Next Article in Journal
Safety, Tolerability, and Immunogenicity of Booster Dose with MVC-COV1901 or MVC-COV1901-Beta SARS-CoV-2 Vaccine in Adults: A Phase I, Prospective, Randomized, Open-Labeled Study
Next Article in Special Issue
Effectiveness of Sinopharm’s BBIBP-CorV Booster Vaccination against COVID-19-Related Severe and Critical Cases and Deaths in Morocco during the Omicron Wave
Previous Article in Journal
BNT162b2 Booster Dose Elicits a Robust Antibody Response in Subjects with Abdominal Obesity and Previous SARS-CoV-2 Infection
Previous Article in Special Issue
mRNA and Adenoviral Vector Vaccine Platforms Utilized in COVID-19 Vaccines: Technologies, Ecosystem, and Future Directions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Insights on Respiratory Syncytial Virus Prevention

by
Edyta Kopera
,
Hanna Czajka
*,
Paweł Zapolnik
and
Artur Mazur
College of Medical Sciences, University of Rzeszów, 35-315 Rzeszów, Poland
*
Author to whom correspondence should be addressed.
Vaccines 2023, 11(12), 1797; https://doi.org/10.3390/vaccines11121797
Submission received: 7 November 2023 / Revised: 20 November 2023 / Accepted: 28 November 2023 / Published: 30 November 2023

Abstract

:
Respiratory syncytial virus (RSV) is a well-known infant pathogen transmitted mainly by droplets. It is a leading cause of upper respiratory tract infections in children, usually with a mild course of illness. RSV has also been a threat to older people, especially those with underlying medical conditions. For a long time, prevention was limited to passive immunoprophylaxis with palivizumab for high-risk infants. There was a strong need to find other treatment or prevention methods against RSV infections. In addition, after the coronavirus disease 2019 (COVID-19) pandemic, some significant changes in RSV epidemiology have been observed. Researchers noticed the shift in RSV seasonality and age distribution and the increased number of cases in older infants and adults. All of these made the need to find other medical options even stronger. Fortunately, two protein-based vaccines against RSV have successfully passed all phases of clinical trials and have been approved for use by adults and older people. One of them is also approved for infants from birth to 6 months of age (after maternal immunisation during pregnancy) and for pregnant women between 24 and 36 weeks of pregnancy. Also, a new passive immunisation option named nirsevimab (a highly potent monoclonal antibody with a long half-life) is now available for the paediatric group. In this review, we will discuss the previous and current RSV prevention methods in the light of structural discoveries of RSV antigens.

1. Introduction

Respiratory syncytial virus (RSV) is a common respiratory virus responsible for upper (URT) and lower respiratory tract (LRT) infections, mainly in infants and older adults. Usually, RSV infections, especially of the upper respiratory tract, are mild with symptoms similar to cold or flu. However, the virus can cause severe and acute respiratory illness. Among those particularly vulnerable to a severe course of RSV infection are premature infants and infants with medically complex conditions or weakened immune system, as well as older adults with underlying comorbidities [1,2,3]. For years, RSV treatment options were very limited, especially for the paediatric population. One of the antiviral drugs, named ribavirin (nucleoside), is licensed only for infants with bronchiolitis and is not recommended for routine RSV treatment. For preventing severe RSV infection in children, passive immunoprophylaxis with palivizumab has been available but only for high-risk infants. The coronavirus disease 2019 (COVID-19) pandemic has significantly influenced RSV infections, including the shift in age distribution, altered patterns of RSV seasonality and an increased number of cases, both in older infants and adults. All these phenomena have accelerated the registration of the first RSV vaccines for adults, infants and pregnant women and other treatment options for infants and young children. There are three main target groups for RSV prevention: paediatric, maternal and elderly. The following review will focus on the prevention of RSV infection in the post-COVID-19 era, mainly in children and older adults.

