Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 282
Filter
2.
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.
Pediatrics ; 154(4)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39257372

ABSTRACT

BACKGROUND AND OBJECTIVES: In the 2023-2024 respiratory syncytial virus (RSV) season, Spain became one of the first countries to introduce universal RSV prophylaxis, during which all infants born at this time were eligible to receive nirsevimab. Locally, most Spanish regions also immunized infants younger than age 6 months at the start of the season (extended catch-up). The aim of this study was to assess how RSV prophylaxis affected the number of infants presenting to pediatric emergency departments with acute respiratory infections. METHODS: A retrospective study was conducted in 15 Spanish pediatric emergency departments from 9 different regions between the 2018 and 2024 epidemic seasons (November-January). We compared the seasons occurring in 2018-2023 and the 2023-2024 season regarding the number of episodes of lower respiratory tract infection and acute bronchiolitis, acute bronchiolitis-related hospital admissions, and PICU admissions. RESULTS: A comparison with the average rates for the previous epidemic seasons revealed a 57.7% decrease in episodes of lower respiratory tract infection in 2023-2024 (95% CI, 56.5-58.8; P < .001; range among hospitals, 4.8-82.8), a 59.2% decrease in episodes of acute bronchiolitis (95% CI, 57.9-60.4; P < .001; range, 6.9-84.1), a 63.1% reduction in acute bronchiolitis-related hospital admissions (95% CI, 60.9-65.2; P < .001; range, 31.4-86.8), and a 63.1% reduction in PICU admissions (95% CI, 58.1-67.9; P < .001; range, 18.2-81.8). Hospitals in regions applying extended catch-up showed better results. CONCLUSIONS: Nirsevimab can protect a broad infant population against RSV infection with high effectiveness. Approaches including extended catch-up are the most effective, although cost- effectiveness must be considered.


Subject(s)
Antiviral Agents , Bronchiolitis , Emergency Service, Hospital , Respiratory Syncytial Virus Infections , Humans , Retrospective Studies , Emergency Service, Hospital/statistics & numerical data , Infant , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/drug therapy , Bronchiolitis/epidemiology , Bronchiolitis/drug therapy , Antiviral Agents/therapeutic use , Acute Disease , Spain/epidemiology , Female , Male , Palivizumab/therapeutic use , Hospitalization/statistics & numerical data , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/prevention & control , Antibodies, Monoclonal, Humanized/therapeutic use , Infant, Newborn
6.
Hum Vaccin Immunother ; 20(1): 2380110, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39118394

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection (LRTI) in infants and young children worldwide. Using routine statutory health insurance claims data including patients from all regions of Germany, we investigated the health-care resource use and costs associated with RSV prophylaxis with palivizumab in Germany. In the database, infants from the birth cohorts 2015-2019 eligible for palivizumab immunization were identified using codes of the 10th revision of the International Classification of Diseases (ICD-10). Health-care resource use and costs related to immunization were determined by inpatient and outpatient administrations. Over the study period, only 1.3% of infants received at least one dose of palivizumab in their first year of life. The mean number of doses per immunized infant was 4.6. From a third-party payer perspective, the mean costs of palivizumab per infant who received at least one dose in the first year of life was €5,435 in the birth cohorts 2015-2019. Despite the substantial risk of severe RSV infection, we found low rates of palivizumab utilization. Novel preventive interventions, featuring broader indications and single-dose administration per season, contribute to mitigating the burden of RSV disease across a more extensive infant population.


Subject(s)
Antiviral Agents , Palivizumab , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/economics , Germany/epidemiology , Palivizumab/administration & dosage , Palivizumab/therapeutic use , Infant , Female , Male , Respiratory Syncytial Virus, Human/immunology , Antiviral Agents/economics , Antiviral Agents/administration & dosage , Antiviral Agents/therapeutic use , Health Care Costs/statistics & numerical data , Infant, Newborn , Vaccination/statistics & numerical data , Vaccination/economics , Immunization/statistics & numerical data , Birth Cohort , Child, Preschool
8.
J Korean Med Sci ; 39(28): e206, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39048301

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV), a highly transmissible virus, is the leading cause of lower respiratory tract infections. We examined molecular changes in the RSV genome before and after the coronavirus disease 2019 (COVID-19) pandemic in Korea, and investigated whether drug-resistant mutations were present. METHODS: In this prospective, single-center study, RSV-positive respiratory samples were collected between September 2019 and December 2022. Long-read whole-genome sequencing (WGS) was performed, and the presence of known drug-resistant substitutions for palivizumab, nirsevimab, and suptavumab was investigated. RESULTS: Overall, 288 respiratory samples were collected from 276 children. WGS data were available for 133 samples (71 and 62 samples from the pre- and post-pandemic periods, respectively). All RSV-A strains (n = 56) belonged to the GA2.3.5 (ON1) genotype, whereas all RSV-B strains (n = 77) belonged to the GB5.0.5a (BA) genotype. No significant differences in genotypes were observed between the pre- and post-pandemic periods. In addition, no notable mutations related to nirsevimab or palivizumab resistance were detected in the F gene. However, the L172Q and S173L substitutions, which are known to confer resistance to suptavumab, were present in all RSV-B samples. CONCLUSION: Despite the unprecedented interruption of RSV seasonality, there were no significant molecular changes in circulating RSV strains in Korea related to nirsevimab or palivizumab resistance before and after the COVID-19 pandemic. However, RSV-specific drug-resistance substitutions for suptavumab were identified.


Subject(s)
COVID-19 , Genotype , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , SARS-CoV-2 , Whole Genome Sequencing , Humans , Prospective Studies , Republic of Korea/epidemiology , COVID-19/virology , COVID-19/epidemiology , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/virology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/isolation & purification , Drug Resistance, Viral/genetics , Antiviral Agents/therapeutic use , Genome, Viral , Palivizumab/therapeutic use , Female , Mutation , Male , Child , Child, Preschool , Infant , Antibodies, Monoclonal, Humanized/therapeutic use
11.
Vaccine ; 42(21): 126164, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39079810

ABSTRACT

BACKGROUND: Health Canada recently authorized the RSVpreF pregnancy vaccine and nirsevimab to protect infants against respiratory syncytial virus (RSV) disease. OBJECTIVE: Assess the cost-effectiveness of RSVpreF and nirsevimab programs in preventing RSV disease in infants, compared to a palivizumab program. METHODS: We used a static cohort model of a Canadian birth cohort during their first RSV season to estimate sequential incremental cost-effectiveness ratios (ICERs) in 2023 Canadian dollars per quality-adjusted life year (QALY) for nine strategies implemented over a one-year time period, from the health system and societal perspectives. Sensitivity and scenario analyses were conducted to explore the impact of uncertainties on the results. RESULTS: All-infants nirsevimab programs averted more RSV-related outcomes than year-round RSVpreF programs, with the most RSV cases averted in a seasonal nirsevimab program with catch-up. Assuming list prices for these immunizing agents, all-infants nirsevimab and year-round RSVpreF programs were never cost-effective, with ICERs far exceeding commonly used cost-effectiveness thresholds. Seasonal nirsevimab with catch-up for infants born outside the RSV season was a cost-effective program if prioritized for infants at moderate/high-risk (ICER <$28,000 per QALY) or those living in settings with higher RSV burden and healthcare costs, such as remote communities where transport would be complex (ICER of $5700 per QALY). Using a $50,000 per QALY threshold, an all-infants nirsevimab program could be optimal if nirsevimab is priced at <$110-190 per dose. A year-round RSVpreF for all pregnant women and pregnant people plus nirsevimab for infants at high-risk was optimal if nirsevimab is priced at >$110-190 per dose and RSVpreF priced at <$60-125 per dose. INTERPRETATION: Prophylactic interventions can substantially reduce RSV disease in infants, and more focused nirsevimab programs are the most cost-effective option at current product prices.


Subject(s)
Antibodies, Monoclonal, Humanized , Cost-Benefit Analysis , Palivizumab , Quality-Adjusted Life Years , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Humans , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/economics , Infant , Canada , Respiratory Syncytial Virus Vaccines/economics , Respiratory Syncytial Virus Vaccines/therapeutic use , Respiratory Syncytial Virus Vaccines/administration & dosage , Antibodies, Monoclonal, Humanized/economics , Antibodies, Monoclonal, Humanized/therapeutic use , Palivizumab/therapeutic use , Palivizumab/economics , Female , Antiviral Agents/economics , Antiviral Agents/therapeutic use , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/drug effects , Male , Infant, Newborn
13.
JAMA Netw Open ; 7(6): e2416077, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38861259

ABSTRACT

Importance: Respiratory syncytial virus (RSV) transmission was disrupted worldwide following the COVID-19 pandemic, and further study is required to better understand these changes. Objective: To compare observed and expected RSV hospital and intensive care unit (ICU) admission rates and characteristics of admitted children during the 2021-2022 and 2022-2023 seasons. Design, Setting, and Participants: A population-based cohort study of all children aged younger than 5 years in Ontario, Canada, July 1, 2017, through March 31, 2023, was conducted. Exposures: Individual and neighborhood-level sociodemographic and clinical characteristics were identified from administrative data, including age, palivizumab eligibility, complex medical conditions, rurality, and living in a marginalized neighborhood. Main Outcomes and Measures: The main outcome was RSV-associated hospitalization. Secondary outcomes included ICU admissions, mechanical ventilation, extracorporeal membrane oxygenation, and in-hospital death. Poisson generalized estimating equations were used to model weekly age- and sex-specific hospitalization rates and estimate expected rates in the postpandemic era; adjusted rate ratios (RRs) and 95% CIs are reported. Results: This cohort study included approximately 700 000 children per study year. Compared with prepandemic years (2017-2018, 2018-2019, and 2019-2020), the 2021-2022 RSV season peaked slightly earlier, but overall admission rates were comparable (289.1 vs 281.4-334.6 per 100 000, or approximately 2000 admissions). The 2022-2023 season peaked a month earlier and resulted in more than twice as many hospitalizations (770.0 per 100 000; n = 4977 admissions). The proportion of children admitted to an ICU in 2022-2023 (13.9%) was slightly higher than prepandemic (9.6%-11.4%); however, the population-based rate was triple the prepandemic levels (106.9 vs 27.6-36.6 per 100 000 children in Ontario). With the exception of palivizumab-eligible children, all sociodemographic and health status characteristics were associated with lower-than-expected RSV hospitalization rates in 2021-2022. In contrast, older age of patients was associated with higher-than-expected rates in 2022-2023 (ie, 24-59 months: RR, 1.90; 95% CI, 1.35-2.66). Conclusions and Relevance: There were notable differences in RSV epidemiologic characteristics in Ontario following the COVID-19 pandemic. It is not yet clear whether and how long atypical RSV epidemics may persist. Clinicians and program planners should consider the potential for ongoing impacts to health care capacity and RSV immunization programs.


Subject(s)
COVID-19 , Hospitalization , Respiratory Syncytial Virus Infections , Humans , Respiratory Syncytial Virus Infections/epidemiology , Hospitalization/statistics & numerical data , Infant , Male , Female , Child, Preschool , Ontario/epidemiology , COVID-19/epidemiology , SARS-CoV-2 , Intensive Care Units/statistics & numerical data , Cohort Studies , Infant, Newborn , Respiration, Artificial/statistics & numerical data , Pandemics , Palivizumab/therapeutic use
15.
Turk J Pediatr ; 66(2): 161-170, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38814297

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) is one of the most common pathogens causing severe lower respiratory tract disease in infancy and childhood. In newborns, young infants, and in infants with co-morbidities, the risk of severe infection is increased. Current protection against severe RSV infection is immunoprophylaxis with the monoclonal antibody palivizumab. The study aimed to assess the effects of palivizumab prophylaxis in the Republic of Montenegro in comparison to the pre-prophylaxis period. METHODS: The study was conducted in prospective/retrospective single center format in Montenegro in the Clinical Center of Podgorica, for the period 2009-2019. RESULTS: A total of 104 high-risk infants in the palivizumab prophylaxis program (2014-2019 RSV seasons) and 168 high-risk children without palivizumab prophylaxis (2009-2013 RSV seasons) were enrolled. A total of 51 children (49.0%) received prophylaxis for prematurity, 33 (31.7%) for bronchopulmonary dysplasia (BPD), 13 (12.5%) for hemodynamically significant heart disease/defect (HSCHD), and 7 (6.8%) for "miscellaneous" indications. In the control group most children had prematurity (101, 60.1%), followed by BPD (59, 35.1%), HSCHD (3, 1.8%), and "miscellaneous" (5, 3.4%) conditions. Readmission to the pediatric intensive care units (PICU) due to RSV infection was significantly lower in prophylaxis group (0.0 vs 16.1%, p<0.001). No lethal outcomes were observed in high-risk children with palivizumab prophylaxis compared to 2.4% in the control group. CONCLUSIONS: The introduction of RSV immunoprophylaxis as well as other new protective treatment strategies for high-risk newborns led to significant improvements in infant and childcare in Montenegro. This is the first report on palivizumab prophylaxis in Montenegro, demonstrating the effectiveness and safety of palivizumab use in clinical settings.


Subject(s)
Antiviral Agents , Palivizumab , Respiratory Syncytial Virus Infections , Palivizumab/therapeutic use , Humans , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/epidemiology , Antiviral Agents/therapeutic use , Male , Infant , Female , Infant, Newborn , Prospective Studies , Retrospective Studies , Montenegro/epidemiology , Infant, Premature
16.
Hum Vaccin Immunother ; 20(1): 2348135, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38738683

ABSTRACT

Nirsevimab has been recently licensed for universal RSV prophylaxis in infants. NIRSE-GAL is a three-year population-based study initiated in Galicia in September 2023. It aims to evaluate nirsevimab effectiveness against RSV-related hospitalizations lower respiratory tract infections (LRTI), severe RSV, all-cause LRTI, and all-cause hospitalization. NIRSE-GAL also aims to estimate nirsevimab impact on primary healthcare use in the short and mid-term, children's wheezing and asthma, and medical prescriptions for RSV. The immunization campaigns will be scheduled based on the expected start week for the RSV season and will last the whole season. Immunization will be offered to: i) infants born during the campaign (seasonal), ii) infants < 6 months at the start of the campaign (catch-up), and iii) infants with high-risk factors, aged 6-24 months at the start of the campaign (high-risk). The follow-up period will start: i) the immunization date for all immunized infants, ii) the start of the campaign, for the non-immunized catch-up or high-risk groups, or iii) the birthdate for the non-immunized seasonal group. Infants will be followed up until outcome occurrence, death, or end of study. Nirsevimab effectiveness will be estimated using Poisson and Cox regression models. Sensitivity and stratified analyses will be undertaken. The number of averted cases and the number needed to immunize will be estimated. Immunization failure and nirsevimab safety will be monitored. NIRSE-GAL was approved by the ethics committee of Galicia (CEIC 2023-377) and registered in ClinicalTrials.gov (ID: NCT06180993). Findings will be mainly shared via peer-reviewed publications and scientific conferences.


Subject(s)
Antiviral Agents , Hospitalization , Respiratory Syncytial Virus Infections , Humans , Respiratory Syncytial Virus Infections/prevention & control , Infant , Hospitalization/statistics & numerical data , Antiviral Agents/therapeutic use , Antiviral Agents/administration & dosage , Respiratory Syncytial Virus, Human/immunology , Female , Male , Respiratory Tract Infections/prevention & control , Immunization Programs , Infant, Newborn , Child, Preschool , Palivizumab/therapeutic use , Palivizumab/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage
18.
Neonatal Netw ; 43(2): 105-115, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38599778

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of hospitalization in infancy in the United States. Nearly all infants are infected by 2 years of age, with bronchiolitis requiring hospitalization often occurring in previously healthy children and long-term consequences of severe disease including delayed speech development and asthma. Incomplete passage of maternal immunity and a high degree of genetic variability within the virus contribute to morbidity and have also prevented successful neonatal vaccine development. Monoclonal antibodies reduce the risk of hospitalization from severe RSV disease, with palivizumab protecting high-risk newborns with comorbidities including chronic lung disease and congenital heart disease. Unfortunately, palivizumab is costly and requires monthly administration of up to five doses during the RSV season for optimal protection.Rapid advances in the past two decades have facilitated the identification of antibodies with broad neutralizing activity and allowed manipulation of their genetic code to extend half-life. These advances have culminated with nirsevimab, a monoclonal antibody targeting the Ø antigenic site on the RSV prefusion protein and protecting infants from severe disease for an entire 5-month season with a single dose. Four landmark randomized controlled trials, the first published in July 2020, have documented the efficacy and safety of nirsevimab in healthy late-preterm and term infants, healthy preterm infants, and high-risk preterm infants and those with congenital heart disease. Nirsevimab reduces the risk of RSV disease requiring medical attention (number needed to treat [NNT] 14-24) and hospitalization (NNT 33-63) with rare mild rash and injection site reactions. Consequently, the Centers for Disease Control and Prevention has recently recommended nirsevimab for all infants younger than 8 months of age entering or born during the RSV season and high-risk infants 8-19 months of age entering their second season. Implementing this novel therapy in this large population will require close multidisciplinary collaboration. Equitable distribution through minimizing barriers and maximizing uptake must be prioritized.


Subject(s)
Antibodies, Monoclonal, Humanized , Respiratory Syncytial Virus Infections , Humans , Infant, Newborn , Antiviral Agents/therapeutic use , Heart Defects, Congenital , Infant, Premature , Palivizumab/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/prevention & control , United States , Randomized Controlled Trials as Topic
19.
BMC Infect Dis ; 24(1): 418, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38641577

ABSTRACT

AIM: Palivizumab has proven effective in reducing hospitalizations, preventing severe illness, improving health outcomes, and reducing healthcare costs for infants at risk of respiratory syncytial virus (RSV) infection. We aim to assess the value of palivizumab in preventing RSV infection in high-risk infants in Colombia, where RSV poses a significant threat, causing severe respiratory illness and hospitalizations. METHODS: We conducted a decision tree analysis to compare five doses of palivizumab with no palivizumab. The study considered three population groups: preterm neonates (≤ 35 weeks gestational age), infants with bronchopulmonary dysplasia (BPD), and infants with hemodynamically significant congenital heart disease (CHD). We obtained clinical efficacy data from IMpact-RSV and Cardiac Synagis trials, while we derived neonatal hospitalization risks from the SENTINEL-1 study. We based hospitalization and recurrent wheezing management costs on Colombian analyses and validated them by experts. We estimated incremental cost-effectiveness ratios and performed 1,000 Monte Carlo simulations for probabilistic sensitivity analyses. RESULTS: Palivizumab is a dominant strategy for preventing RSV infection in preterm neonates and infants with BPD and CHD. Its high efficacy (78% in preventing RSV in preterm infants), the substantial risk of illness and hospitalization, and the high costs associated with hospitalization, particularly in neonatal intensive care settings, support this finding. The scatter plots and willingness-to-pay curves align with these results. CONCLUSION: Palivizumab is a cost-saving strategy in Colombia, effectively preventing RSV infection in preterm neonates and infants with BPD and CHD by reducing hospitalizations and lowering healthcare costs.


Subject(s)
Heart Defects, Congenital , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Infant , Infant, Newborn , Humans , Palivizumab/therapeutic use , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/prevention & control , Cost-Benefit Analysis , Colombia/epidemiology , Antiviral Agents/therapeutic use , Infant, Premature , Antibodies, Monoclonal, Humanized/therapeutic use , Hospitalization
20.
J Pediatric Infect Dis Soc ; 13(Supplement_2): S103-S109, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38577737

ABSTRACT

The efforts to prevent respiratory syncytial virus (RSV) infection in infants span over half a century. RSV vaccine development began in the 1960s, and it confronted a significant disappointment after testing a formalin-inactivated RSV (FI RSV) vaccine candidate. This inactivated RSV vaccine was not protective. A large number of the vaccinated RSV-naive children, when subsequently exposed to natural RSV infection from wild-type virus in the community, developed severe lung inflammation termed enhanced respiratory disease. This resulted in a halt in RSV vaccine development. In the 1990s, attention turned to the potential for passive protection against severe RSV disease with immunoglobulin administration. This led to studies on using standard intravenous immunoglobulins in high-risk infants, followed by high-titer RSV immunoglobulin preparation and, subsequently, the development of RSV monoclonal antibodies. Over the past 25 years, palivizumab has been recognized as a safe and effective monoclonal antibody as a prevention strategy for RSV in high-risk children. Its high cost and need for monthly administration, however, has hindered its use to ~2% of the birth cohort, neglecting the vast majority of newborns, including healthy full-term infants who comprise the largest portion of RSV hospitalizations and the greatest part of the burden of RSV disease. Still these efforts, helped pave the way for the present advances in RSV prevention that hold promise for mitigating severe RSV disease for all infants.


Subject(s)
Immunization, Passive , Palivizumab , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/history , Humans , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus Vaccines/therapeutic use , Respiratory Syncytial Virus Vaccines/administration & dosage , History, 20th Century , Immunization, Passive/methods , Palivizumab/therapeutic use , History, 21st Century , Infant , Respiratory Syncytial Virus, Human/immunology , Vaccines, Inactivated/immunology , Vaccine Development , Antiviral Agents/therapeutic use , Antiviral Agents/administration & dosage , Immunoglobulins, Intravenous/therapeutic use , Antibodies, Monoclonal/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL