Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 73
1.
Virology ; 586: 115-121, 2023 09.
Article En | MEDLINE | ID: mdl-37542818

BACKGROUND: Monoclonal antibody (palivizumab), intravenous immune globulin (IGIV), or respiratory syncytial virus (RSV)-polyclonal-hyperimmune-globulin (RSV-IG as Respigam®, RI-001, RI-002) are used with ribavirin in RSV-infected immunocompromised patients, with debated efficacy. Palivizumab-resistance (PR) can arise during treatment of persistent infections in this population. RSV-IG may confer benefit in PR-RSV infection. METHODS: RSV-IG [RI-001] was provided for an immunocompromised infant with RSV-pneumonitis refractory to ribavirin and palivizumab. RSV-neutralizing antibody, respiratory RSV load (qPCR), and F-gene-sequence-detection of PR was determined. Prophylactic RSV-IG [RI-002] or palivizumab was administered in a cotton-rat model infected with wild-type and PR-RSV. Lung RSV load and neutralizing antibody were measured. RESULTS: As protective RI-001-neutralizing antibody titers waned in the infant, a subpopulation of PR-escape mutants were detected with a fatal RSV-burden in the lungs. In PR-RSV-infected cotton rats, prophylactic RI-002 reduced RSV-load in the lungs (2.45 vs 0.28 log10 PFU/g lung-tissue reduction, respectively, p < 0.05) and provided protective RSV-neutralizing antibody. CONCLUSIONS: RSV-IG and ribavirin use in immunocompromised patients requires further study.


Drug Resistance, Viral , Palivizumab , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Palivizumab/therapeutic use , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus Infections/drug therapy , Immunocompromised Host , Animals , Sigmodontinae , Lung/pathology , Lung/virology , Immunoglobulins/administration & dosage , Antibodies, Neutralizing/administration & dosage , Female , Infant , Fatal Outcome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications
2.
Influenza Other Respir Viruses ; 17(7): e13176, 2023 07.
Article En | MEDLINE | ID: mdl-37502622

Background: Respiratory syncytial virus (RSV) infection is a cause of substantial morbidity and mortality in young children. There is currently no effective therapy available. Methods: This was a Phase 2 study of the oral RSV fusion protein inhibitor AK0529 in infants aged 1-24 months, hospitalized with RSV infection. In Part 1, patients (n = 24) were randomized 2:1 to receive a single dose of AK0529 up to 4 mg/kg or placebo. In Part 2, patients (n = 48) were randomized 2:1 to receive AK0529 at 0.5, 1, or 2 mg/kg bid or placebo for 5 days. Sparse pharmacokinetic samples were assessed using population pharmacokinetics modelling. Safety, tolerability, viral load, and respiratory signs and symptoms were assessed daily during treatment. Results: No safety or tolerability signals were detected for AK0529: grade ≥3 treatment-emergent adverse events occurring in 4.1% of patients in AK0529 and 4.2% in placebo groups, respectively, and none led to death or withdrawal from the study. In Part 2, targeted drug exposure was reached with 2 mg/kg bid. A numerically greater reduction in median viral load with 2 mg/kg bid AK0529 than with placebo at 96 h was observed. A -4.0 (95% CI: -4.51, -2.03) median reduction in Wang Respiratory Score from baseline to 96 h was observed in the 2 mg/kg group compared with -2.0 (95% CI: -3.42, -1.82) in the placebo group. Conclusions: AK0529 was well tolerated in hospitalized RSV-infected infant patients. Treatment with AK0529 2 mg/kg bid was observed to reduce viral load and Wang Respiratory Score. Clinical Trials Registration: NCT02654171.


Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Child , Infant , Humans , Child, Preschool , Respiratory Syncytial Virus Infections/epidemiology , Sulfones/pharmacology , Sulfones/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use
3.
Sci Transl Med ; 15(693): eade6422, 2023 04 26.
Article En | MEDLINE | ID: mdl-37023209

Respiratory syncytial virus (RSV) is the leading, global cause of serious respiratory disease in infants and is an important cause of respiratory illness in older adults. No RSV vaccine is currently available. The RSV fusion (F) glycoprotein is a key antigen for vaccine development, and its prefusion conformation is the target of the most potent neutralizing antibodies. Here, we describe a computational and experimental strategy for designing immunogens that enhance the conformational stability and immunogenicity of RSV prefusion F. We obtained an optimized vaccine antigen after screening nearly 400 engineered F constructs. Through in vitro and in vivo characterization studies, we identified F constructs that are more stable in the prefusion conformation and elicit ~10-fold higher serum-neutralizing titers in cotton rats than DS-Cav1. The stabilizing mutations of the lead construct (847) were introduced onto F glycoprotein backbones of strains representing the dominant circulating genotypes of the two major RSV subgroups, A and B. Immunization of cotton rats with a bivalent vaccine formulation of these antigens conferred complete protection against RSV challenge, with no evidence of disease enhancement. The resulting bivalent RSV prefusion F investigational vaccine has recently been shown to be efficacious against RSV disease in two pivotal phase 3 efficacy trials, one for passive protection of infants by immunization of pregnant women and the second for active protection of older adults by direct immunization.


Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Pregnancy , Female , Humans , Animals , Antibodies, Viral , Antibodies, Neutralizing , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/genetics , Glycoproteins , Sigmodontinae , Viral Fusion Proteins/genetics
4.
Influenza Other Respir Viruses ; 14(6): 747-756, 2020 11.
Article En | MEDLINE | ID: mdl-32790065

The controlled human infection model and specifically the human viral challenge model are not dissimilar to standard clinical trials while adding another layer of complexity and safety considerations. The models deliberately infect volunteers, with an infectious challenge agent to determine the effect of the infection and the potential benefits of the experimental interventions. The human viral challenge model studies can shorten the time to assess the efficacy of a new vaccine or treatment by combining this with the assessment of safety. The newly emerging SARS-CoV-2 virus is highly contagious, and an urgent race is on to develop a new vaccine against this virus in a timeframe never attempted before. The use of the human viral challenge model has been proposed to accelerate the development of the vaccine. In the early 2000s, the authors successfully developed a pathogenic human viral challenge model for another virus for which there was no effective treatment and established it to evaluate potential therapies and vaccines against respiratory syncytial virus. Experience gained in the development of that model can help with the development of a COVID-19 HVCM and the authors describe it here.


Betacoronavirus/pathogenicity , Coronavirus Infections/pathology , Models, Biological , Pneumonia, Viral/pathology , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , COVID-19 , COVID-19 Vaccines , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Humans , Pandemics/prevention & control , Patient Selection , Pneumonia, Viral/drug therapy , Pneumonia, Viral/immunology , Pneumonia, Viral/prevention & control , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/pathogenicity , SARS-CoV-2 , Safety , Viral Load/drug effects , Viral Vaccines/adverse effects , Viral Vaccines/therapeutic use
5.
N Engl J Med ; 383(5): 415-425, 2020 07 30.
Article En | MEDLINE | ID: mdl-32726528

BACKGROUND: Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract infection in infants, and a need exists for prevention of RSV in healthy infants. Nirsevimab is a monoclonal antibody with an extended half-life that is being developed to protect infants for an entire RSV season with a single intramuscular dose. METHODS: In this trial conducted in both northern and southern hemispheres, we evaluated nirsevimab for the prevention of RSV-associated lower respiratory tract infection in healthy infants who had been born preterm (29 weeks 0 days to 34 weeks 6 days of gestation). We randomly assigned the infants in a 2:1 ratio to receive nirsevimab, at a dose of 50 mg in a single intramuscular injection, or placebo at the start of an RSV season. The primary end point was medically attended RSV-associated lower respiratory tract infection through 150 days after administration of the dose. The secondary efficacy end point was hospitalization for RSV-associated lower respiratory tract infection through 150 days after administration of the dose. RESULTS: From November 2016 through November 2017, a total of 1453 infants were randomly assigned to receive nirsevimab (969 infants) or placebo (484 infants) at the start of the RSV season. The incidence of medically attended RSV-associated lower respiratory tract infection was 70.1% lower (95% confidence interval [CI], 52.3 to 81.2) with nirsevimab prophylaxis than with placebo (2.6% [25 infants] vs. 9.5% [46 infants]; P<0.001) and the incidence of hospitalization for RSV-associated lower respiratory tract infection was 78.4% lower (95% CI, 51.9 to 90.3) with nirsevimab than with placebo (0.8% [8 infants] vs. 4.1% [20 infants]; P<0.001). These differences were consistent throughout the 150-day period after the dose was administered and across geographic locations and RSV subtypes. Adverse events were similar in the two trial groups, with no notable hypersensitivity reactions. CONCLUSIONS: A single injection of nirsevimab resulted in fewer medically attended RSV-associated lower respiratory tract infections and hospitalizations than placebo throughout the RSV season in healthy preterm infants. (Funded by AstraZeneca and Sanofi Pasteur; ClinicalTrials.gov number, NCT02878330.).


Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antiviral Agents/administration & dosage , Infant, Premature , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human , Respiratory Tract Infections/prevention & control , Viral Fusion Proteins/antagonists & inhibitors , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Antiviral Agents/adverse effects , Female , Hospitalization/statistics & numerical data , Humans , Incidence , Infant , Infant, Newborn , Injections, Intramuscular , Kaplan-Meier Estimate , Male , Poisson Distribution , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology
6.
J Clin Virol ; 129: 104339, 2020 08.
Article En | MEDLINE | ID: mdl-32512375

Respiratory syncytial virus (RSV) immunoprophylaxis (IP) has been shown to reduce RSV hospitalization rates in high-risk infants; however, it is unclear whether RSV IP is associated with increased risk of non-RSV disease, particularly non-RSV hospitalizations. We conducted a systematic literature review to understand the occurrences of non-RSV disease and/or non-RSV hospitalizations in published studies of RSV IP. Cochrane, Embase, and PubMed databases were searched and reviewed to summarize data regarding the incidence of RSV and non-RSV respiratory disease among RSV IP recipients and controls in randomized and non-randomized studies. Independent investigators screened and selected studies for inclusion. Risk-of-bias assessment was conducted to assess strength/validity of the data using the Jadad scoring system and Downs and Black quality assessment tool, where appropriate. Twenty studies were included for review (5 randomized controlled trials [RCTs]; 15 non-randomized studies). RCTs of RSV IP demonstrated reductions in RSV hospitalizations and all-cause hospitalizations, with no increase in hospitalizations for non-RSV disease. Non-randomized studies also demonstrated reduced RSV hospitalizations in RSV IP recipients but had mixed results in assessments of hospitalizations for non-RSV disease. When RSV IP recipients and controls were more similar in disease severity risk, results of non-randomized studies aligned more closely with RCTs. Observations of increased non-RSV hospitalization rates among RSV IP recipients in some non-randomized studies could be primarily explained by differences in the clinical characteristics between RSV IP recipients and controls.


Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Databases, Factual , Hospitalization , Humans , Incidence , Infant
7.
Am J Perinatol ; 37(4): 421-429, 2020 03.
Article En | MEDLINE | ID: mdl-30991438

OBJECTIVE: The SENTINEL1 observational study characterized confirmed respiratory syncytial virus hospitalizations (RSVH) among U.S. preterm infants born at 29 to 35 weeks' gestational age (wGA) not receiving respiratory syncytial virus (RSV) immunoprophylaxis (IP) during the 2014 to 2015 and 2015 to 2016 RSV seasons. STUDY DESIGN: All laboratory-confirmed RSVH at participating sites during the 2014 to 2015 and 2015 to 2016 RSV seasons (October 1-April 30) lasting ≥24 hours among preterm infants 29 to 35 wGA and aged <12 months who did not receive RSV IP within 35 days before onset of symptoms were identified and characterized. RESULTS: Results were similar across the two seasons. Among infants with community-acquired RSVH (N = 1,378), 45% were admitted to the intensive care unit (ICU) and 19% required invasive mechanical ventilation (IMV). There were two deaths. Infants aged <6 months accounted for 78% of RSVH observed, 84% of ICU admissions, and 91% requiring IMV. Among infants who were discharged from their birth hospitalization during the RSV season, 82% of RSVH occurred within 60 days of birth hospitalization discharge. CONCLUSION: Among U.S. preterm infants 29 to 35 wGA not receiving RSV IP, RSVH are often severe with almost one-half requiring ICU admission and about one in five needing IMV.


Hospitalization/statistics & numerical data , Infant, Premature, Diseases/epidemiology , Infant, Premature , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus, Human , Antiviral Agents/therapeutic use , Community-Acquired Infections/epidemiology , Female , Humans , Infant , Infant, Newborn , Infant, Premature, Diseases/prevention & control , Infant, Premature, Diseases/therapy , Intensive Care Units, Pediatric , Male , Multivariate Analysis , Odds Ratio , Palivizumab/therapeutic use , Respiration, Artificial , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/therapy , United States/epidemiology
8.
PLoS One ; 14(4): e0214708, 2019.
Article En | MEDLINE | ID: mdl-30986239

Many aspects of the respiratory syncytial virus (RSV) are still poorly understood. Yet these knowledge gaps have had and could continue to have adverse, unintended consequences for the efficacy and safety of antivirals and vaccines developed against RSV. Mathematical modelling was used to test and evaluate hypotheses about the rate of loss of RSV infectivity and the mechanisms and kinetics of RSV infection spread in SIAT cells in vitro. While the rate of loss of RSV integrity, as measured via qRT-PCR, is well-described by an exponential decay, the latter mechanism failed to describe the rate at which RSV A Long loses infectivity over time in vitro based on the data presented herein. This is unusual given that other viruses (HIV, HCV, influenza) have been shown to lose their infectivity exponentially in vitro, and indeed an exponential rate of loss of infectivity is always assumed in mathematical modelling and experimental analyses. The infectivity profile of RSV in HEp-2 and SIAT cells remained consistent over the course of an RSV infection, over time and a large range of infectivity. However, SIAT cells were found to be ∼ 100× less sensitive to RSV infection than HEp-2 cells. In particular, we found that RSV spreads inefficiently in SIAT cells, in a manner we show is consistent with the establishment of infection resistance in uninfected cells. SIAT cells are a good in vitro model in which to study RSV in vivo dissemination, yielding similar infection timescales. However, the higher sensitivity of HEp-2 cells to RSV together with its RSV infectivity profile being similar to that of SIAT cells, makes HEp-2 cells more suitable for quantifying RSV infectivity over the course of in vitro RSV infections in SIAT cells. Our findings highlight the importance and urgency of resolving the mechanisms at play in the dissemination of RSV infections in vitro, and the processes by which this infectivity is lost.


Models, Theoretical , Respiratory Syncytial Virus, Human/physiology , Cell Line , Humans , Influenza A virus/physiology , Kinetics , RNA, Viral/analysis , Real-Time Polymerase Chain Reaction , Respiratory Syncytial Virus, Human/genetics
9.
J Antimicrob Chemother ; 74(2): 442-452, 2019 02 01.
Article En | MEDLINE | ID: mdl-30376079

Background: Respiratory syncytial virus (RSV) causes high morbidity, with mortality rates approaching or exceeding that of influenza in adult and infant patient populations, respectively. Lumicitabine (ALS-008176 or JNJ-64041575) is an oral nucleoside analogue prodrug in clinical development to treat RSV infections. This prodrug converts to plasma-circulating ALS-8112, and then to the 5'-active nucleoside triphosphate (NTP) form within host cells. We conducted an RSV-A challenge study in healthy adults to evaluate lumicitabine's activity during an active RSV infection. Objectives: To develop a semi-mechanistic mathematical model describing RSV kinetics, and the pharmacokinetics (PK) and pharmacodynamics (PD) of lumicitabine during treatment. Methods: Nasopharyngeal viral load and concentrations of ALS-8112 and ALS-8144 (uridine metabolite) were measured frequently over the study duration. Population viral kinetic and PK/PD models were developed using NONMEM. The RSV life-cycle was described using a target-cell-limited model that included a physiological delay. Results: The estimated clearances of ALS-8112 and ALS-8144 were 54.2 and 115 L/h/70 kg, respectively. A semi-physiological model was linked to predict ALS-8112 conversion to active intracellular NTP. Extensive and rapid RSV reduction occurred after lumicitabine treatment (EC50 = 1.79 µM), with >99% viral inhibition at 2 h after loading dose. Simulated NTP exposures and time to EC50 attainment suggested that rapid therapeutic effects and reduced dosing frequency are achievable in adult and paediatric patients. Conclusions: The semi-mechanistic model characterizes RSV kinetics and the antiviral effectiveness of lumicitabine in an adult challenge population. This model is applicable to guide dose selection in adult and paediatric patients.


Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Deoxycytidine/analogs & derivatives , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Virus Replication/drug effects , Adult , Antiviral Agents/blood , Deoxycytidine/blood , Deoxycytidine/pharmacokinetics , Deoxycytidine/therapeutic use , Double-Blind Method , Healthy Volunteers , Humans , Models, Theoretical , Nasopharynx/virology , Respiratory Syncytial Virus, Human/physiology , Viral Load/drug effects
10.
J Clin Virol ; 107: 29-37, 2018 10.
Article En | MEDLINE | ID: mdl-30170224

BACKGROUND: Herpes Simplex Virus encephalitis (HSVE) is a devastating disease of all ages. Rigorous studies correlating viral load with neuroradiological and clinical severity have not been performed, particularly in neonates. Understanding these relationships may improve therapies. OBJECTIVES: To correlate molecularly quantified HSV in cerebrospinal fluid (CSF) and disease severity. STUDY DESIGN: HSV loads (VL) were evaluated by real-time PCR from the CSF of 33 patients (20 neonates, 5 children, 8 adults) with HSVE. We studied relationships between CSF VL and structural and volumetric brain abnormalities (MRI); hospital morbidity; and discharge and long-term (>3 month) clinical outcomes. RESULTS: Initial CSF VL did not differ in neonates vs non-neonates (median 4.6 vs 5.1 log10 copies/mL, p = 0.75). Initial CSF VL was higher in neonates with HSV-2 vs HSV-1 (median 4.8 vs 3.2 log10 copies/mL, respectively, p = 0.02). Persistently detectable DNA in CSF despite acyclovir trended towards higher odds of unfavorable outcome at discharge for neonates [0.87 (CI 0.75-1), p = 0.07]. Initial VL correlated with higher CSF protein concentrations for the cohort and for neonates (p = 0.03 and 0.01, respectively), but not with lesion volume or subarachnoid exposure of involved brain (p all >0.05), hospital morbidity (p all >0.05), nor with higher odds of unfavorable discharge or long-term outcomes for the cohort [OR = 0.9(CI 0.5-1.6), p = 0.72; OR = 1.0(CI 0.5-1.8), p = 0.9] or for neonates [OR = 1.3(CI 0.5-3.3), p = 0.57; OR = 2.3(CI 0.7-8), p = 0.2]. CONCLUSIONS: Initial HSV VL did not predict neuroradiological or clinical outcomes in patients with HSVE, suggesting host inflammatory factors contribute to disease in treated patients with good viral clearance.


Encephalitis, Herpes Simplex/cerebrospinal fluid , Herpesvirus 1, Human/isolation & purification , Herpesvirus 2, Human/isolation & purification , Viral Load/methods , Acyclovir/therapeutic use , Adolescent , Adult , Brain/pathology , Brain/virology , Child , Child, Preschool , DNA, Viral/cerebrospinal fluid , Encephalitis, Herpes Simplex/drug therapy , Female , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/genetics , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Polymerase Chain Reaction , Severity of Illness Index , Young Adult
11.
Antiviral Res ; 158: 103-112, 2018 10.
Article En | MEDLINE | ID: mdl-30086337

Antiviral therapy can lead to drug resistance, but multiple factors determine the frequency of drug resistance mutations and the clinical consequences. When chronic infections caused by Human Immunodeficiency Virus (HIV), Hepatitis C Virus (HCV) and Hepatitis B Virus (HBV) are compared with acute infections such as influenza virus, respiratory syncytial virus (RSV), and other respiratory viruses, there are similarities in how and why antiviral resistance substitutions occur, but the clinical significance can be quite different. Emergence of resistant variants has implications for design of new therapeutics, treatment guidelines, clinical trial design, resistance monitoring, reporting, and interpretation. In this discussion paper, we consider the molecular factors contributing to antiviral drug resistance substitutions, and a comparison is made between chronic and acute infections. The implications of resistance are considered for clinical trial endpoints and public health, as well as the requirements for therapeutic monitoring in clinical practice with acute viral infections.


Antiviral Agents/pharmacology , Drug Resistance, Viral , Virus Diseases/drug therapy , Viruses/drug effects , Biological Evolution , Drug Resistance, Viral/genetics , HIV/drug effects , HIV Infections/drug therapy , HIV Infections/virology , Hepacivirus/drug effects , Hepatitis B/drug therapy , Hepatitis B/virology , Hepatitis B virus/drug effects , Hepatitis C/drug therapy , Hepatitis C/virology , Humans , Mutation , Mutation Rate , Orthomyxoviridae/drug effects , Respiratory Syncytial Viruses/drug effects , Virus Diseases/virology , Virus Replication/drug effects
12.
MMWR Morb Mortal Wkly Rep ; 67(30): 825-828, 2018 Aug 03.
Article En | MEDLINE | ID: mdl-30070981

Angiostrongyliasis is caused by infection and migration to the brain of larvae of the parasitic nematode Angiostrongylus cantonensis, or rat lungworm. Adult A. cantonensis reside in the lungs of the definitive wild rodent host, where they produce larvae passed in feces, which are then ingested by snails and slugs (gastropods). Human infection typically occurs when gastropods containing mature larvae are inadvertently ingested by humans. Although human infection often is asymptomatic or involves transient mild symptoms, larval migration to the brain can lead to eosinophilic meningitis, focal neurologic deficits, coma, and death. The majority of cases of human angiostrongyliasis occur in Asia and the Pacific Islands, including Hawaii, but autochthonous and imported cases have been reported in the continental United States (1,2), underscoring the importance of provider recognition to ensure prompt identification and treatment. The epidemiologic and clinical features of 12 angiostrongyliasis cases in the continental United States were analyzed. These cases were identified through A. cantonensis polymerase chain reaction (PCR) testing (3) of cerebrospinal fluid (CSF) submitted to CDC from within the continental United States. Six cases were likely a result of autochthonous transmission in the southern United States. All 12 patients had CSF pleocytosis and eosinophilia, consistent with eosinophilic meningitis. Health care providers need to be aware of the possibility of angiostrongyliasis in patients with eosinophilic meningitis, especially in residents in the southern United States or persons who have traveled outside the continental United States and have a history of ingestion of gastropods or contaminated raw vegetables.


Angiostrongylus cantonensis/isolation & purification , Central Nervous System Diseases/epidemiology , Strongylida Infections/complications , Strongylida Infections/diagnosis , Adolescent , Adult , Aged , Angiostrongylus cantonensis/genetics , Animals , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , United States/epidemiology , Young Adult
13.
Infect Control Hosp Epidemiol ; 39(9): 1074-1079, 2018 09.
Article En | MEDLINE | ID: mdl-30019659

OBJECTIVE: To describe an adenovirus outbreak in a neonatal intensive care unit (NICU), including the use of qualitative and semiquantitative real-time polymerase chain reaction (qPCR) data to inform the outbreak response. DESIGN: Mixed prospective and retrospective observational study. SETTING: A level IV NICU in the southeastern United States.PatientsTwo adenovirus cases were identified in a NICU. Screening of all inpatients with qPCR on nasopharyngeal specimens revealed 11 additional cases.InterventionsOutbreak response procedures, including enhanced infection control policies, were instituted. Serial qPCR studies were used to screen for new infections among exposed infants and to monitor viral clearance among cases. Changes to retinopathy of prematurity (ROP) exam procedures were made after an association was noted in those patients. At the end of the outbreak, a retrospective review allowed for comparison of clinical factors between the infected and uninfected groups. RESULTS: There were no new cases among patients after outbreak identification. One adenovirus-infected patient died; the others recovered their clinical baselines. The ROP exams were associated with an increased risk of infection (odds ratio [OR], 84.6; 95% confidence interval [CI], 4.5-1,601). The duration of the outbreak response was 33 days, and the previously described second wave of cases after the end of the outbreak did not occur. Revisions to infection control policies remained in effect following the outbreak. CONCLUSIONS: Retinopathy of prematurity exams are potential mechanisms of adenovirus transmission, and autoclaved or single-use instruments should be used to minimize this risk. Real-time molecular diagnostic and quantification data guided outbreak response procedures, which rapidly contained and fully terminated a NICU adenovirus outbreak.


Adenovirus Infections, Human/transmission , Disease Outbreaks , Infection Control/methods , Neonatal Screening/adverse effects , Real-Time Polymerase Chain Reaction , Retinopathy of Prematurity/diagnosis , Adenoviruses, Human/classification , Cross Infection/virology , Female , Humans , Infant, Newborn , Intensive Care Units, Neonatal , Male , Neonatal Screening/methods , Prospective Studies , Retrospective Studies , Serogroup , Tennessee
15.
Emerg Infect Dis ; 23(10): 1756-1758, 2017 10.
Article En | MEDLINE | ID: mdl-28930003

Angiostrongylus cantonensis, the rat lungworm, is the most common infectious cause of eosinophilic meningoencephalitis worldwide. This parasite is endemic to Southeast Asia and the Pacific Islands, and its global distribution is increasing. We report A. cantonensis meningoencephalitis in a 12-month-old boy in Tennessee, USA, who had not traveled outside of southwestern Tennessee or northwestern Mississippi.


Angiostrongylus cantonensis/isolation & purification , Eosinophilia/diagnosis , Meningoencephalitis/diagnosis , Strongylida Infections/diagnosis , Adrenal Cortex Hormones/therapeutic use , Albendazole/therapeutic use , Angiostrongylus cantonensis/drug effects , Angiostrongylus cantonensis/immunology , Angiostrongylus cantonensis/pathogenicity , Animals , Antibodies, Helminth/blood , Eosinophilia/drug therapy , Eosinophilia/parasitology , Humans , Infant , Male , Meningoencephalitis/drug therapy , Meningoencephalitis/parasitology , Rats , Strongylida Infections/drug therapy , Strongylida Infections/parasitology , Strongylida Infections/transmission
16.
Pediatr Res ; 82(5): 872-880, 2017 Nov.
Article En | MEDLINE | ID: mdl-28723886

BackgroundLongitudinal respiratory syncytial virus (RSV) dynamics have not been well studied despite the existence of factors favoring prolonged RSV replication including high mutation rates allowing rapid evolution and potential escape from immune control. We therefore measured viral load in previously RSV-naive infants over prolonged time spans.MethodsDuring 2014-2015, quantitative nasal aspirates were collected from 51 RSV-PCR+ infants. Multiple parallel assessments of viral loads were quantified at each collected time point using a well-validated real-time quantitative reverse transcriptase polymerase chain reaction assay. After observing viral load rebound phenomenon in some infants, the viral dynamics of 27 infants with sufficient longitudinal viral load data points were analyzed using the pre-defined criteria for viral rebound. Additional analyses were performed comparing age with viral rebound, viral clearance rates, and viral load area-under-the-curve (AUCVL).ResultsThe 51 infants (303 nasal aspirate samples; mean of 5.9 per patient) exhibited slower than expected viral clearance. Lower age trended toward slower viral clearance and greater AUCVL. Six infants had detectable viral loads ≥1 month after symptom onset. Ten of twenty-seven evaluable subjects exhibited viral rebound and this rebound was age-dependent (P=0.0259). All but one rebounder were <70 days old.ConclusionInfants struggle to control primary RSV infections allowing prolonged viral replication and previously undescribed viral rebound; likely representing viral mutational immune escape.


Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/growth & development , Virus Replication , DNA, Viral/genetics , Female , Host-Pathogen Interactions , Humans , Infant , Infant, Newborn , Kinetics , Longitudinal Studies , Male , Nasal Cavity/virology , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus, Human/genetics , Viral Load
17.
Article En | MEDLINE | ID: mdl-28652242

Although respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract infection in infants and young children, attempts to develop an effective therapy have so far proved unsuccessful. Here we report the preclinical profiles of PC786, a potent nonnucleoside RSV L protein polymerase inhibitor, designed for inhalation treatment of RSV infection. PC786 demonstrated a potent and selective antiviral activity against laboratory-adapted or clinical isolates of RSV-A (50% inhibitory concentration [IC50], <0.09 to 0.71 nM) and RSV-B (IC50, 1.3 to 50.6 nM), which were determined by inhibition of cytopathic effects in HEp-2 cells without causing detectable cytotoxicity. The underlying inhibition of virus replication was confirmed by PCR analysis. The effects of PC786 were largely unaffected by the multiplicity of infection (MOI) and were retained in the face of established RSV replication in a time-of-addition study. Persistent anti-RSV effects of PC786 were also demonstrated in human bronchial epithelial cells. In vivo intranasal once daily dosing with PC786 was able to reduce the virus load to undetectable levels in lung homogenates from RSV-infected mice and cotton rats. Treatment with escalating concentrations identified a dominant mutation in the L protein (Y1631H) in vitro In addition, PC786 potently inhibited RSV RNA-dependent RNA polymerase (RdRp) activity in a cell-free enzyme assay and minigenome assay in HEp-2 cells (IC50, 2.1 and 0.5 nM, respectively). Thus, PC786 was shown to be a potent anti-RSV agent via inhibition of RdRp activity, making topical treatment with this compound a novel potential therapy for the treatment of human RSV infections.


Antiviral Agents/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Respiratory Tract Infections/drug therapy , Spiro Compounds/pharmacology , Virus Replication/drug effects , Animals , Benzamides , Benzazepines , Cell Line , Epithelial Cells/virology , Humans , Mice , Rats , Respiratory Mucosa/virology , Respiratory Tract Infections/virology , Viral Load/drug effects , Viral Proteins/biosynthesis
19.
Transpl Infect Dis ; 19(2)2017 Apr.
Article En | MEDLINE | ID: mdl-28054734

BACKGROUND: Respiratory syncytial virus (RSV) may cause fatal lower respiratory tract infection (LRTI) in immunocompromised patients. Ribavirin with or without standard intravenous immunoglobulin (IVIG) is frequently given although efficacy is debated. Infusion of IVIG with high levels of neutralizing antibody against RSV may offer benefit in these patients. METHODS: RI-001 contains standardized levels of high-titer anti-RSV neutralizing antibody and was provided for compassionate use to 15 patients with RSV LRTI who either failed conventional therapy or had significant risk of progression. Patients were treated on day 1 with RI-001 1500 mg/kg, followed 2 days later with 750 mg/kg. Pre- and post-infusion sera were measured for RSV neutralizing antibody. Patient data were analyzed for safety related to infusion of RI-001, and clinical outcomes. RESULTS: Patients ranged in age from 2 months to 71 years and 80% had hematologic malignancy or were bone marrow or hematopoietic stem cell transplant recipients. Administration was well tolerated. Pre-infusion neutralizing titers ranged from 51 to 1765 geometric mean titer (mean 646±519) and all patients demonstrated at least a 4-fold rise (mean 6410±4470) 5-10 days post infusion. Eleven of 15 improved and were discharged from the hospital. Days from positive RSV test to RI-001 treatment was shorter in survivors compared to non-survivors (4.4±2.8 vs. 20.3±21.0 days, P=.02). CONCLUSION: Administration of RI-001 was well tolerated and resulted in significant increases in serum neutralizing antibody titers to RSV. Our data suggest that early identification of RSV and treatment with RI-001 may offer benefit.


Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Antiviral Agents/therapeutic use , Drugs, Investigational/therapeutic use , Immunoglobulins, Intravenous/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/immunology , Respiratory Tract Infections/drug therapy , Adolescent , Adult , Aged , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/adverse effects , Antibodies, Viral/administration & dosage , Antibodies, Viral/adverse effects , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Child , Child, Preschool , Compassionate Use Trials , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Female , Humans , Immunocompromised Host , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/adverse effects , Infant , Infusions, Intravenous , Investigational New Drug Application , Male , Middle Aged , Respiratory Syncytial Virus Infections/virology , Respiratory Tract Infections/virology , Treatment Outcome , Young Adult
20.
Am J Perinatol ; 34(1): 51-61, 2017 01.
Article En | MEDLINE | ID: mdl-27233106

Objective SENTINEL1 characterized U.S. preterm infants 29 to 35 weeks' gestational age (wGA) < 12 months old hospitalized for laboratory-confirmed respiratory syncytial virus (RSV) disease and not receiving RSV immunoprophylaxis during the 2014 to 2015 RSV season. Study Design This is a noninterventional, observational, cohort study. Results A total of 702 infants were hospitalized with community-acquired RSV disease, of whom an estimated 42% were admitted to the intensive care unit (ICU) and 20% required invasive mechanical ventilation (IMV). Earlier gestational age and younger chronologic age were associated with an increased frequency of RSV-confirmed hospitalization (RSVH), ICU admission, and IMV. Among infants 29 to 32 wGA and < 3 months of age, 68% required ICU admission and 44% required IMV. One death occurred of an infant 29 wGA. Among the 212 infants enrolled for in-depth analysis of health care resource utilization, mean and median RSVH charges were $55,551 and $27,461, respectively, which varied by intensity of care required. Outpatient visits were common, with 63% and 62% of infants requiring visits before and within 1 month following the RSVH, respectively. Conclusion Preterm infants 29 to 35 wGA are at high risk for severe RSV disease, which imposes a substantial health burden, particularly in the first months of life.


Ambulatory Care/statistics & numerical data , Hospital Costs , Hospitalization/statistics & numerical data , Respiratory Syncytial Virus Infections/epidemiology , Ambulatory Care/economics , Antiviral Agents/therapeutic use , Cohort Studies , Female , Gestational Age , Health Care Costs , Humans , Infant , Infant, Newborn , Infant, Premature , Intensive Care Units, Pediatric , Male , Palivizumab/therapeutic use , Respiration, Artificial , Respiratory Syncytial Virus Infections/prevention & control , United States/epidemiology
...