2. General Overview of RSV Structure

RSV is a negative-sense, single-stranded ribonucleic acid (RNA) virus with filamentous morphology [4]. The genetic material of the virus is surrounded by an envelope, which is a lipid bilayer derived from the host cell membrane. RSV phylogenetically belongs to the Orthopneumovirus genus of the Pneumoviridae family. The 15.2 kbp RSV genome consists of ten RNA segments, which are translated into 11 different proteins. Three of them, named the attachment (G), the fusion (F) and the small hydrophobic (SH) protein, are embedded in the lipid bilayer/membrane. Underneath this membrane, there is a layer of non-glycosylated matrix (M) protein that provides structural support [5]. M protein interacts with M2-1 protein, which creates an intermediate layer and mediates the association between ribonucleocapsid and the matrix protein.
M2-1 is crucial in viral sequential transcription as a transcription antitermination factor [6]. M2-2 protein is generated from another open reading frame (ORF) of the M2 gene and was shown to be involved in the balance between transcription and RNA replication. Interestingly, with the M2-2 knockout mutation, the virus growth is attenuated, but the expression of the gene is increased [7], which is a very attractive phenotype for vaccine development. Other viral proteins include nucleoprotein (N), large polymerase subunit (L) and phosphoprotein, and polymerase cofactor (P). N protein binds tightly to the viral RNA, encapsidating and protecting it within the ribonucleoprotein structure. L protein is involved in RNA synthesis, and P acts as a linker between N and L proteins [5,8]. There are also two non-structural proteins: NS1 and NS2. These proteins are independent pathogenic factors. The first mediates the inhibition and RNA replication, while the second is not essential for RNA replication but improves virus growth in cell culture [9]. Both proteins play a crucial role in suppressing the innate immune response and could be a target of RSV therapy [10]. An in-depth understanding of the virus structure, its genome, and encoded proteins will undoubtedly contribute to the development of effective new-generation therapies.

The Main Virus Antigens

The RSV G and F glycoproteins are the main virus antigens that are very important for the virus pathogenesis and the host immune response. The attachment G protein (RSV G) plays a crucial role in viral attachment to host cells, facilitating the initial step of viral infection. It is synthesized both as a membrane-bound and secreted form. The first one is responsible for viral attachment through binding to specific receptors (including glycosaminoglycans) on the host cell surface. The second one mediates immune evasion [11,12]. The membrane-bound form is a multi-domain protein. It consists of a short cytoplasmic tail, transmembrane domain, and two mucin-like regions, which are highly glycosylated, and flank two other protein regions: a central conserved domain with a CX3C motif and a heparin-binding domain [13]. The unique feature of the RSV G protein is its glycosylation pattern with ~5 N-linked glycans and ~40 O-linked glycans. It was shown that carbohydrates constitute not less than 50% of the molecular weight of the mature molecule [14,15]. RSV G protein undergoes rapid changes and variations. Antigenic differences in the G protein are the main basis for classifying RSV subtypes [16,17]. Human RSV is classified into A or B subtypes. There are nine various genotypes in RSV-A group (GA1-GA7, SAA1 and NA1) and not less than 32 genotypes in RSV-B (BA1-14, GB1-GB5, SAB1-4, URU1-2, NZB1-2, BA-CCA, BA-CCB, BA-C, CBB and CB1) [18,19]. Viruses of both subtypes commonly co-circulate during the annual epidemic period. However, RSV-A viruses are usually predominant [20,21]. Targeting the attachment protein is a potential approach for anti-RSV drug development and vaccine design. Certainly, the antigenic diversity of the protein is a major challenge to achieving these goals.
The fusion F protein (RSV F) is another main glycoprotein found on the virus surface. It plays a crucial role in the infection process not only by mediating the fusion of the viral envelope with the host cell membrane but also by assisting in virus attachment. Unlike variable RSV G protein, RSV F is highly conserved. It is synthesized as an inactive F0 precursor, post-translationally glycosylated at 5 or 6 N-glycosylation sites and cleaved into two subunits by furin-like host enzymes. The resulting F1 (C-terminal) and F2 (N-terminal) subunits are linked via two disulfide bonds [22]. The proteolytic cleavage of the RSV F protein is required for its activity. The mature protein forms compact trimers on the virus surface [23]. Cleaved and trimerized F glycoprotein adopts a metastable prefusion (RSV pre-F) conformation characterized by a compact structure with the hydrophobic fusion peptide buried within the protein [24]. During the fusion process, the protein undergoes major conformational changes and adopts extremely stable postfusion (RSV post-F) conformation, which represents the final state of the fusion protein [25]. These conformational changes are essential for the successful fusion of the virus particle with the host cell membrane during the infection process. In the RSV post-F conformation, the hydrophobic fusion peptide is exposed at the F1 subunit, allowing it to insert into the host cell membrane.
Moreover, the F1 subunit adopts, at some stage, a hairpin-like structure, bringing the viral and host cell membranes into close proximity. Stabilization of the RSV pre-F conformation revealed significant immunogenic characteristics of the protein. It is the pre-F conformation that elicits highly neutralizing antibodies, which are crucial for blocking virus infection [23,26]. Two of the RSV pre-F epitopes, site ϴ and V become particularly important for the selection of highly potent monoclonal antibodies with therapeutic potential [27]. They are present only on the RSV pre-F protein. The other antigenic sites (I, II, III and IV), which are less immunogenic, are present on both RSV pre-F and post-F forms, except for epitope I, which is present only on post-F [25,28]. The main characteristics of RSV F epitopes are presented in Table 1.
Understanding the structure and conformational changes of the fusion protein was essential for developing antiviral therapies. The RSV pre-F conformation became a crucial antigen for developing an effective RSV vaccine [28,29,30,31,32].

