1 Cation Channel/metabolism , Amino Acid Sequence , Animals , Ankyrin Repeat , Humans , Models, Molecular , Protein Binding , Rats , Sequence Alignment , Small Molecule Libraries/metabolism , TRPA1 Cation Channel/antagonists & inhibitors , TRPA1 Cation Channel/genetics
8.
Structure ; 26(4): 533-544.e3, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29576321

ABSTRACT

Small conductance potassium (SK) ion channels define neuronal firing rates by conducting the after-hyperpolarization current. They are key targets in developing therapies where neuronal firing rates are dysfunctional, such as in epilepsy, Parkinson's, and amyotrophic lateral sclerosis (ALS). Here, we characterize a binding pocket situated at the intracellular interface of SK2 and calmodulin, which we show to be shared by multiple small-molecule chemotypes. Crystallization of this complex revealed that riluzole (approved for ALS) and an analog of the anti-ataxic agent (4-chloro-phenyl)-[2-(3,5-dimethyl-pyrazol-1-yl)-pyrimidin-4-yl]-amine (CyPPA) bind to and allosterically modulate via this site. Solution-state nuclear magnetic resonance demonstrates that riluzole, NS309, and CyPPA analogs bind at this bipartite pocket. We demonstrate, by patch-clamp electrophysiology, that both classes of ligand interact with overlapping but distinct residues within this pocket. These data define a clinically important site, laying the foundations for further studies of the mechanism of action of riluzole and related molecules.


Subject(s)
Calmodulin/chemistry , Indoles/chemistry , Oximes/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , Riluzole/chemistry , Small-Conductance Calcium-Activated Potassium Channels/chemistry , Allosteric Regulation , Amino Acid Motifs , Anticonvulsants/chemistry , Anticonvulsants/metabolism , Binding Sites , Calmodulin/genetics , Calmodulin/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Indoles/metabolism , Models, Molecular , Oximes/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Pyrazoles/metabolism , Pyrimidines/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Riluzole/metabolism , Small-Conductance Calcium-Activated Potassium Channels/genetics , Small-Conductance Calcium-Activated Potassium Channels/metabolism
9.
ACS Med Chem Lett ; 8(6): 666-671, 2017 Jun 08.
Article in English | MEDLINE | ID: mdl-28626530

ABSTRACT

A series of TRPA1 antagonists is described which has as its core structure an indazole moiety. The physical properties and in vitro DMPK profiles are discussed. Good in vivo exposure was obtained with several analogs, allowing efficacy to be assessed in rodent models of inflammatory pain. Two compounds showed significant activity in these models when administered either systemically or topically. Protein chimeras were constructed to indicate compounds from the series bound in the S5 region of the channel, and a computational docking model was used to propose a binding mode for example compounds.

10.
Chem Sci ; 8(1): 40-62, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-28451148

ABSTRACT

Suzuki-Miyaura cross-coupling reactions of heteroaryl polyhalides with aryl boronates are surveyed. Drawing on data from literature sources as well as bespoke searches of Pfizer's global chemistry RKB and CAS Scifinder® databases, the factors that determine the site-selectivity of these reactions are discussed with a view to rationalising the trends found.

