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.
J Med Chem ; 67(16): 13723-13736, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39105710

ABSTRACT

Respiratory syncytial virus (RSV) is an RNA virus infecting the upper and lower respiratory tract and is recognized as a major respiratory health threat, particularly to older adults, immunocompromised individuals, and young children. Around 64 million children and adults are infected every year worldwide. Despite two vaccines and a new generation monoclonal antibody recently approved, no effective antiviral treatment is available. In this manuscript, we present the medicinal chemistry efforts resulting in the identification of compound 28 (JNJ-8003), a novel RSV non-nucleoside inhibitor displaying subnanomolar activity in vitro as well as prominent efficacy in mice and a neonatal lamb models.


Subject(s)
Antiviral Agents , Pyridines , Animals , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , Humans , Mice , Pyridines/pharmacology , Pyridines/chemistry , Pyridines/chemical synthesis , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/virology , Structure-Activity Relationship , Sheep , Drug Discovery , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Viruses/drug effects
2.
Antiviral Res ; 227: 105907, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38772503

ABSTRACT

Respiratory syncytial virus (RSV) can cause pulmonary complications in infants, elderly and immunocompromised patients. While two vaccines and two prophylactic monoclonal antibodies are now available, treatment options are still needed. JNJ-7184 is a non-nucleoside inhibitor of the RSV-Large (L) polymerase, displaying potent inhibition of both RSV-A and -B strains. Resistance selection and hydrogen-deuterium exchange experiments suggest JNJ-7184 binds RSV-L in the connector domain. JNJ-7184 prevents RSV replication and transcription by inhibiting initiation or early elongation. JNJ-7184 is effective in air-liquid interface cultures and therapeutically in neonatal lambs, acting to drastically reverse the appearance of lung pathology.


Subject(s)
Antiviral Agents , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Virus Replication , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/virology , Animals , Humans , Virus Replication/drug effects , Respiratory Syncytial Virus, Human/drug effects , Sheep , Drug Resistance, Viral , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Viral Proteins/genetics , Lung/virology
3.
Adv Sci (Weinh) ; 11(12): e2306729, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38225749

ABSTRACT

Respiratory syncytial virus (RSV) causes substantial morbidity and mortality in infants, the immunocompromised, and the elderly. RSV infects the airway epithelium via the apical membrane and almost exclusively sheds progeny virions back into the airway mucus (AM), making RSV difficult to target by systemically administered therapies. An inhalable "muco-trapping" variant of motavizumab (Mota-MT), a potent neutralizing mAb against RSV F is engineered. Mota-MT traps RSV in AM via polyvalent Fc-mucin bonds, reducing the fraction of fast-moving RSV particles in both fresh pediatric and adult AM by ≈20-30-fold in a Fc-glycan dependent manner, and facilitates clearance from the airways of mice within minutes. Intranasal dosing of Mota-MT eliminated viral load in cotton rats within 2 days. Daily nebulized delivery of Mota-MT to RSV-infected neonatal lambs, beginning 3 days after infection when viral load is at its maximum, led to a 10 000-fold and 100 000-fold reduction in viral load in bronchoalveolar lavage and lung tissues relative to placebo control, respectively. Mota-MT-treated lambs exhibited reduced bronchiolitis, neutrophil infiltration, and airway remodeling than lambs receiving placebo or intramuscular palivizumab. The findings underscore inhaled delivery of muco-trapping mAbs as a promising strategy for the treatment of RSV and other acute respiratory infections.


Subject(s)
Antibodies, Monoclonal , Respiratory Syncytial Virus Infections , Humans , Infant , Child , Animals , Sheep , Mice , Aged , Antibodies, Monoclonal/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Palivizumab/therapeutic use , Respiratory Syncytial Viruses , Lung
4.
PLoS One ; 16(3): e0235026, 2021.
Article in English | MEDLINE | ID: mdl-33705390

ABSTRACT

Respiratory syncytial virus (RSV) is the primary cause of viral bronchiolitis resulting in hospitalization and a frequent cause of secondary respiratory bacterial infection, especially by Streptococcus pneumoniae (Spn) in infants. While murine studies have demonstrated enhanced morbidity during a viral/bacterial co-infection, human meta-studies have conflicting results. Moreover, little knowledge about the pathogenesis of emerging Spn serotype 22F, especially the co-pathologies between RSV and Spn, is known. Here, colostrum-deprived neonate lambs were divided into four groups. Two of the groups were nebulized with RSV M37, and the other two groups were mock nebulized. At day three post-RSV infection, one RSV group (RSV/Spn) and one mock-nebulized group (Spn only) were inoculated with Spn intratracheally. At day six post-RSV infection, bacterial/viral loads were assessed along with histopathology and correlated with clinical symptoms. Lambs dually infected with RSV/Spn trended with higher RSV titers, but lower Spn. Additionally, lung lesions were observed to be more frequent in the RSV/Spn group characterized by increased interalveolar wall thickness accompanied by neutrophil and lymphocyte infiltration and higher myeloperoxidase. Despite lower Spn in lungs, co-infected lambs had more significant morbidity and histopathology, which correlated with a different cytokine response. Thus, enhanced disease severity during dual infection may be due to lesion development and altered immune responses rather than bacterial counts.


Subject(s)
Pneumococcal Infections/pathology , Respiratory Syncytial Virus Infections/pathology , Streptococcus pneumoniae/isolation & purification , Animals , Animals, Newborn , Bronchoalveolar Lavage Fluid/microbiology , Bronchoalveolar Lavage Fluid/virology , Cytokines/metabolism , Disease Models, Animal , Lung/microbiology , Lung/pathology , Lung/virology , Lymphocytes/cytology , Lymphocytes/immunology , Neutrophils/cytology , Neutrophils/immunology , Peroxidase/metabolism , Pneumococcal Infections/complications , Pneumococcal Infections/epidemiology , Pneumococcal Infections/microbiology , RNA, Viral/metabolism , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/isolation & purification , Serogroup , Sheep , Streptococcus pneumoniae/genetics
5.
Cell Tissue Res ; 380(2): 313-324, 2020 May.
Article in English | MEDLINE | ID: mdl-32347384

ABSTRACT

Many animal models have been established for respiratory syncytial virus (RSV) infection of infants with the purpose of studying the pathogenesis, immunological response, and pharmaceutical testing and the objective of finding novel therapies and preventive measures. This review centers on a neonatal lamb model of RSV infection that has similarities to RSV infection of infants. It includes a comprehensive description of anatomical and immunological similarities between ovine and human lungs along with comparison of pulmonary changes and immune responses with RSV infection. These features make the newborn lamb an effective model for investigating key aspects of RSV infection in infants. The importance of RSV lamb model application in preclinical therapeutic trials and current updates on new studies with the RSV-infected neonatal lamb are also highlighted.


Subject(s)
Respiratory Syncytial Virus Infections/diagnosis , Animals , Animals, Newborn , Disease Models, Animal , Humans
6.
Nat Commun ; 8(1): 167, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28761099

ABSTRACT

Respiratory syncytial virus is a major cause of acute lower respiratory tract infection in young children, immunocompromised adults, and the elderly. Intervention with small-molecule antivirals specific for respiratory syncytial virus presents an important therapeutic opportunity, but no such compounds are approved today. Here we report the structure of JNJ-53718678 bound to respiratory syncytial virus fusion (F) protein in its prefusion conformation, and we show that the potent nanomolar activity of JNJ-53718678, as well as the preliminary structure-activity relationship and the pharmaceutical optimization strategy of the series, are consistent with the binding mode of JNJ-53718678 and other respiratory syncytial virus fusion inhibitors. Oral treatment of neonatal lambs with JNJ-53718678, or with an equally active close analog, efficiently inhibits established acute lower respiratory tract infection in the animals, even when treatment is delayed until external signs of respiratory syncytial virus illness have become visible. Together, these data suggest that JNJ-53718678 is a promising candidate for further development as a potential therapeutic in patients at risk to develop respiratory syncytial virus acute lower respiratory tract infection.Respiratory syncytial virus causes lung infections in children, immunocompromised adults, and in the elderly. Here the authors show that a chemical inhibitor to a viral fusion protein is effective in reducing viral titre and ameliorating infection in rodents and neonatal lambs.


Subject(s)
Imidazolidines/metabolism , Indoles/metabolism , Respiratory Syncytial Virus, Human/metabolism , Viral Fusion Protein Inhibitors/metabolism , Viral Fusion Proteins/metabolism , Animals , Animals, Newborn , Cell Line, Tumor , Chlorocebus aethiops , Epithelial Cells , Humans , Imidazolidines/pharmacology , Imidazolidines/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Molecular Structure , Pneumonia, Viral/drug therapy , Rats , Respiratory Mucosa/cytology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/metabolism , Sheep , Structure-Activity Relationship , Vero Cells , Viral Fusion Protein Inhibitors/pharmacology , Viral Fusion Protein Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL