Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 107(1): 308-13, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19966279

ABSTRACT

Six-helix bundle (6HB) formation is an essential step for many viruses that rely on a class I fusion protein to enter a target cell and initiate replication. Because the binding modes of small molecule inhibitors of 6HB formation are largely unknown, precisely how they disrupt 6HB formation remains unclear, and structure-based design of improved inhibitors is thus seriously hampered. Here we present the high resolution crystal structure of TMC353121, a potent inhibitor of respiratory syncytial virus (RSV), bound at a hydrophobic pocket of the 6HB formed by amino acid residues from both HR1 and HR2 heptad-repeats. Binding of TMC353121 stabilizes the interaction of HR1 and HR2 in an alternate conformation of the 6HB, in which direct binding interactions are formed between TMC353121 and both HR1 and HR2. Rather than completely preventing 6HB formation, our data indicate that TMC353121 inhibits fusion by causing a local disturbance of the natural 6HB conformation.


Subject(s)
Antiviral Agents/metabolism , Benzimidazoles/metabolism , Pyridines/metabolism , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Amino Acid Sequence , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Cell Fusion , Crystallography, X-Ray , HeLa Cells , Humans , Membrane Fusion/physiology , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Structure, Secondary , Pyridines/chemistry , Pyridines/pharmacology , Repetitive Sequences, Amino Acid , Respiratory Syncytial Virus, Human/chemistry , Sequence Alignment , Structure-Activity Relationship , Viral Fusion Proteins/antagonists & inhibitors , Viral Fusion Proteins/genetics
2.
J Biol Chem ; 283(37): 25273-25280, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18625705

ABSTRACT

An estimated one-third of the world population is latently infected with Mycobacterium tuberculosis. These nonreplicating, dormant bacilli are tolerant to conventional anti-tuberculosis drugs, such as isoniazid. We recently identified diarylquinoline R207910 (also called TMC207) as an inhibitor of ATP synthase with a remarkable activity against replicating mycobacteria. In the present study, we show that R207910 kills dormant bacilli as effectively as aerobically grown bacilli with the same target specificity. Despite a transcriptional down-regulation of the ATP synthase operon and significantly lower cellular ATP levels, we show that dormant mycobacteria do possess residual ATP synthase enzymatic activity. This activity is blocked by nanomolar concentrations of R207910, thereby further reducing ATP levels and causing a pronounced bactericidal effect. We conclude that this residual ATP synthase activity is indispensable for the survival of dormant mycobacteria, making it a promising drug target to tackle dormant infections. The unique dual bactericidal activity of diarylquinolines on dormant as well as replicating bacterial subpopulations distinguishes them entirely from the current anti-tuberculosis drugs and underlines the potential of R207910 to shorten tuberculosis treatment.


Subject(s)
Adenosine Triphosphate/chemistry , Gene Expression Regulation, Bacterial , Homeostasis , Mycobacterium/metabolism , Quinolines/pharmacology , Antitubercular Agents/pharmacology , Mitochondrial Proton-Translocating ATPases/chemistry , Models, Biological , Mycobacterium bovis/drug effects , Mycobacterium bovis/metabolism , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Nitric Oxide/chemistry , Oxygen/chemistry , RNA, Messenger/metabolism , Time Factors
3.
J Med Chem ; 51(4): 875-96, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18254606

ABSTRACT

A preceding paper (Bonfanti et al. J. Med Chem. 2007, 50, 4572-4584) reported the optimization of the pharmacokinetic profile of substituted benzimidazoles by reducing their tissue retention. However, the modifications that were necessary to achieve this goal also led to a significant drop in anti-RSV activity. This paper describes a molecular modeling study followed by a lead optimization program that led to the recovery of the initial potent antiviral activity and the selection of TMC353121 as a clinical candidate.


Subject(s)
Antiviral Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Morpholines/chemical synthesis , Pyridines/chemical synthesis , Respiratory Syncytial Viruses/drug effects , Viral Fusion Proteins/genetics , Virus Internalization/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Binding Sites , Drug Resistance, Viral , Male , Models, Molecular , Morpholines/pharmacokinetics , Morpholines/pharmacology , Mutation , Protein Binding , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Respiratory Syncytial Viruses/physiology , Sigmodontinae , Structure-Activity Relationship
4.
J Med Chem ; 50(19): 4572-84, 2007 Sep 20.
Article in English | MEDLINE | ID: mdl-17722899

ABSTRACT

We previously reported the discovery of substituted benzimidazole fusion inhibitors with nanomolar activity against respiratory syncytial virus (Andries, K.; et al. Antiviral Res. 2003, 60, 209-219). A lead compound of the series was selected for preclinical evaluation. This drug candidate, JNJ-2408068 (formerly R170591, 1), showed long tissue retention times in several species (rat, dog, and monkey), creating cause for concern. We herein describe the optimization program to develop compounds with improved properties in terms of tissue retention. We have identified the aminoethyl-piperidine moiety as being responsible for the long tissue retention time of 1. We have investigated the replacement or the modification of this group, and we suggest that the pKa of this part of the molecules influences both the antiviral activity and the pharmacokinetic profile. We were able to identify new respiratory syncytial virus inhibitors with shorter half-lives in lung tissue.


Subject(s)
Antiviral Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Piperidines/chemical synthesis , Pyridines/chemical synthesis , Respiratory Syncytial Viruses/drug effects , Viral Fusion Proteins/antagonists & inhibitors , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , HeLa Cells , Humans , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Male , Piperidines/pharmacokinetics , Piperidines/pharmacology , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tissue Distribution
5.
Nat Chem Biol ; 3(6): 323-4, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17496888

ABSTRACT

The diarylquinoline R207910 (TMC207) is a promising candidate in clinical development for the treatment of tuberculosis. Though R207910-resistant mycobacteria bear mutations in ATP synthase, the compound's precise target is not known. Here we establish by genetic, biochemical and binding assays that the oligomeric subunit c (AtpE) of ATP synthase is the target of R207910. Thus targeting energy metabolism is a new, promising approach for antibacterial drug discovery.


Subject(s)
ATP Synthetase Complexes/metabolism , Antitubercular Agents/pharmacology , Quinolines/pharmacology , ATP Synthetase Complexes/chemistry , ATP Synthetase Complexes/drug effects , Bacterial Proteins/chemistry , Bacterial Proteins/drug effects , Bacterial Proteins/metabolism , Bacterial Proton-Translocating ATPases , Binding Sites , Diarylquinolines , Electrophoresis, Gel, Two-Dimensional , Kinetics , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Protein Subunits/drug effects , Protein Subunits/isolation & purification , Protein Subunits/metabolism
6.
Antiviral Res ; 60(3): 209-19, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14638397

ABSTRACT

A cell-based assay was used to discover compounds inhibiting respiratory syncytial virus (RSV)-induced fusion in HeLa/M cells. A lead compound was identified and subsequent synthesis of >300 analogues led to the identification of JNJ 2408068 (R170591), a low molecular weight (MW 395) benzimidazole derivative with an EC(50) (0.16 nM) against some lab strains almost 100,000 times better than that of ribavirin (15 microM). Antiviral activity was confirmed for subgroup A and B clinical isolates of human RSV and for a bovine RSV isolate. The compound did not inhibit the growth of representative viruses from other Paramyxovirus genera, i.e. HPIV2 and Mumps Virus (genus Rubulavirus), HPIV3 (genus Respirovirus), Measles virus (genus Morbillivirus) and hMPV. Efficacy in cytopathic effect inhibition assays correlated well with efficacy in virus yield reduction assays. A concentration of 10nM reduced RSV production 1000-fold in multi-cycle experiments, irrespective of the multiplicity of infection. Time of addition studies pointed to a dual mode of action: inhibition of virus-cell fusion early in the infection cycle and inhibition of cell-cell fusion at the end of the replication cycle. Two resistant mutants were raised and shown to have single point mutations in the F-gene (S398L and D486N). JNJ 2408068 was also shown to inhibit the release of proinflammatory cytokines IL-6, IL-8 and Rantes from RSV-infected A549 cells.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Respiratory Syncytial Viruses/drug effects , Antiviral Agents/chemistry , Cell Fusion , Cytokines/metabolism , Cytopathogenic Effect, Viral/drug effects , DNA Mutational Analysis , Drug Resistance, Viral/genetics , HeLa Cells , Humans , Metapneumovirus/drug effects , Metapneumovirus/growth & development , Molecular Weight , Morbillivirus/drug effects , Morbillivirus/growth & development , Point Mutation , Respiratory Syncytial Viruses/growth & development , Respiratory Syncytial Viruses/isolation & purification , Respiratory Syncytial Viruses/pathogenicity , Respirovirus/drug effects , Respirovirus/growth & development , Rubulavirus/drug effects , Rubulavirus/growth & development , Viral Fusion Proteins/genetics , Viral Plaque Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...