12.
Mol Pharm ; 13(11): 4001-4012, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27704838

ABSTRACT

Selective modulators of the γ-amino butyric acid (GABAA) family of receptors have the potential to treat a range of disease states related to cognition, pain, and anxiety. While the development of various α subunit-selective modulators is currently underway for the treatment of anxiety disorders, a mechanistic understanding of the correlation between their bioactivity and efficacy, based on ligand-target interactions, is currently still lacking. In order to alleviate this situation, in the current study we have analyzed, using ligand- and structure-based methods, a data set of 5440 GABAA modulators. The Spearman correlation (ρ) between binding activity and efficacy of compounds was calculated to be 0.008 and 0.31 against the α1 and α2 subunits of GABA receptor, respectively; in other words, the compounds had little diversity in structure and bioactivity, but they differed significantly in efficacy. Two compounds were selected as a case study for detailed interaction analysis due to the small difference in their structures and affinities (ΔpKi(comp1_α1 - comp2_α1) = 0.45 log units, ΔpKi(comp1_α2 - comp2_α2) = 0 log units) as compared to larger relative efficacies (ΔRE(comp1_α1 - comp2_α1) = 1.03, ΔRE(comp1_α2 - comp2_α2) = 0.21). Docking analysis suggested that His-101 is involved in a characteristic interaction of the α1 receptor with both compounds 1 and 2. Residues such as Phe-77, Thr-142, Asn-60, and Arg-144 of the γ chain of the α1γ2 complex also showed interactions with heterocyclic rings of both compounds 1 and 2, but these interactions were disturbed in the case of α2γ2 complex docking results. Binding pocket stability analysis based on molecular dynamics identified three substitutions in the loop C region of the α2 subunit, namely, G200E, I201T, and V202I, causing a reduction in the flexibility of α2 compared to α1. These amino acids in α2, as compared to α1, were also observed to decrease the vibrational and dihedral entropy and to increase the hydrogen bond content in α2 in the apo state. However, freezing of both α1 and α2 was observed in the ligand-bound state, with an increased number of internal hydrogen bonds and increased entropy. Therefore, we hypothesize that the amino acid differences in the loop C region of α2 are responsible for conformational changes in the protein structure compared to α1, as well as for the binding modes of compounds and hence their functional signaling.


Subject(s)
Receptors, GABA/metabolism , Amino Acid Sequence , Animals , Benzodiazepines/pharmacology , Butyric Acid/pharmacology , GABA-A Receptor Agonists/pharmacology , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Molecular Sequence Data , Principal Component Analysis , Protein Structure, Secondary , Receptors, GABA/chemistry
13.
Bioorg Med Chem Lett ; 26(20): 4919-4924, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27641472

ABSTRACT

TRESK (Twik RElated Spinal cord K+ channel) is a member of the Twin Pore Domain potassium channel (K2P) family responsible for regulating neuronal excitability in dorsal root ganglion (DRG) and trigeminal (TG) neurons, peripheral neurons involved in pain transmission. As channel opening causes an outward K+ current responsible for cell hyperpolarisation, TRESK represents a potentially interesting target for pain treatment. However, as no crystal structure exists for this protein, the mechanisms involved in the opening action of its ligands are still poorly understood, making the development of new potent and selective openers challenging. In this work we present a structure activity relationship (SAR) of the known TRESK opener flufenamic acid (FFA) and some derivatives, investigating the functional effects of chemical modifications to build a TRESK homology model to support the biological results. A plausible binding mode is proposed, providing the first predictive hypothesis of a human TRESK opener binding site.


Subject(s)
Flufenamic Acid/chemistry , Flufenamic Acid/pharmacology , Potassium Channels/chemistry , Animals , Binding Sites , HEK293 Cells , Humans , Mice , Neurons/drug effects , Structure-Activity Relationship
14.
Nature ; 537(7618): 117-121, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27556947

ABSTRACT

Ca2+ antagonist drugs are widely used in therapy of cardiovascular disorders. Three chemical classes of drugs bind to three separate, but allosterically interacting, receptor sites on CaV1.2 channels, the most prominent voltage-gated Ca2+ (CaV) channel type in myocytes in cardiac and vascular smooth muscle. The 1,4-dihydropyridines are used primarily for treatment of hypertension and angina pectoris and are thought to act as allosteric modulators of voltage-dependent Ca2+ channel activation, whereas phenylalkylamines and benzothiazepines are used primarily for treatment of cardiac arrhythmias and are thought to physically block the pore. The structural basis for the different binding, action, and therapeutic uses of these drugs remains unknown. Here we present crystallographic and functional analyses of drug binding to the bacterial homotetrameric model CaV channel CaVAb, which is inhibited by dihydropyridines and phenylalkylamines with nanomolar affinity in a state-dependent manner. The binding site for amlodipine and other dihydropyridines is located on the external, lipid-facing surface of the pore module, positioned at the interface of two subunits. Dihydropyridine binding allosterically induces an asymmetric conformation of the selectivity filter, in which partially dehydrated Ca2+ interacts directly with one subunit and blocks the pore. In contrast, the phenylalkylamine Br-verapamil binds in the central cavity of the pore on the intracellular side of the selectivity filter, physically blocking the ion-conducting pathway. Structure-based mutations of key amino-acid residues confirm drug binding at both sites. Our results define the structural basis for binding of dihydropyridines and phenylalkylamines at their distinct receptor sites on CaV channels and offer key insights into their fundamental mechanisms of action and differential therapeutic uses in cardiovascular diseases.


Subject(s)
Amines/chemistry , Amines/pharmacology , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Dihydropyridines/chemistry , Dihydropyridines/pharmacology , Allosteric Regulation/drug effects , Amines/adverse effects , Amlodipine/chemistry , Amlodipine/pharmacology , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites/drug effects , Binding Sites/genetics , Calcium/chemistry , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Line , Crystallography, X-Ray , Dihydropyridines/adverse effects , Lipids/chemistry , Models, Molecular , Moths , Mutation , Niacin/analogs & derivatives , Niacin/chemistry , Niacin/pharmacology , Protein Subunits/chemistry , Protein Subunits/metabolism , Verapamil/chemistry , Verapamil/pharmacology
15.
ACS Med Chem Lett ; 6(4): 419-24, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25893043

ABSTRACT

The transient receptor potential (TRP) family of ion channels comprises nonselective cation channels that respond to a wide range of chemical and thermal stimuli. TRPM8, a member of the melastatin subfamily, is activated by cold temperatures (<28 °C), and antagonists of this channel have the potential to treat cold induced allodynia and hyperalgesia. However, TRPM8 has also been implicated in mammalian thermoregulation and antagonists have the potential to induce hypothermia in patients. We report herein the identification and optimization of a series of TRPM8 antagonists that ultimately led to the discovery of PF-05105679. The clinical finding with this compound will be discussed, including both efficacy and its ability to affect thermoregulation processes in humans.

16.
Proc Natl Acad Sci U S A ; 111(23): 8428-33, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24850863

ABSTRACT

Voltage-gated sodium channels are important targets for the development of pharmaceutical drugs, because mutations in different human sodium channel isoforms have causal relationships with a range of neurological and cardiovascular diseases. In this study, functional electrophysiological studies show that the prokaryotic sodium channel from Magnetococcus marinus (NavMs) binds and is inhibited by eukaryotic sodium channel blockers in a manner similar to the human Nav1.1 channel, despite millions of years of divergent evolution between the two types of channels. Crystal complexes of the NavMs pore with several brominated blocker compounds depict a common antagonist binding site in the cavity, adjacent to lipid-facing fenestrations proposed to be the portals for drug entry. In silico docking studies indicate the full extent of the blocker binding site, and electrophysiology studies of NavMs channels with mutations at adjacent residues validate the location. These results suggest that the NavMs channel can be a valuable tool for screening and rational design of human drugs.


Subject(s)
Alphaproteobacteria/metabolism , Bacterial Proteins/metabolism , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Sodium Channels/metabolism , Alphaproteobacteria/chemistry , Alphaproteobacteria/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites/genetics , Crystallography, X-Ray , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Lamotrigine , Membrane Potentials/drug effects , Membrane Potentials/physiology , Models, Molecular , Molecular Sequence Data , Mutation , NAV1.1 Voltage-Gated Sodium Channel/chemistry , NAV1.1 Voltage-Gated Sodium Channel/genetics , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Sodium Channel Blockers/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/chemistry , Sodium Channels/genetics , Triazines/metabolism , Triazines/pharmacology
17.
Angew Chem Int Ed Engl ; 53(24): 6126-30, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24821300

ABSTRACT

The benzoxazinone and dihydroquinoxalinone fragments were employed as novel acetyl lysine mimics in the development of CREBBP bromodomain ligands. While the benzoxazinone series showed low affinity for the CREBBP bromodomain, expansion of the dihydroquinoxalinone series resulted in the first potent inhibitors of a bromodomain outside the BET family. Structural and computational studies reveal that an internal hydrogen bond stabilizes the protein-bound conformation of the dihydroquinoxalinone series. The side chain of this series binds in an induced-fit pocket forming a cation-π interaction with R1173 of CREBBP. The most potent compound inhibits binding of CREBBP to chromatin in U2OS cells.


Subject(s)
CREB-Binding Protein/genetics , Cations/chemistry , Epigenomics/methods , Ligands , Models, Molecular , Protein Binding
18.
ChemMedChem ; 9(7): 1378-86, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24729513

ABSTRACT

Nonstructural protein 5A (NS5A) represents a novel target for the treatment of hepatitis C virus (HCV). Daclatasvir, recently reported by Bristol-Myers-Squibb, is a potent NS5A inhibitor currently under investigation in phase 3 clinical trials. While the performance of daclatasvir has been impressive, the emergence of resistance could prove problematic and as such, improved analogues are being sought. By varying the biphenyl-imidazole unit of daclatasvir, novel inhibitors of HCV NS5A were identified with an improved resistance profile against mutant strains of the virus while retaining the picomolar potency of daclatasvir. One compound in particular, methyl ((S)-1-((S)-2-(4-(4-(6-(2-((S)-1-((methoxycarbonyl)-L-valyl)pyrrolidin-2-yl)-1H-imidazol-5-yl)quinoxalin-2-yl)phenyl)-1H-imidazol-2-yl)pyrrolidin-1-yl)-3-methyl-1-oxobutan-2-yl)carbamate (17), exhibited very promising activity and showed good absorption and a long predicted human pharmacokinetic half-life. This compound represents a promising lead that warrants further evaluation.


Subject(s)
Protease Inhibitors/chemistry , Quinoxalines/chemistry , Valine/analogs & derivatives , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Cell Line , Dogs , Drug Evaluation, Preclinical , Drug Resistance, Viral , Half-Life , Hepacivirus/metabolism , Humans , Microsomes, Liver/metabolism , Protease Inhibitors/pharmacokinetics , Quinoxalines/chemical synthesis , Quinoxalines/pharmacokinetics , Rats , Structure-Activity Relationship , Valine/chemical synthesis , Valine/chemistry , Valine/pharmacokinetics , Viral Nonstructural Proteins/metabolism
19.
ChemMedChem ; 9(7): 1387-96, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24729518

ABSTRACT

In ongoing studies towards novel hepatitis C virus (HCV) therapeutics, inhibitors of nonstructural protein 5A (NS5A) were evaluated. Specifically, starting from previously reported lead compounds, peripheral substitution patterns of a series of biaryl-linked pyrrolidine NS5A replication complex inhibitors were probed and structure-activity relationships were elucidated. Using molecular modelling and a supercritical fluid chromatographic (SFC) technique, intramolecular H-bonding and peripheral functional group topology were evaluated as key determinants of activity and membrane permeability. The novel compounds exhibited retained potency as compared with the lead compounds, and also showed promising results against a panel of resistance viruses. Together, the results of the study take us a step closer towards understanding the potency of daclatasvir, a clinical candidate upon which the compounds were based, and to designing improved analogues as second-generation antiviral agents targeting NS5A.


Subject(s)
Protease Inhibitors/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Cell Membrane Permeability/drug effects , Cell Proliferation/drug effects , Dogs , Drug Evaluation, Preclinical , Drug Resistance, Viral , Hepacivirus/metabolism , Humans , Hydrogen Bonding , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Rats , Structure-Activity Relationship , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
20.
Article in English | MEDLINE | ID: mdl-24079272

ABSTRACT

As part of a project to generate a library of nucleosides as potential antiviral agents, a small subset of novel acyclic phosphonic acid nucleosides was prepared. Practical synthetic routes are described for three targets, which were then tested against HIV, hepatitis C virus (HCV), and Dengue virus.


Subject(s)
Acids, Acyclic/chemical synthesis , Antiviral Agents/chemical synthesis , Nucleosides/chemical synthesis , Phosphorous Acids/chemical synthesis , Acids, Acyclic/chemistry , Antiviral Agents/chemistry , Dengue/drug therapy , Dengue Virus/drug effects , HIV/drug effects , HIV Infections/drug therapy , Hepacivirus/drug effects , Humans , Molecular Structure , Nucleosides/chemistry , Phosphorous Acids/chemistry , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL