Key information

Mode of transmission

Transmitted from respiratory secretions to nasopharyngeal or conjunctival mucosa, through close or direct contact with large droplets or fomites.

Incubation period

Ranges from two to eight days

Period of communicability

Average eight days, up to three weeks

Available vaccines

Unfunded – RSVpreF vaccine (Arexvy) – for active immunisation

Unfunded – palivizumab – for passive immunoprophylaxis

Dose, presentation, route

RSV vaccine (Arexvy)

-       0.5 mL per dose

-       Two glass vials – antigen powder must be reconstituted with adjuvant suspension.

-       Intramuscular injection

 

Palivizumab

-       Single-use vials containing 50 mg / 0.5 mL or 100 mg / 1.0 mL palivizumab

-       Recommended dose 15 mg/Kg, monthly during RSV season

-       Intramuscular injection

Vaccine indications and schedule

RSV vaccine (Arexvy)

Unfunded

One dose, for use in adults aged 60 years and over

For use prior to or during RSV season (autumn/winter months)

 

Palivizumab

Unfunded

For use in infants aged under 24 months at high risk of severe RSV-LRTI, particularly recommended for:

-       born preterm ≤28 weeks gestation

-       born preterm ≤32 weeks gestation with chronic lung disease of prematurity

-       with haemodynamically significant congenital heart disease

-       severely immunocompromised infants.

Vaccine effectiveness

RSV vaccine (Arexvy):

Clinical trial data: efficacy of 83 percent against RSV-LRTI and 94 percent against severe RSV disease in adults aged over 60 and over 70 years.

Protection is maintained for at least two RSV seasons.

18.1. Virology

Respiratory syncytial virus (RSV) is an Orthopneumovirus of the Pneumoviridae family. There are two main antigenic subtypes in humans, subtype A and B. Virus-induced fusion of infected cells forms multinucleated cells known as syncytia.

18.2. Clinical features

Respiratory syncytial virus is a highly contagious and common virus that can cause lower respiratory tract infections (LRTIs), particularly in the late autumn and winter months. Most children will have been exposed to RSV by the age of 2 years but reinfection is common.[1]


It is transmitted in large droplets or on fomites from respiratory secretions, infecting the nasopharyngeal or conjunctiva mucosa.[2] Incubation period ranges from two to eight days and viral shedding can last for an average of eight days to up to three weeks from immunocompetent individuals.[2]


Infection starts in the upper respiratory tract and then spreads rapidly by intercellular transmission to the lower airways to the terminal bronchioles.[2] RSV infection disrupts ciliated epithelial cells in the airways, stimulating mononuclear cell infiltration, mucosal oedema and syncytia formation. It disrupts the cell-cycle regulatory proteins to boost viral replication. Severity and infectivity is directly related to viral load.[3]


With subsequent infections, acquired humoral and cell-mediated immunity attenuate the severity of each infection assisted by increasing age, size and maturity of the airways during childhood. Conversely, in older adults, declines in cell-mediated immunity and lung function increase the severity of RSV infection.

18.2.1. Adults

18.2.1. Adults

RSV is a major cause of upper respiratory tract infection (URTI) in adults. By adulthood, individuals are unlikely to be immunologically naïve to RSV, with repeat exposures occurring throughout our lifetimes.[3] Most infected adults experience mild to moderate clinical disease. RSV-associated hospitalisation rates increase exponentially from the age of 60 years. For those with immunocompromise and older age, RSV infection can result in hospitalisation for severe lower respiratory tract disease (LRTD), viral pneumonia and exacerbation of underlying comorbidities.[4]

 

RSV is a highly prevalent disease and the virus has been identified as the causative agent in up to 12 percent of medically-attended acute respiratory illnesses.[4] It is associated with exacerbation of pre-existing illness in adults and with excess deaths predominantly in elderly adults. Hospital admission is on average three to six days and overall mortality is 6–8 percent in adults. Among hospitalised adults with a positive-RSV test, 10–31 percent require intensive care admission and 3–17 percent need mechanical ventilation.[4]

 

Risk factors for progression of RSV infection to viral pneumonia include Down syndrome, immunocompromise, underlying lung or heart disease, increasing age from 65 years, frailty, living in long-term care facilities and living at high altitudes.[4] Further factors associated with more severe RSV-LRTD include vitamin D deficiency, colder weather, air pollution and exposure to tobacco smoke.[4] The burden of RSV in adults is likely to be higher when consideration is given to the exacerbation of underlying disease, such as heart and lung disease, beyond those recorded as having RSV-LRTI.[5] In New Zealand, being of Māori or Pacific ethnicity, and/or living in areas of high deprivation independently increases the risk of RSV-associated hospitalisation.[6]

18.2.2. Infants

18.2.2. Infants

Compared with less than one percent of all adults, a higher proportion (around 12 percent) of young children with RSV require hospital care. Risk factors for poor outcomes in infants are: age under 3 months and gradually decreasing with increasing age, congenital heart disease, chronic conditions diagnosed before age 6 months, including congenital heart, congenital lung and chronic lung disease or neurological disease; prematurity (born before 37 weeks gestation) and extreme prematurity (born before 32 weeks gestation).[7,8,9] Another risk factor for hospitalisation with RSV-LRTI in infants are young maternal age (under 20 years). Preschool and primary school children in the household are significant sources of infection to infants.[10] In New Zealand, being of Māori or Pacific ethnicity, and/or living in areas of high deprivation is also independently associated with an increased risk for RSV-associated hospitalisation of infants.[11]

18.3. Epidemiology

18.3.1. Global burden of disease

18.3.1. Global burden of disease

More than 80 percent of children are infected with RSV at least once by the age of 2 years, and half of these have been infected twice.[12] Globally, RSV is associated with a substantial burden in young infants and is the most common cause of hospitalisation of infants under 2 years for acute LRTI. The virus is also linked to an increased risk of childhood asthma.[9,13]

In adults, the available epidemiology of RSV is less robust than for children. The frequency of clinical testing is lower, and in ambulatory settings, adults often present later to clinical care. Despite this, RSV is a highly prevalent disease associated with excess deaths predominantly in the older adults. The incidence of medically-attended RSV infection in adults generally increases with age, and RSV-associated pneumonia in adults age 65 years and over has the highest annual mortality rate (7.2 per 100,000 persons per year).[4] The odds of an adult with comorbidity being hospitalised with RSV acute respiratory illness is approximately four times that of individuals without comorbidity.[5] The burden is likely to be underestimated, and would be greater if exacerbation of underlying diseases, such as heart and lung disease, due to RSV infection was considered.

18.3.2. New Zealand epidemiology

18.3.2. New Zealand epidemiology

RSV is not a notifiable disease in Aotearoa New Zealand, but the Institute of Environmental Surveillance and Research (ESR) maintains several surveillance systems to monitor respiratory virus activity and severity for the Public Health Agency. Virus detection forms part of the surveillance for community respiratory infections and severe acute respiratory infection (SARI) hospitalisation, including RSV testing. In 2023, like other seasonal respiratory viruses, RSV was predominantly detected in the community during the winter months but RSV-SARI occurred throughout the year.[14]

One study examining the RSV-associated hospitalisation rates in adults in Auckland during 2012–2015 found that approximately 8 percent of SARI hospitalisations were associated with RSV infection and 90 percent of these cases occurred during winter.[6] Adults aged over 80 years had the highest incidence of RSV-SARI, at 190.8 (95% CI 137.6-244.0) per 100,000 compared with 23.6 (21.0-26.1) per 100,000 for all adults. The incidence rate ratio was over 30 times higher for oldest adults than those aged under 50 years.[6]

18.4. Vaccines

An early respiratory syncytial virus vaccine candidate in the 1960s, containing formalin-inactivated whole virus, was discontinued due to the occurrence of vaccine-associated enhanced disease. During a clinical trial some participants experienced more severe RSV after exposure to RSV infection following vaccination. Advances in virology have since identified two forms of the F protein, the protein used by the virus to attach to epithelial cells of the airways: namely, a prefusion form and a post fusion form. DNA technology to produce recombinant proteins has enabled the production of the F protein in its prefusion conformation only. The risk of vaccine-associated enhanced disease, which is driven by T cell and antibody responses against the post-fusion F protein, has been overcome in the current vaccines by targeting specific antigenic sites expressed on the prefusion F protein only.

18.4.1. Available vaccines

18.4.1. Available vaccines

New Zealand approved vaccine (unfunded)

Adjuvanted RSVpreF vaccine (Arexvy, GSK)

Each 0.5 ml dose of RSVpreF contains:

  • 120 µg RSV glycoprotein F stabilized in the prefusion conformation (RSVPreF3) produced by recombinant DNA technology in Chinese hamster ovary (CHO) cells
  • AS01E adjuvant, containing 25 µg Quillaja saponaria Molin fraction 21 (QS-21) and 25 µg 3-O-desacyl-4’-monophophoryl lipid A (MPL) from Salmonella minnesota)
  • it also contains trehalose dihydrate, polysorbate 80, monobasic potassium phosphate, dibasic potassium phosphate, dioleoyl phosphatidylcholine, cholesterol, sodium chloride and water.

 

Palivizumab (Synagis, AstraZeneca)

Palivizumab is a humanised IgG1κ monoclonal antibody targeting antigen site A (II) of the RSV fusion protein F to provide monthly passive immunity (immunoprophylaxis) against RSV in infants and young children at very high risk of RSV disease.

Each 0.5 mL dose contains 50mg or 1 mL dose contains 100 mg of palivizumab.

  • Palivizumab is produced by DNA technology in recombinant mouse myeloma cells. It also contains histidine, glycine and water.

Other RSV vaccines, for use in older adults or during pregnancy to provide passive protection for infants, and immunoprophylactic candidates for young children, are approved for use or under regulatory review elsewhere, including in Australia, Europe and North America.[15] Some of which are also undergoing regulatory review in New Zealand.

18.4.2. Efficacy and effectiveness

18.4.2. Efficacy and effectiveness

Immunogenicity of adjuvanted RSVpreF vaccine, Arexvy

One dose of adjuvanted RSVpreF vaccine (Arexvy) induces robust RSV-A and RSV-B neutralising antibodies that are likely to be seroprotective for at least one year after vaccination in older adults (aged ≥60 years).[16] The T cell profile is strongly CD4+ Th1-driven, and there is no evidence of induction of CD8+ T cells associated with risk for vaccine-associated enhanced respiratory disease.[17] An anamnestic response is seen after a booster dose given 20 months after the first dose.[18] Although clinical trials evaluated a two-dose primary course, it appears that one dose induces an adequate immune response to provide protection for longer than one year.[16,17]

RSVpreF vaccine has similar immunogenicity in adults aged 18-50 years to that seen in older adults. Similar efficacy is also seen in those aged 50-59 years with comorbidities that increase the risk of severe RSV (study to date included chronic heart failure, disease, chronic respiratory disease, advanced renal or liver disease, diabetes mellitus type 1 or 2, but exclude those with immunocompromise).[19,20] Further data is anticipated over more than two RSV seasons, in special high-risk groups and immunocompromised groups, and concomitant administration with other vaccines given to older adults.

 

Efficacy of adjuvanted RSVpreF vaccine, Arexvy

Vaccine efficacy of adjuvanted RSVpreF was consistently high among adults aged from 60 years against RSV-LRTI, severe RSV-LRTD and RSV-ARI during a first RSV season in a phase 3 RCT (NCT0488659).[21] Over one RSV season, vaccine efficacy was around 83 percent (96.95% CI: 58 – 94 percent) against RSV-LRTI; 94 percent (97.5% CI: 62 – 99.9 percent) against severe RSV-LRTD; and 72 percent (56 – 82 percent) against RSV-ARI in adults aged from 60 years,[21] and was similar in adults over 70 years. Similarly, efficacy of 95 percent (66–99.9 percent) was also shown against RSV-LRTD in those with at least one comorbidity that increases the risk of severe RSV.[19] A second dose, given a year after the first, provided little additional benefit than that provided by one dose over two seasons.[22] The efficacy of one dose over two seasons (adjusted for seasonality) was maintained around 80 percent (53–92 percent) against severe RSV-LRTD and 67 percent (48–80) against all RSV-LRTI.[22] Further data is required to assess the optimal timing of revaccination.

 

Efficacy of palivizumab

Licensure was based on the IMpact-RSV randomised control trial, conducted during the 1996-1997 RSV season, which included 1502 infants and young children born at or before 35 weeks gestation (some with chronic lung disease (CLD) associated with prematurity).[23] Compared with the placebo cohort (RSV hospitalisation rate of 10.6 percent), an absolute reduction of 5.8% (p < 0.001) was seen in hospitalisation of those who received palivizumab (4.8 percent); equivalent to a relative risk reduction of 54.7 percent.[23] A second clinical trial in 1287 children with haemodynamically significant congenital heart disease reported an absolute reduction in hospitalisation of 4.4 percent (p < 0.003; 9.7 percent in placebo group and 5.3 percent in palivizumab group); with relative risk reduction of 45.3 percent.[23]

There is limited and conflicting data on effectiveness of palivizumab. Whilst retrospective studies have not shown consistent increases nor decrease in RSV-related hospitalisation for those born 29–35 weeks receiving palivizumab, many did not examine cohorts separately based on comorbidities, such as chronic lung disease of prematurity, and acknowledge that adherence with injections is challenging.[23,24,25] A systematic review reported that palivizumab reduces hospitalisation due to RSV infection by 56 percent (relative risk 0.44; 95% CI 0.30-0.64), but evidence is uncertain around mortality (relative risk 0.69; 0.42-1.15).[26]

18.4.3. Transport, storage and handling

18.4.3. Transport, storage and handling

Transport according to the National Standards for Vaccine Storage and Transportation for Immunisation Providers 2017 (2nd edition).

Store at +2°C to +8°C. Do not freeze. Protect from light.

18.4.4. Dosage and administration

18.4.4. Dosage and administration

A single 0.5 mL dose of Arexvy is administered by intramuscular injection (see section 2.2.3).

This vaccine requires reconstitution prior to administration. It consists of two components, a vial containing powder of recombinant prefusion F protein and a vial containing a suspension of adjuvant, AS01E. Draw up the adjuvant suspension into a syringe and add the entire contents into the power. Gently swirl until the powder is dissolved. Use within 4 hours of reconstitution.

Both vials have a non-latex, butyl rubber stopper.

 

Palivizumab

Palivizumab is administered by intramuscular injection, monthly according to infant weight at 15 mg/Kg. Injection volumes of over 1mL should be given as a divided dose. Single-use vials contain 50 mg / 0.5 mL or 100 mg / 1.0 mL of palivizumab.

 

Co-administration with other vaccines

RSVpreF vaccine can be given concomitantly with standard seasonal quadrivalent influenza vaccine.

There is currently no data for concomitant administration with other vaccines, but as a non-live vaccine there are unlikely to be any concerns. As this vaccine contains AS01E adjuvant, when given with other adjuvanted vaccines such as ZV (Shingrix), rCV (Nuvaxovid) or aQIV (FluAd Quad), there could be an increase in the frequency of mild to moderate reactions. It is preferable to give these vaccines is different limbs, where possible.

Palivizumab can be administered at any time before or after any routine childhood immunisations. Since the monoclonal antibody in palivizumab is specific for RSV, it is not expected to interfere with the immune response to vaccines, including live viral vaccines.

18.5.1. Recommended groups (unfunded) for RSVpreF vaccine

18.5.1. Recommended groups (unfunded) for RSVpreF vaccine

RSVpreF (Arexvy) is indicated for active immunisation for the prevention of lower respiratory tract disease (LRTD) caused by respiratory syncytial virus RSV-A and RSV-B subtypes in adults 60 years of age and older.

It is currently unfunded. It can be considered for individuals who are aged over 60, especially the very elderly and those with multiple comorbidities that increase the risk of severe RSV-ARI.  Offered as one dose of RSVpreF vaccine given prior to the start of the RSV season in late autumn or as soon as possible during the RSV season in the winter.

18.5.2. Recommended groups (unfunded) for palivizumab

18.5.2. Recommended groups (unfunded) for palivizumab

For use in infants at high risk of severe RSV-LRTI, particularly recommended for those aged under 1 year prior to or during the RSV season:

  • born preterm up to 28 weeks gestation
  • born preterm up to 32 weeks gestation with chronic lung disease of prematurity
  • with haemodynamically significant congenital heart disease
  • severely immunocompromised infants.

18.5.3. Pregnancy and breastfeeding

18.5.3. Pregnancy and breastfeeding

RSVpreF (Arexvy) is indicated from the age of 60 years and is not recommended for use in pregnancy or while lactating/breast-feeding.

Palivizumab is not indicated for adolescent or adult use.

18.5.4. Immunocompromised individuals

18.5.4. Immunocompromised individuals

Although safety and immunogenicity data are not yet available in immunocompromised adults, as a non-live vaccine, there are no safety concerns around giving RSVpreF to individuals who are immunocompromised and who are at increased risk of being hospitalised with severe RSV infection. Individuals receiving immunosuppressive therapy or who have immunodeficiency could have a reduced immune response to RSV vaccine.

Palivizumab is available for use in infants with immunocompromise, particularly those with severe immunodeficiencies, that increases their risk of severe RSV-LRTD up to age 24 months.

18.6. Contraindications and precautions

See also section 2.1.3 for pre-vaccination screening guidelines and section 2.1.4 for general contraindications for all vaccines.

18.6.1. Contraindications

18.6.1. Contraindications

There are no specific contraindications to RSVpreF (Arexvy) or palivizumab, except for anaphylaxis to a previous dose or any component of the vaccine.

18.6.2. Precautions

18.6.2. Precautions

The RSVpreF, Arexvy, is not recommended for use during pregnancy. During a single clinical trial, a 1.5-times increase in preterm births was observed in the vaccinated cohort compared to placebo following administration of an investigational unadjuvanted formulation of RSVpreF3 to 3,557 pregnant women; the clinical trial was discontinued. Stratification by economic region showed that the increase in preterm births was seen in low-income and not in high-income countries.[27] The mechanism for this is unknown.

18.7. Potential responses and AEFIs

18.7.1. Potential responses

18.7.1. Potential responses

During clinical trials, RSVpreF was well-tolerated in adults aged from 18 years and aged from 50 years. The most frequently reported responses were injection-site pain, headache, fatigue, myalgia and arthralgia.[21] Co-administration with seasonal influenza vaccine increased the incidence of pain at the RSV vaccine injection site but there are no concerns about safety.[28]

The reported responses to palivizumab were not significantly different from placebo and the adverse event profiles were similar during clinical studies.

18.7.2. AEFI

18.7.2. AEFI

No serious adverse events or potential immune-mediated disease were attributed to RSVPreF3 OA in diverse older populations (aged over 60 years)[21] or in adults aged 50­-59 years with comorbidities that increase the risk for severe RSV.[29] Clinical trial safety follow-up has been reported for at least 18 months, to date.[22] One case of Guillain-Barré syndrome was temporally associated with vaccination but diagnosis of this was inconclusive as it did not meet the Brighton Collaboration Working Group case definition.[16]

18.8. Public health measures

Respiratory syncytial virus is not a notifiable infection in Aotearoa New Zealand. The methods of preventing and controlling RSV are the same as those for other seasonal respiratory viruses.

  • breastfeeding
  • hand hygiene (ie, regularly washing hands for at least 20 seconds and drying them for 20 seconds, or regularly using an alcohol-based hand rub)
  • respiratory hygiene (ie, cough and sneeze etiquette, and wearing of face masks in some settings for example poorly ventilated or overcrowded)
  • social distancing (ie, persuading those with symptoms to avoid others in the community by staying away from school and work when sick; in particular, infected individuals should avoid contact with the elderly, the chronically ill, and infants and babies)
  • regularly cleaning flat surfaces such as bathroom sinks, bedside cabinets, desks and tabletops
  • immunisation (as available)
  • consider healthy homes initiatives.

18.9. Variations from the vaccine data sheet

None.

References

References

References
  1. Andeweg SP, Schepp RM, van de Kassteele J, et al. Population-based serology reveals risk factors for RSV infection in children younger than 5 years. Scientific Reports, 2021. 11(1): p. 8953.
  2. Piedimonte G ,Perez MK. Respiratory syncytial virus infection and bronchiolitis. Pediatrics in Review, 2014. 35(12): p. 519-30.
  3. Taleb SA, Al Thani AA, Al Ansari K,Yassine HM. Human respiratory syncytial virus: pathogenesis, immune responses, and current vaccine approaches. European Journal of Clinical Microbiology and Infectious Diseases, 2018. 37(10): p. 1817-1827.
  4. Nam HH ,Ison MG. Respiratory syncytial virus infection in adults. BMJ, 2019. 366: p. l5021.
  5. Shi T, Vennard S, Jasiewicz F, et al. Disease burden estimates of respiratory syncytial virus related acute respiratory infections in adults with comorbidity: A systematic review and meta-analysis. Journal of Infectious Diseases, 2022. 226(Suppl 1): p. S17-S21.
  6. Prasad N, Newbern EC, Trenholme AA, et al. The health and economic burden of respiratory syncytial virus associated hospitalizations in adults. PloS One, 2020. 15(6): p. e0234235.
  7. Hardelid P, Verfuerden M, McMenamin J, et al. The contribution of child, family and health service factors to respiratory syncytial virus (RSV) hospital admissions in the first 3 years of life: birth cohort study in Scotland, 2009 to 2015. Euro Surveillance, 2019. 24(1).
  8. Munro APS, Martinon-Torres F, Drysdale SB,Faust SN. The disease burden of respiratory syncytial virus in infants. Current Opinion in Infectious Diseases, 2023. 36(5): p. 379-384.
  9. Shi T, Vennard S, Mahdy S,Nair H. Risk factors for poor outcome or death in young children with respiratory syncytial virus-associated acute lower respiratory tract infection: A systematic review and meta-analysis. Journal of Infectious Diseases, 2022. 226(Suppl 1): p. S10-S16.
  10. Agoti CN, Phan MVT, Munywoki PK, et al. Genomic analysis of respiratory syncytial virus infections in households and utility in inferring who infects the infant. Scientific Reports, 2019. 9(1): p. 10076.
  11. Prasad N, Newbern EC, Trenholme AA, et al. Respiratory syncytial virus hospitalisations among young children: a data linkage study. Epidemiology and Infection, 2019. 147.
  12. Eiland LS. Respiratory syncytial virus: diagnosis, treatment and prevention. J Pediatr Pharmacol Ther, 2009. 14(2): p. 75-85.
  13. Brenes-Chacon H, Garcia-Maurino C, Moore-Clingenpeel M, et al. Age-dependent interactions among clinical characteristics, viral loads and disease severity in young children with respiratory syncytial virus infection. Pediatric Infectious Disease Journal, 2021. 40(2): p. 116-122.
  14. Institute of Environmental Surveillance and Research (ESR). 2024 Influenza and other viral respiratory surveillance. ESR; 2024 [updated 26 May 2024]; URL: https://www.esr.cri.nz/our-research/nga-kete/infectious-disease-intelligence/influenza-and-respiratory-surveillance/. (accessed 31 May 2024)
  15. Ruckwardt TJ. The road to approved vaccines for respiratory syncytial virus. npj Vaccines, 2023. 8(1).
  16. Schwarz TF, Hwang SJ, Ylisastigui P, et al. Immunogenicity and safety following one dose of AS01E-adjuvanted respiratory syncytial virus prefusion F protein vaccine in older adults: a phase 3 trial. Journal of Infectious Diseases, 2023.
  17. Leroux-Roels I, Davis MG, Steenackers K, et al. Safety and immunogenicity of a respiratory syncytial virus prefusion F (RSVPreF3) candidate vaccine in older adults: Phase 1/2 randomized clinical trial. Journal of Infectious Diseases, 2023. 227(6): p. 761-772.
  18. Leroux-Roels I, Van Ranst M, Vandermeulen C, et al. Safety and immunogenicity of a revaccination with a respiratory syncytial virus prefusion F vaccine in older adults: A Phase 2b study. Journal of Infectious Diseases, 2023.
  19. Feldman RG, Antonelli-Incalzi R, Steenackers K, et al. Respiratory syncytial virus prefusion F protein vaccine Is efficacious in older adults with underlying medical conditions. Clinical Infectious Diseases, 2023.
  20. Gerber S. 2023 GSK's RSVPreF3 + ASO1E vaccine (Arexvy). ACIP meeting 25 October 2023. URL: https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2023-10-25-26/02-gerber-adult-RSV-508.pdf. (accessed 31 May 2024)
  21. Papi A, Ison MG, Langley JM, et al. Respiratory syncytial virus Prefusion Fprotein vaccine in older adults. New England Journal of Medicine, 2023. 388(7): p. 595-608.
  22. Ison MG, Papi A, Athan E, et al. Efficacy and safety of respiratory syncytial virus prefusion F protein vaccine (RSVPreF3 OA) in older adults over 2 RSV seasons. Clinical Infectious Diseases, 2024.
  23. Caserta MT, O'Leary ST, Munoz FM,Ralston SL. Palivizumab Prophylaxis in Infants and Young Children at Increased Risk of Hospitalization for Respiratory Syncytial Virus Infection. Pediatrics, 2023. 152(1).
  24. Link-Gelles R, Ciesla AA, Mak J, et al. Early estimates of updated 2023-2024 (monovalent XBB.1.5) COVID-19 vaccine effectiveness against symptomatic SARS-CoV-2 infection attributable to co-circulating omicron variants among immunocompetent adults - Increasing Community Access to Testing Program, United States, September 2023-January 2024. MMWR: Morbidity and Mortality Weekly Report, 2024. 73(4): p. 77-83.
  25. Wu P, Escobar GJ, Gebretsadik T, et al. Effectiveness of respiratory syncytial virus immunoprophylaxis in reducing bronchiolitis hospitalizations among high-risk infants. American Journal of Epidemiology, 2018. 187(7): p. 1490-1500.
  26. Garegnani L, Styrmisdóttir L, Roson Rodriguez P, et al. Palivizumab for preventing severe respiratory syncytial virus (RSV) infection in children. Cochrane Database Syst Rev, 2021. 11(11): p. CD013757.
  27. Department of Health and Social Care. 2023 Respiratory syncytial virus (RSV) immunisation programme for infants and older adults: JCVI full statement, 11 September 2023. Crown. URL: https://www.gov.uk/government/publications/rsv-immunisation-programme-jcvi-advice-7-june-2023/respiratory-syncytial-virus-rsv-immunisation-programme-for-infants-and-older-adults-jcvi-full-statement-11-september-2023. (accessed 2023 February 7)
  28. Chandler R, Montenegro N, Llorach C, et al. Immunogenicity, reactogenicity, and safety of AS01E-adjuvanted RSV prefusion F protein-based candidate vaccine (RSVPreF3 OA) when co-administered with a seasonal quadrivalent influenza vaccine in older adults: results of a phase 3, open-label, randomized controlled trial. Clinical Infectious Diseases, 2024.
  29. Ferguson M, Schwarz TF, Núnez SA, et al., Respiratory syncytial virus prefusion F protein vaccine (RSVPreF3 OA) is immunogenic and well-tolerated in adults 50–59 years, including adults at increased risk for RSV disease, in 8th ReSViNET Conference. 2024: Mumbai, India.