Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.806
Filter
1.
Virol J ; 21(1): 235, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39350281

ABSTRACT

BACKGROUND: Cell-penetrating peptides (CPPs) are effective for delivering therapeutic molecules with minimal toxicity. This study focuses on the use of penetratin, a well-characterized CPP, to deliver a DNA vector encoding short hairpin RNA (shRNA) targeting the respiratory syncytial virus (RSV) F gene into infected cells. RSV is known to cause severe lower respiratory infections in infants and poses significant risks to immunocompromised individuals and the elderly. We evaluated the antiviral efficacy of the penetratin-shRNA complex by comparing its ability to inhibit RSV replication and induce apoptosis with ribavirin treatment. METHODS: Penetratin-shRNA complexes were prepared at different ratios and analyzed using gel retardation assays, dynamic light scattering, and zeta potential measurements. The complexes were tested in HEp-2 and A549 cells for transfection efficiency, cytotoxicity, viral load, and apoptosis using plaque assays, real-time reverse transcription-polymerase chain reaction (RT-PCR), DNA fragmentation, propidium iodide staining, and caspase 3/7 activation assays. RESULTS: The gel shift assay determined that a 20:1 CPP-to-shRNA ratio was optimal for effective complexation, resulting in particles with a size of 164 nm and a zeta potential of 8.7 mV. Transfection efficiency in HEp-2 cells was highest at this ratio, reaching up to 93%. The penetratin-shRNA complex effectively silenced the RSV F gene, reduced viral titers, and decreased DNA fragmentation and apoptosis in infected cells. CONCLUSION: Penetratin effectively delivers shRNA targeting the RSV F gene, significantly reducing viral load and preventing apoptosis without toxicity. This approach surpasses Lipofectamine and shows potential for future therapeutic interventions, especially when combined with ribavirin, against RSV infection.


Subject(s)
Antiviral Agents , Apoptosis , Cell-Penetrating Peptides , RNA, Small Interfering , Virus Replication , Humans , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/chemistry , Apoptosis/drug effects , Antiviral Agents/pharmacology , Virus Replication/drug effects , RNA, Small Interfering/genetics , Cell Line , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/physiology
2.
Vaccine ; 42(24): 126294, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-39241353

ABSTRACT

Respiratory syncytial virus (RSV) causes substantial morbidity and mortality in older adults. In May 2023, two subunit RSV vaccines (Arexvy [GSK] and Abrysvo [Pfizer]) received approval from the U.S. Food and Drug Administration (FDA). In June 2023, ACIP recommended that adults aged ≥60 years may receive a single dose of RSV vaccine, using shared clinical decision-making. In support of development of this policy, our objective was to assess the cost-effectiveness of RSV vaccination in the general population in this age group. We used a decision-analytical model of RSV over a two-year timeframe using data from published literature, FDA documents, epidemiological databases, and manufacturer data. We tracked RSV-associated outpatient, emergency department, inpatient healthcare utilization, RSV-attributable deaths, quality-adjusted life-years lost (QALYs), and societal costs. The societal cost per QALY saved from RSV vaccination depended on age group and product: adults aged ≥60 years, $196,842 for GSK's vaccine and $176,557 for Pfizer's vaccine; adults ≥65 years, $162,138 for GSK and $146,543 for Pfizer; adults 60- <65 years, $385,829 for GSK and $331,486 for Pfizer. Vaccine efficacy, incidence of RSV hospitalization, and vaccine cost had the greatest influence on cost per QALY. Cost per QALY saved decreased as the age of those vaccinated increased. Inputs such as long-term efficacy are uncertain. RSV vaccination in adults aged ≥60 years may be cost-effective, particularly in those of more advanced age. Lower vaccine acquisition costs and persistent efficacy beyond two RSV seasons would render RSV vaccination more cost-effective for a broader target population. PRIMARY FUNDING SOURCE: US Centers for Disease Control and Prevention.


Subject(s)
Cost-Benefit Analysis , Quality-Adjusted Life Years , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Vaccination , Humans , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/economics , Respiratory Syncytial Virus Vaccines/economics , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus Vaccines/administration & dosage , Aged , Middle Aged , Vaccination/economics , Vaccination/methods , Respiratory Syncytial Virus, Human/immunology , Male , Aged, 80 and over , Female , United States/epidemiology
3.
Vaccine ; 42(24): 126276, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-39241352

ABSTRACT

BACKGROUND: Nirsevimab is an extended half-life monoclonal antibody (mAb) licensed for the prevention of respiratory syncytial virus (RSV)-associated lower respiratory tract disease in neonates, infants and medically vulnerable children. We characterized RSV isolates recovered from participants enrolled in MEDLEY: a randomized, palivizumab-controlled phase 2/3 trial of nirsevimab in infants born preterm and/or with congenital heart disease or chronic lung disease of prematurity. METHODS: Participants were assessed in two RSV seasons (Season 1 and 2). Season 1 participants were randomized (2:1) to receive a single dose of nirsevimab (50 mg if weight <5 kg or 100 mg if weight ≥5 kg in Season 1; 200 mg in Season 2) followed by four monthly doses of placebo, or five once-monthly doses of palivizumab (15 mg/kg weight per dose). Season 2 participants continued nirsevimab and placebo (nirsevimab/nirsevimab) or were re-randomized (1:1) to switch to nirsevimab (palivizumab/nirsevimab) or continue palivizumab (palivizumab/palivizumab). Cases of RSV infection were identified by central testing of nasal swabs from participants seeking medical attention for respiratory illnesses. Nirsevimab and palivizumab binding site substitutions were assessed via microneutralization assay. RESULTS: Twenty-five cases of confirmed RSV infection were observed during the trial and sequenced: 12 in nirsevimab recipients and 10 in palivizumab recipients during Season 1, and 1 case in each Season 2 group. Molecular sequencing of RSV A (n = 14) isolates detected no nirsevimab binding site substitutions, and 3 palivizumab neutralization-resistant substitutions (Lys272Met, Lys272Thr, Ser275Leu). The nirsevimab binding site Ile206Met:Gln209Arg and Ile206Met:Gln209Arg:Ser211Asn substitutions were the only anti-RSV mAb binding site substitutions detected among RSV B isolates (n = 11). Nirsevimab neutralized all nirsevimab and palivizumab binding site substitutions in RSV A and B isolates recovered from MEDLEY participants. CONCLUSION: No binding site substitution detected during MEDLEY affected RSV susceptibility to nirsevimab neutralization.


Subject(s)
Antibodies, Monoclonal, Humanized , Antiviral Agents , Palivizumab , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Palivizumab/therapeutic use , Palivizumab/administration & dosage , Respiratory Syncytial Virus Infections/prevention & control , Infant , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antiviral Agents/therapeutic use , Antiviral Agents/administration & dosage , Double-Blind Method , Male , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/genetics , Female , Infant, Newborn , Antibodies, Viral/immunology , Child, Preschool , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood
4.
BMC Infect Dis ; 24(1): 1009, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300365

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV), a leading cause of lower respiratory tract infection (LRTI) among children, has resurged in the form of endemic or even pandemic in many countries and areas after the easing of COVID-19 containment measures. This study aimed to investigate the differences in epidemiological and clinical characteristics of children hospitalized for RSV infection during pre- and post-COVID-19 eras in Yunnan, China. METHODS: A total of 2553 pediatric RSV inpatients from eight hospitals in Yunnan were retrospectively enrolled in this study, including 1451 patients admitted in 2018-2019 (pre-COVID-19 group) and 1102 patients admitted in 2023 (post-COVID-19 group). According to the presence or absence of severe LRTI (SLRTI), patients in the pre- and post-COVID-19 groups were further divided into the respective severe or non-severe subgroups, thus analyzing the risk factors for RSV-associated SLRTI in the two eras. Demographic, epidemiological, clinical, and laboratory data of the patients were collected for the final analysis. RESULTS: A shift in the seasonal pattern of RSV activity was observed between the pre-and post-COVID-19 groups. The peak period of RSV hospitalizations in the pre-COVID-19 group was during January-April and October-December in both 2018 and 2019, whereas that in the post-COVID-19 group was from April to September in 2023. Older age, more frequent clinical manifestations (fever, acute otitis media, seizures), and elevated laboratory indicators [neutrophil-to-lymphocyte ratio (NLR), c-reactive protein (CRP), interleukin 6 (IL-6), co-infection rate] were identified in the post-COVID-19 group than those in the pre-COVID-19 group (all P < 0.05). Furthermore, compared to the pre-COVID-19 group, the post-COVID-19 group displayed higher rates of SLRTI and mechanical ventilation, with a longer length of hospital stay (all P < 0.05). Age, low birthweight, preterm birth, personal history of atopy, underlying condition, NLR, IL-6 were the shared independent risk factors for RSV-related SLRTI in both pre- and post-COVID-19 groups, whereas seizures and co-infection were independently associated with SLRTI only in the post-COVID-19 group. CONCLUSIONS: An off-season RSV endemic was observed in Yunnan during the post-COVID-19 era, with changed clinical features and increased severity. Age, low birthweight, preterm birth, personal history of atopy, underlying condition, NLR, IL-6, seizures, and co-infection were the risk factors for RSV-related SLRTI in the post-COVID-19 era.


Subject(s)
COVID-19 , Hospitalization , Respiratory Syncytial Virus Infections , Humans , Retrospective Studies , Respiratory Syncytial Virus Infections/epidemiology , COVID-19/epidemiology , Female , Male , Infant , Child, Preschool , China/epidemiology , Hospitalization/statistics & numerical data , Child , Risk Factors , SARS-CoV-2 , Respiratory Syncytial Virus, Human , Seasons , Infant, Newborn , Adolescent
5.
Lancet ; 404(10458): 1143-1156, 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39265587

ABSTRACT

The global burden of respiratory syncytial virus (RSV) lower respiratory tract infection (LRTI) in young children is high. The RSV prevention strategies approved in 2023 will be essential to lowering the global disease burden. In this Series paper, we describe clinical presentation, burden of disease, hospital management, emerging therapies, and targeted prevention focusing on developments and groundbreaking publications for RSV. We conducted a systematic search for literature published in the past 15 years and used a non-systematic approach to analyse the results, prioritising important papers and the most recent reviews per subtopic. Annually, 33 million episodes of RSV LRTI occur in children younger than 5 years, resulting in 3·6 million hospitalisations and 118 200 deaths. RSV LRTI is a clinical diagnosis but a clinical case definition and universal clinical tool to predict severe disease are non-existent. The advent of molecular point-of-care testing allows rapid and accurate confirmation of RSV infection and could reduce antibiotic use. There is no evidence-based treatment of RSV, only supportive care. Despite widespread use, evidence for high-flow nasal cannula (HFNC) therapy is insufficient and increased paediatric intensive care admissions and intubation indicate the need to remove HFNC therapy from standard care. RSV is now a vaccine-preventable disease in young children with a market-approved long-acting monoclonal antibody and a maternal vaccine targeting the RSV prefusion protein. To have a high impact on life-threatening RSV infection, infants at high risk, especially in low-income and middle-income countries, should be prioritised as an interim strategy towards universal immunisation. The implementation of RSV preventive strategies will clarify the full burden of RSV infection. Vaccine probe studies can address existing knowledge gaps including the effect of RSV prevention on transmission dynamics, antibiotic misuse, the respiratory microbiome composition, and long-term sequalae.


Subject(s)
Antiviral Agents , Respiratory Syncytial Virus Infections , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Antiviral Agents/therapeutic use , Cost of Illness , Hospitalization , Respiratory Syncytial Virus Infections/therapy , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/therapy , Respiratory Tract Infections/virology
6.
Lancet ; 404(10458): 1157-1170, 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39265588

ABSTRACT

The search for safe and efficacious products to prevent severe respiratory syncytial virus (RSV) disease in young infants has lasted more than 60 years. In high-income and middle-income countries, two new products have been authorised: an RSV monoclonal antibody for administration to infants (nirsevimab) and an RSV prefusion F maternal vaccine (RSVpreF [Pfizer, Puurs, Belgium]) for administration to pregnant people. These products are not yet available in low-income and lower-middle-income countries, where most RSV deaths occur. Other papers in this Series describe the acute burden of RSV disease in young children, the effects of RSV infection in early childhood on long-term lung health, and the burden of RSV disease and disease prevention products in older adults. In this Series paper, we briefly review the efficacy, effectiveness, and safety of nirsevimab and RSVpreF maternal vaccine for protection of infants. We then explore potential regulatory, policy, and implementation pathways and provide case studies of intervention uptake in Spain and Argentina, and considerations for use in Kenya. We also explore the health economic evidence to inform product introduction decisions. With sufficient political will and affordable pricing, RSV disease prevention in infants can become a global reality.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Humans , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Infant , Female , Pregnancy , Respiratory Syncytial Virus, Human/immunology , Vaccination , Child, Preschool
7.
Physiol Genomics ; 56(10): 691-697, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39222066

ABSTRACT

The severity of respiratory syncytial virus (RSV) may be linked to host genetic susceptibility. Surfactant protein (SP) genetic variants have been associated with RSV severity, but the impact of single-nucleotide polymorphism (SNP)-SNP interactions remains unexplored. Therefore, we used a novel statistical model to investigate the association of SNP-SNP interactions of SFTP genes with RSV severity in two- and three-interaction models. We analyzed available genotype and clinical data from prospectively enrolled 405 children diagnosed with RSV, categorizing them into moderate or severe RSV groups. Using Wang's statistical model, we studied significant associations of SNP-SNP interactions with RSV severity in a case-control design. We observed, first, association of three interactions with increased risk of severe RSV in a two-SNP model. One intragenic interaction was between SNPs of SFTPA2, and the other two were intergenic, involving SNPs of hydrophilic and hydrophobic SPs alone. We also observed, second, association of 22 interactions with RSV severity in a three-SNP model. Among these, 20 were unique, with 12 and 10 interactions associated with increased or decreased risk of RSV severity, respectively, and included at least one SNP of either SFTPA1 or SFTPA2. All interactions were intergenic except one, among SNPs of SFTPA1. The remaining interactions were either among SNPs of hydrophilic SPs alone (n = 8) or among SNPs of both hydrophilic or hydrophobic SPs (n = 11). Our findings indicate that SNPs of all SFTPs may contribute to genetic susceptibility to RSV severity. However, the predominant involvement of SFTPA1 and/or SFTPA2 SNPs in these interactions underscores their significance in RSV severity.NEW & NOTEWORTHY Although surfactant protein (SP) genetic variants are associated with respiratory syncytial virus (RSV) severity, the impact of single-nucleotide polymorphism (SNP)-SNP interactions of SP genes remained unexplored. Using advanced statistical models, we uncovered 22 SNP-SNP interactions associated with RSV severity, with notable involvement of SFTPA1 and SFTPA2 SNPs. This highlights the comprehensive role of all SPs in genetic susceptibility to RSV severity, shedding light on potential avenues for targeted interventions.


Subject(s)
Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Respiratory Syncytial Virus Infections , Severity of Illness Index , Humans , Respiratory Syncytial Virus Infections/genetics , Polymorphism, Single Nucleotide/genetics , Female , Male , Infant , Genetic Predisposition to Disease/genetics , Case-Control Studies , Pulmonary Surfactant-Associated Protein A/genetics , Child, Preschool , Prospective Studies , Genotype , Child , Respiratory Syncytial Virus, Human/genetics , Infant, Newborn
8.
MMWR Morb Mortal Wkly Rep ; 73(38): 837-843, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39325675

ABSTRACT

Respiratory syncytial virus (RSV) is the most common cause of hospitalization among U.S. infants. CDC recommends RSV vaccination for pregnant persons or administration of RSV antibody (nirsevimab) to infants aged <8 months to prevent RSV lower respiratory tract disease among infants. To determine maternal and infant RSV immunization coverage for the 2023-24 RSV season, CDC conducted an Internet panel survey during March 26-April 11, 2024. Among 678 women at 32-36 weeks' gestation during September 2023-January 2024, 32.6% reported receipt of an RSV vaccine any time during pregnancy. Among 866 women with an infant born during August 2023-March 2024, 44.6% reported receipt of nirsevimab by the infant. Overall, 55.8% of infants were protected by maternal RSV vaccine, nirsevimab, or both. Provider recommendation for maternal vaccination or infant nirsevimab was associated with higher immunization coverage, whereas lack of a provider recommendation was the main reason for not getting RSV immunization. The main reason for definitely or probably not getting nirsevimab for infants was concern about the long-term safety for the infant. Activities supporting providers to make RSV prevention recommendations and have informative conversations with patients might increase the proportion of infants protected against severe RSV disease. CDC and the American College of Obstetricians and Gynecologists have resources to assist providers in effectively communicating the importance of immunization.


Subject(s)
Antibodies, Monoclonal, Humanized , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Humans , United States , Female , Respiratory Syncytial Virus Infections/prevention & control , Infant , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/immunology , Pregnancy , Antibodies, Monoclonal, Humanized/therapeutic use , Adult , Antibodies, Viral/blood , Vaccination/statistics & numerical data , Young Adult , Infant, Newborn , Respiratory Syncytial Virus, Human/immunology
9.
Science ; 385(6716): 1484-1491, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39325881

ABSTRACT

The respiratory syncytial virus (RSV) fusion (F) glycoprotein is highly immunogenic in its prefusion (pre-F) conformation. However, the protein is unstable, and its conformation must be stabilized for it to function effectively as an immunogen in vaccines. We present a mutagenesis strategy to arrest the RSV F protein in its pre-F state by blocking localized changes in protein structure that accompany large-scale conformational rearrangements. We generated a series of mutants and screened them in vitro to assess their potential for forming a stable pre-F. In animals, the immunogenicity of a representative mutant F protein, with a conformation confirmed by cryo-electron microscopy, elicited levels of neutralizing antibodies and protection against RSV-induced lung damage that were comparable to those of DS-Cav1, a pre-F used in a licensed vaccine.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Viral Fusion Proteins , Animals , Humans , Mice , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cryoelectron Microscopy , Immunogenicity, Vaccine , Lung/virology , Mice, Inbred BALB C , Mutagenesis , Mutation , Protein Conformation , Protein Stability , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/immunology , Viral Fusion Proteins/genetics
11.
Viruses ; 16(9)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39339830

ABSTRACT

Recent studies have documented prolonged expression of viral antigens and RNA and associated inflammation after infection with SARS-CoV-2 in a substantial proportion of infected patients. The persisting SARS-CoV-2 effects and findings, with inflammation associated with continued detection of viral antigens, especially resemble those previously reported for influenza virus, as well as respiratory syncytial virus (RSV). The reports indicate the need for improved insight into the mechanisms whereby post-SARS-CoV-2 infection-related illness is apparently more common and perhaps even more persistent after infection than observed for other respiratory viruses.


Subject(s)
Antigens, Viral , COVID-19 , Inflammation , Respiratory Syncytial Virus Infections , SARS-CoV-2 , Humans , COVID-19/immunology , COVID-19/virology , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Antigens, Viral/immunology , Inflammation/virology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/immunology , Orthomyxoviridae/immunology , Orthomyxoviridae/genetics , Influenza, Human/virology , Influenza, Human/immunology , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/genetics
12.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39337288

ABSTRACT

Respiratory syncytial virus (RSV) is a common respiratory pathogen that causes respiratory illnesses, ranging from mild symptoms to severe lower respiratory tract infections in infants and older adults. This virus is responsible for one-third of pneumonia deaths in the pediatric population; however, there are currently only a few effective vaccines. A better understanding of the RSV-host relationship at the molecular level may lead to a more effective management of RSV-related symptoms. The fractalkine (CX3CL1) receptor (CX3CR1) is a co-receptor for RSV expressed by airway epithelial cells and diverse immune cells. RSV G protein binds to the CX3CR1 receptor via a highly conserved amino acid motif (CX3C motif), which is also present in CX3CL1. The CX3CL1-CX3CR1 axis is involved in the activation and infiltration of immune cells into the infected lung. The presence of the RSV G protein alters the natural functions of the CX3CR1-CX3CL1 axis and modifies the host's immune response, an aspects that need to be considered in the development of an efficient vaccine and specific pharmacological treatment.


Subject(s)
CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/metabolism , Chemokine CX3CL1/metabolism , CX3C Chemokine Receptor 1/metabolism , Respiratory Syncytial Virus, Human/immunology , Animals
13.
Front Cell Infect Microbiol ; 14: 1445115, 2024.
Article in English | MEDLINE | ID: mdl-39220282

ABSTRACT

The human respiratory syncytial virus (RSV) is a significant health concern, particularly for infants, young children, and the elderly. This virus is known to evolve continuously due to environmental factors and herd immunity. In light of this, our study aimed to analyze the genetic variability of the G protein in RSV-A and RSV-B genotypes in Kuwait from 2020 to 2022. Between January 2020 and September 2022, we collected 490 respiratory samples from hospitalized patients with acute respiratory tract infections. These samples were tested and confirmed positive for RSV using multiplex Real-Time PCR. Subsequently, the samples underwent nucleic acid sequencing using the advanced Nanopore sequencing technology to analyze the full-length G gene. Sequence analysis showed that 64 isolates (76%) were RSV-A, and 20 isolates (24%) were RSV-B. The G genes of RSV-A belonged to genotype GA2.3.5, while all the RSV-B genotypes belonged to GB5.0.5a. New lineages and sub-lineages of RSV-A and RSV-B were detected, indicating the circulation of new strains in Kuwait. Many unique and new amino acid changes, including insertions, were found in the G proteins of Kuwaiti isolates, with the highest variability in the second hypervariable region. An increased number of N and O-linked glycosylation sites were also identified in the G protein, which could speculate to alter the antigenicity of RSV. The identified changes in the G protein of RSV-A and RSV-B genotypes might result from immune pressure and could affect the antigenic characteristics of circulating strains in Kuwait. This could potentially lead to new RSV variants that can evade the immune response. Our in-depth analysis of the G proteins of both RSV-A and RSV-B could aid in the development of more potent treatments and vaccines.


Subject(s)
Genetic Variation , Genotype , Phylogeny , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Kuwait , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/classification , Respiratory Syncytial Virus, Human/isolation & purification , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus Infections/virology , Glycosylation , Infant , Child, Preschool , Female , Male , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Adult , Child , Middle Aged , Aged , Young Adult , Adolescent , Respiratory Tract Infections/virology , Viral Envelope Proteins/genetics
14.
Cleve Clin J Med ; 91(9 suppl 1): S19-S25, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231601

ABSTRACT

Respiratory syncytial virus (RSV) is a threat to infants globally causing bronchiolitis and pneumonia. Despite decades of research, RSV outbreaks occur with only modest advancements in prevention or treatment. Vaccine development faced challenges because past attempts caused enhanced disease and treatment options demonstrated limited efficacy. Recent advancements, including maternal vaccines focusing on the prefusion form of the F glycoprotein are now showing significant benefits in preventing severe RSV-related illness in infants. Additionally, monoclonal antibodies offer prevention directly to newborns within 1 week of birth. These innovations have the potential to substantially change the impact of RSV.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Humans , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/therapy , Infant , Respiratory Syncytial Virus, Human , Infant, Newborn , Child , Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use
15.
Influenza Other Respir Viruses ; 18(9): e70008, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39284784

ABSTRACT

BACKGROUND: Older adults in nursing and care homes (NCHs) are vulnerable to severe respiratory syncytial virus (RSV) infection, hospitalization, and death. This study aimed to gather data on RSV disease among older adults in NCHs and identify reported risk factors for RSV hospitalization and case fatality. METHODS: The study protocol was registered in PROSPERO (CRD42022371908). We searched MEDLINE, EMBASE, and Global Health databases to identify articles published between 2000 and 2023. Observational and experimental studies conducted among older adults in NCHs requiring assistive care and reporting RSV illness were included and relevant data were extracted. RESULTS: Of 18,690 studies screened, 32 were selected for full-text review, and 20 were included. Overall, the number of NCH residents ranged from 42 to 1459 with a mean age between 67.6 and 85 years. Attack rates ranged from 6.7% to 47.6% and annual incidence ranged from 0.5% to 14%. Case fatality rates ranged from 7.7% to 23.1%. We found similar annual incidence rates of RSV-positive acute respiratory infection (ARI) of 4582 (95% CI: 3259-6264) and 4785 (95% CI: 2258-10,141) per 100,000 reported in two studies. Annual incidence rate of RSV-positive lower respiratory tract infection was 3040 (95% CI: 1986-4454) cases per 100,000 adults. Annual RSV-ARI hospital admission rates were between 600 (95% CI: 190-10,000) and 1104 (95% CI: 350-1930) per 100,000 person-years. Among all RSV disease cases, commonly reported chronic medical conditions included chronic obstructive pulmonary disease (COPD), heart failure, ischemic heart disease, coronary artery disease, hypertension, diabetes, kidney dysfunction, cerebrovascular accident, malignancies, dementia, and those with a Charlson comorbidity score > 6.5. CONCLUSION: Data on RSV infection among NCH residents are limited and largely heterogeneous but document a high risk of illness, frequent hospitalization, and high mortality. Preventive interventions, such as vaccination, should be considered for this high-risk population. Nationally representative epidemiologic studies and NCH-based viral pathogen surveillance could more precisely assess the burden on NCH residents.


Subject(s)
Hospitalization , Nursing Homes , Respiratory Syncytial Virus Infections , Humans , Respiratory Syncytial Virus Infections/epidemiology , Nursing Homes/statistics & numerical data , Aged , Hospitalization/statistics & numerical data , Aged, 80 and over , Incidence , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/isolation & purification , Risk Factors , Male , Female , Cost of Illness
16.
EBioMedicine ; 107: 105318, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39217853

ABSTRACT

Severe respiratory syncytial virus (RSV) disease is a significant contributor to the global burden of disease in infants and children. The RSV attachment protein (G) has been shown to be critical in invading airway epithelial cells through its CX3C motif interacting with the host receptor CX3CR1. The ubiquitous expression of this receptor on immune cells may explain their susceptibility to RSV infection. The RSV G protein may enhance disease severity through reprogramming of normal cellular functionality leading to inhibition of antiviral responses. While existing preventives targeting the RSV fusion (F) protein are highly effective, there are no RSV therapeutics based on the G protein to limit RSV pathogenesis. Monoclonal antibodies targeting the RSV G protein administered as post-infection therapeutics in mice have been shown to improve the antiviral response, reduce viral load and limit disease severity. Further research is required to better understand how RSV infection of immune cells contributes to pathogenesis for the development of more targeted and efficacious therapeutics.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/metabolism , Animals , Respiratory Syncytial Virus, Human/immunology , Host-Pathogen Interactions/immunology , CX3C Chemokine Receptor 1/metabolism , Viral Fusion Proteins/immunology , Viral Fusion Proteins/metabolism , Mice , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology
17.
Front Cell Infect Microbiol ; 14: 1435294, 2024.
Article in English | MEDLINE | ID: mdl-39286815

ABSTRACT

Objectives: We aim to investigate the prevalence of respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) among pediatric patients with acute respiratory tract illness (ARTI) in southern China both pre- and post-COVID-19 pandemic, as well as identify associated risk factors for severe infections. Methods: The study conducted a real-time PCR analysis on hospitalized children with ARTI from 2012 to 2023, specifically targeting RSV, hMPV, and other respiratory pathogens. Additionally, demographic data was collected during this analysis. Results: The prevalence of RSV occurs triennially, and likewise, the temporal pattern of hMPV outbreaks mirrors that of RSV. The peak infection rates of RSV and hMPV occurred during and following the implementation of COVID-19 epidemic prevention and control measures. The incidence of RSV infection exhibited bimodal peaks in 2022, while hMPV demonstrated seasonal peaks during the spring, fall, and winter periods post-COVID-19 pandemic. After the COVID-19 outbreak, there has been an upward trend in the proportion of female patients and patients aged one year and older presenting with ARTI, RSV infections, and hMPV infections. Infant (OR = 4.767, 95%CI: [3.888-5.846], p < 0.0001), presence of co-infection (OR = 0.540, 95%CI: [0.404-0.722], p < 0.0001), and existence of comorbidities (OR = 1.582, 95%CI: [1.285-1.949], p < 0.0001) was the risk ratio for the severity of RSV infection. Children infected with hMPV under the age of 1 year (OR = 0.322, 95%CI: [0.180 - 0.575], p < 0.0001), as well as those with comorbidities (OR = 8.809, 95%CI: [4.493 - 17.272], p < 0.0001), have a higher risk of developing severe illness. Conclusion: The changing epidemiological patterns have the potential to lead to widespread severe outbreaks among children, particularly those with underlying medical conditions who may experience more severe symptoms. Conducting surveillance for pneumoviridae viruses in children is an imperative measure to establish a robust foundation for future epidemic prevention and treatment strategies.


Subject(s)
COVID-19 , Metapneumovirus , Paramyxoviridae Infections , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Metapneumovirus/genetics , Metapneumovirus/isolation & purification , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/virology , Infant , Female , Male , Child, Preschool , Paramyxoviridae Infections/epidemiology , Paramyxoviridae Infections/virology , Retrospective Studies , China/epidemiology , Child , COVID-19/epidemiology , COVID-19/virology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/isolation & purification , Prevalence , Risk Factors , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Adolescent , Seasons , Infant, Newborn , Incidence , Severity of Illness Index
18.
BMC Public Health ; 24(1): 2410, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232690

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) is one of the main causes of hospitalization for lower respiratory tract infection in children under five years of age globally. Maternal vaccines and monoclonal antibodies for RSV prevention among infants are approved for use in high income countries. However, data are limited on the economic burden of RSV disease from low- and middle-income countries (LMIC) to inform decision making on prioritization and introduction of such interventions. This study aimed to estimate household and health system costs associated with childhood RSV in Kenya. METHODS: A structured questionnaire was administered to caregivers of children aged < 5 years admitted to referral hospitals in Kilifi (coastal Kenya) and Siaya (western Kenya) with symptoms of acute lower respiratory tract infection (LRTI) during the 2019-2021 RSV seasons. These children had been enrolled in ongoing in-patient surveillance for respiratory viruses. Household expenditures on direct and indirect medical costs were collected 10 days prior to, during, and two weeks post hospitalization. Aggregated health system costs were acquired from the hospital administration and were included to calculate the cost per episode of hospitalized RSV illness. RESULTS: We enrolled a total of 241 and 184 participants from Kilifi and Siaya hospitals, respectively. Out of these, 79 (32.9%) in Kilifi and 21(11.4%) in Siaya, tested positive for RSV infection. The total (health system and household) mean costs per episode of severe RSV illness was USD 329 (95% confidence interval (95% CI): 251-408 ) in Kilifi and USD 527 (95% CI: 405- 649) in Siaya. Household costs were USD 67 (95% CI: 54-80) and USD 172 (95% CI: 131- 214) in Kilifi and Siaya, respectively. Mean direct medical costs to the household during hospitalization were USD 11 (95% CI: 10-12) and USD 67 (95% CI: 51-83) among Kilifi and Siaya participants, respectively. Observed costs were lower in Kilifi due to differences in healthcare administration. CONCLUSIONS: RSV-associated disease among young children leads to a substantial economic burden to both families and the health system in Kenya. This burden may differ between Counties in Kenya and similar multi-site studies are advised to support cost-effectiveness analyses.


Subject(s)
Hospitalization , Respiratory Syncytial Virus Infections , Respiratory Tract Infections , Humans , Kenya/epidemiology , Respiratory Syncytial Virus Infections/economics , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/therapy , Child, Preschool , Infant , Female , Male , Hospitalization/economics , Hospitalization/statistics & numerical data , Respiratory Tract Infections/economics , Respiratory Tract Infections/therapy , Respiratory Tract Infections/virology , Health Care Costs/statistics & numerical data , Cost of Illness , Surveys and Questionnaires , Respiratory Syncytial Virus, Human , Health Expenditures/statistics & numerical data , Infant, Newborn
19.
Euro Surveill ; 29(39)2024 Sep.
Article in English | MEDLINE | ID: mdl-39328156

ABSTRACT

BackgroundRespiratory syncytial virus (RSV) is a leading cause of acute respiratory infections and hospitalisations in infants (age < 1 year) and young children. Little is known on RSV epidemiology and related inpatient healthcare resource use (HCRU) in Switzerland.AimTo explore RSV-related hospitalisations, inpatient HCRU and medical costs in all age groups, and risk factors for infant hospitalisations in Switzerland.MethodsWe used national hospital registry data from 2003 to 2021 identifying RSV cases with ICD-10-GM codes, and described demographic characteristics, HCRU and associated medical costs of RSV inpatients. The effect of risk factors on infant hospitalisation was estimated with logistic regression.ResultsWe observed a general increase and biannual pattern in RSV hospitalisations between 2003/04 and 2018/19, with 3,575 hospitalisations in 2018/19 and 2,487 in 2019/20 before numbers declined in 2020/21 (n = 902). Around two thirds of all hospitalisations occurred in infants. Mean (median) age was 118 (85) days in hospitalised infants and 74 (77) years in hospitalised adult patients (> 18 years); 7.2% of cases required intensive care unit stay. Mean inpatient medical costs were estimated at EUR 8,046. Most (90.8%) hospitalised infants with RSV were born after 35 weeks of gestation without bronchopulmonary dysplasia or congenital heart disease. Low birth weight, gestational age and congenital disorders were associated with a higher risk for hospitalisation.ConclusionsRSV leads to a substantial number of hospitalisations and peaks in hospital capacity utilisation. Measures to protect all infants from an RSV hospitalisation are essential in addressing this public health challenge.


Subject(s)
Hospitalization , Inpatients , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/economics , Switzerland/epidemiology , Infant , Hospitalization/statistics & numerical data , Hospitalization/economics , Female , Male , Child, Preschool , Child , Adult , Middle Aged , Adolescent , Respiratory Syncytial Virus, Human/isolation & purification , Aged , Risk Factors , Infant, Newborn , Inpatients/statistics & numerical data , Young Adult , Registries , Aged, 80 and over , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Respiratory Tract Infections/economics , Health Care Costs/statistics & numerical data , Cost of Illness , Length of Stay/statistics & numerical data
20.
Emerg Infect Dis ; 30(10): 2199-2202, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39320243

ABSTRACT

In a population-based birth cohort study of respiratory syncytial virus surveillance in the United States, 897/1,680 (53.4%) children were infected during infancy; 25 (2.8%) of those were hospitalized. Among symptomatic infants, 143/324 (44.1%) had lower respiratory tract infections. These data provide benchmarks to monitor effects of maternal vaccines and extended half-life monoclonal antibodies.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/virology , United States/epidemiology , Risk Factors , Infant , Prevalence , Female , Infant, Newborn , Male , Birth Cohort
SELECTION OF CITATIONS
SEARCH DETAIL