3. Epidemiology of RSV Virus after COVID-19 Pandemic

RSV is especially dangerous for premature infants. Also, babies younger than six months of age and children with bronchopulmonary dysplasia, congenital heart disease and immunodeficiency are at risk for RSV-associated severe illness and death [33,34]. In 2019, RSV was responsible for over 30 million lower respiratory infections, 3 million hospitalisations associated with acute respiratory infections and more than 100,000 deaths in young children (0–5 years) globally [33]. More than 95% of RSV-associated acute lower respiratory infection episodes and more than 97% of RSV-attributable deaths across all age bands were in low-income and middle-income countries (LMICs) [33]. The association of early RSV bronchiolitis with recurrent wheezing and asthma has been reported [35]. RSV also imposes a significant risk to older patients (age ≥ 65), especially with underlying comorbidities [2,3,35]. The clinical manifestations of RSV infection in this age group are nonspecific and variable; thus, diagnosis might be problematic [36]. According to late reports from the Centers for Disease Control and Prevention (CDC), due to RSV infections, there are ca. 60,000–160,000 hospitalisations among older adults and 6000–10,000 deaths in this age group annually. RSV also poses a threat to pregnant women [37]. For years, the epidemic curve for RSV was relatively constant. The COVID-19 pandemic has impacted the epidemiology of RSV, although the full extent and implications are still being studied. During the COVID-19 pandemic, epidemiologists reported a marked reduction in influenza, RSV and other respiratory viruses activity in several countries. Implementing non-pharmaceutical interventions such as social distancing and isolation aimed at reducing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission also limited the spread of RSV [38,39,40,41]. Currently, in Poland and other countries, an increase in RSV infections is reported, with an increasing number of infections among an older cohort of susceptible children [42,43,44,45]. Also, changes in RSV seasonality patterns have been observed. Typically, the RSV season occurs during the fall, lasts through the winter and ends in the late spring. During the COVID-19 pandemic, the timing and seasonality patterns have been disrupted. Numerous countries, such as Israel, Australia, South Africa, New Zealand, Italy, France, the United States and Japan, have experienced unexpected seasonality shifts [38,43,46,47]. The temporary decline in RSV circulation during the COVID-19 pandemic may have led to an increase in the number of susceptible individuals in the population, including older children and adults who may have had limited exposure to RSV in recent months. This accumulation of susceptible individuals potentially results in more significant outbreaks when RSV activity resumes.

4. Immunoprophylaxis

There are two general methods of immunoprophylaxis: passive and active immunisation, however, in the case of RSV infection, for years, only passive immunisation was available and for high-risk infants, exclusively. We are currently witnessing a breakthrough with the registration of the first two RSV vaccines for older adults and a new monoclonal neutralising antibody with a much longer half-life for infants and young children. In the near future, we can certainly expect new RSV interventions in the field of passive and active immunisation since there are several products in clinical trials. An important aspect of immunoprophylaxis is the vaccination of pregnant women [48]. The strategy to protect infants through transplacental transfer of vaccine-induced maternal antibodies is under development for RSV prevention. In the following sections, the two methods of RSV immunoprophylaxis will be discussed, both current status and prospects for the future.

4.1. Passive Immunisation Using Monoclonal Antibodies

The first passive immunoprophylaxis used in RSV infections was the intravenous administration of immune globulin RSV-IGIV (RespiGam, MedImmune, Gaithersburg, MD, USA). The product consisted of polyclonal antibodies derived from human plasma, rich in the neutralizing anti-RSV activity. RespiGam was dedicated to premature infants and young children with chronic lung disease; however, the treatment exhibited a moderate efficacy and generated high costs. Discoveries in the field of the molecular structure of the RSV F protein, together with the advances in the isolation of monoclonal antibodies, resulted in the development of palivizumab. Palivizumab (Synagis, AstraZeneca, Cambridge, UK) is a humanized murine anti-IgG1κ monoclonal antibody, which is produced via DNA technology in mouse myeloma cells. It targets the highly conserved epitope II on the RSV F protein (pre-F and post-F) and possesses fusion-inhibiting effects on the virus (RSV A and B subtypes). It is licenced as an immunoprophylaxis for preterm infants (born at ≤35 weeks of gestation) and infants younger than six months of age when the RSV infection season begins, also for children younger than two years with bronchopulmonary dysplasia or haemodynamically significant heart disease [49]. The need for monthly administration during the period of expected risk of RSV infection and relatively high cost are the heavy burden for health services and the main barriers to the extension of indications. Caserta et al. thoroughly discussed in their recently published report all these aspects and other more concerning palivizumab immunoprophylaxis [50].
There is a need for the next generation antibodies with a longer half-life and higher efficacy. Two monoclonal antibodies hoped to fulfil this need—suptavumab and motavizumab. The first one (MEDI-8897, targeting epitope V on the pre-F) successfully passed Phase I and II but was discontinued after receiving results from the Phase III study in infants. Suptavumab prevented RSV A infections while increasing the rate of RSV B. It did not reduce overall RSV hospitalisations and outpatient lower respiratory tract infections [51]. The second one, named motavizumab (MEDI-524), was derived from palivizumab and targeted the same antigenic site—epitope II. Motavizumab had 20 times higher in vitro neutralisation activity compared to palivizumab and a longer half-life [52]. Regardless of these attractive features, its clinical trial was discontinued due to increased incidence of hypersensitivity reactions. In the meantime, a monoclonal antibody D25, which selectively binds to the antigen ϴ site of the RSV pre-F, was isolated. Through binding to this specific epitope, the antibody is able to block protein conversion to the postfusion form at the stage of the prehairpin intermediate. The antibody was further optimised by introducing mutations (YTE) in the Fc region, which allowed to obtain the variant with a longer half-life and enhanced potency. This antibody variant named nirsevimab (Beyfortus, AstraZeneca, Cambridge, UK, and Sanofi, Paris, France) became the second monoclonal antibody (fully human) licensed for infant immunoprophylaxis. It is recommended for the prevention of RSV lower respiratory tract disease in infants and children up to 24 months of age with a single injection for protection throughout the RSV season. In the study among healthy late preterm and full-term infants, nirsevimab showed an efficacy of 75% against RSV lower respiratory tract infections and a reduction in RSV-related hospitalisations compared to placebo [53]. In another study among preterm infants and infants at higher risk for severe RSV infection (congenital heart and lung diseases), the efficacy and safety of nirsevimab were compared to palivizumab. The study showed a similar efficacy and safety profile to that of palivizumab [54,55]. The latest results from the Phase IIIb HARMONIE (international clinical trial) study demonstrated an 83.2% reduction in hospitalisations due to RSV-related LRT infection in infants under 12 months who received nirsevimab, compared to placebo. Nirsevimab also reduced the incidence of hospitalisations due to severe RSV-related LRT disease by 75.71% in infants under 12 months compared to placebo [56].
Clesrovimab (MK-1654) is another monoclonal antibody (fully human) with a long half-life and proven safety [57]. It targeted highly conserved site IV of the RSV F protein. Clesrovimab is currently in Phase IIb/III to evaluate the efficacy and safety in healthy pre-term and full-term infants versus placebo (NCT04767373) and in Phase III to evaluate the safety, efficacy and pharmacokinetics in infants and children at increased risk for severe RSV disease compared to palivizumab (NCT04938830). To reduce the costs of passive immunisation, other products are under development. The first one is a Ø site monoclonal antibody named RSM01. It is currently in a Phase I trial (NCT05118386) to evaluate the safety, tolerability and pharmacokinetics of single ascending doses in healthy adults. The second one is TNM001 (Trinomab Biotech, Zhuhai, China). It is a fully human monoclonal antibody which acts by targeting the RSV F protein. Currently, in Phase Ib/IIa study (NCT05630573), the safety, tolerability and pharmacokinetics profile of TNM001 injections in Chinese healthy and term infants is being assessed. A summary of monoclonal antibodies used for RSV prophylaxis is presented in Table 2.

4.2. Active Immunisation

Since the first isolation of RSV from chimpanzees with coryza, various studies have been conducted to develop an effective vaccine. The process was long and full of humble discoveries of the virus’s nature. Initially, well-tolerated formalin-inactivated RSV vaccine tested in children eventually gave aberrant results, with the death of two patients. It was found that inactivated vaccines evoked too few virus-blocking antibodies but a major inflammatory response, including pulmonary neutrophil infiltration [58]. These caused difficulties in RSV vaccine development due to obvious safety concerns. The development process was challenging due to the unique characteristics of the virus. The breakthrough in the field came after the stabilisation of the RSV pre-F conformation. Recently, two vaccines, both based on the RSV F in its pre-F form, were approved by the Food and Drug Administration (FDA). One of them is AREXEVY (GlaxoSmithKline, Brentford, UK), which has also been approved in the European Union by the European Medicines Agency (EMA). It is a monovalent adjuvanted vaccine, which contains 120 µg of recombinant RSV pre-F glycoprotein (RSVPreF3 antigen) and AS01E as an adjuvant for intramuscular injection. The RSVPreF3 antigen is produced in Chinese hamster ovary (CHO) cells using recombinant DNA technology and then thoroughly purified in a multi-step process. The AS01E adjuvant was selected in the Phase I/II trial when the safety and immunogenicity of the vaccine were studied in adults (N = 48, aged 18–40 years) and older adults (N = 1005, aged 60–80 years). The vaccine proved to be safe, and AS01E adjuvant was superior to AS01B [59]. In Phase III, participants received one dose of RSVPreF vaccine (N = 12,467) or placebo (N = 12,499). Vaccine efficacy against RSV-related lower respiratory tract disease, severe RSV-related LRT disease and RSV-related acute respiratory infection was shown to be 82.6%, 94.1% and 71.1%, respectively. Vaccine efficacy against infections of RSV subtypes A and B was similar. The safety profiles were proved regardless of underlying medical conditions [60].
The second approved vaccine is ABRYSVO (Pfizer, New York, NY, USA) bivalent vaccine. This vaccine represents a different strategy—a final formulation containing RSV pre-F antigens from both RSV subtypes A and B without any adjuvant. The ABRYSVO vaccine contains in total 120 µg of protein (60 µg RSVpreF A and 60 µg RSVpreF B) for single intramuscular injection. Both proteins are expressed in CHO cells and then purified through a series of column chromatography and filtration steps. In Phase IIa (healthy adults, 18 to 50 years), the vaccine showed 86.7% efficacy for symptomatic RSV infection confirmed by any detectable viral RNA on at least two consecutive days (NCT05035212, RENOIR) [61]. In Phase 3, the efficacy, immunogenicity, and safety of a single dose of the vaccine were studied in adults 60 years of age and older. Participants had received RSVpreF vaccine (N = 17,215) or placebo (N = 17,069). The authors showed that vaccine efficacy for preventing RSV-associated LRT illness and RSV-associated acute respiratory illness was 85.7% and 62.1%, respectively [62]. Both already registered vaccines have been further studied for other clinical purposes. The RSVpreF vaccine is checked for immunogenicity, safety and reactogenicity when co-administered with the influenza vaccine (Fluzone High-Dose Quadrivalent, Sanofi, Paris, France) in adults aged 65 and older compared to separate administration of both vaccines (NCT05559476, NCT05568797). Also, the bivalent RSVpreF vaccine is studied to evaluate the safety, tolerability and immunogenicity when co-administered with seasonal inactivated influenza vaccine in adults aged 65 and older (NCT05301322). Lately (July 2023), the bivalent RSVpreF vaccine (ABRYSVO) was approved by EMA for infants from birth to 6 months of age (after maternal immunisation during pregnancy) and for pregnant women between 24 and 36 weeks of pregnancy. It is the first vaccine against RSV approved to protect pregnant women by active immunization and their infants in consequence by passive immunisation. It was approved after receiving the results from the Phase III clinical study. This was run to study the efficacy and safety of maternal immunisation against medically attended LRT illness (MA-LRTI). The efficacy, safety and immunogenicity were assessed in infants born to healthy women who received the vaccine while being pregnant. Simultaneously, the safety and immunogenicity in the pregnant women were evaluated (NCT04424316). In this study, 3682 pregnant women received the vaccine, and 3676 received a placebo, then 3570 (vaccinated mothers) and 3558 infants (placebo mothers) were tested. The results showed that vaccine efficacy for preventing medically attended severe LRT illness within 90 and 180 days after birth was 81.8% and 69.4%, respectively [63]. Under clinical development is another maternal vaccine, which is based on RSVpreF antigen. In the Phase IIb study, the safety, tolerability, and immunogenicity of an RSVpreF subunit vaccine (RSV vaccine) are evaluated in pregnant women who receive either one of two doses of the vaccine, formulated with/without aluminium hydroxide or placebo, as well as the safety and characteristics of transplacentally transferred antibodies in their infants (NCT04032093). In this clinical study, 406 women and 403 infants were included, and 327 women (80.5%) received the RSVpreF vaccine. The RSVpreF vaccine proved to elicit neutralising antibody responses with efficient transplacental transfer [64].
The COVID-19 pandemic undoubtedly brought a breakthrough in the field of vaccines since for the first time in the history, vaccines based on nucleic acids such as mRNA vaccines were used. The possibility of using this type of vaccines was the result of many years of brilliant scientific research, which has been honoured this year with the Nobel Prize. The most advance mRNA vaccine against RSV is the mRNA-1345 vaccine developed by Moderna. The vaccine is adjuvanted with LNP and aim to produce RSV F protein. It is currently under Phase III of clinical trials [NCT05330975]. The study is divided into Part A, B and C. The purpose of Part A study is to evaluate the safety, tolerability and immunogenicity of mRNA-1345 co-administered with a seasonal influenza vaccine (Alfuria®, quadrivalent). The second purpose is to evaluate both the impact of co-administered influenza vaccine on the immune response to RSV-A and the impact of RSV vaccine on the immune response to influenza. The purpose of Part B study is to evaluate the safety, tolerability and immunogenicity of mRNA-1345 co-administered with mRNA-1273.214. The other purpose of the study is to evaluate both the impact of co-administered mRNA-1273.214 vaccine on the immune response to RSV-A and to evaluate the effect of the RSV vaccine on the immune response to SARS-CoV-2. Finally, the purpose of Part C study is to evaluate the safety and tolerability of booster dose of mRNA 1345 administered one year after a primary dose. It is a continuation of the study after obtaining positive results from Phase I (NCT04528719) when the tolerability and reactogenicity of a single injection of up to five dose levels of mRNA-1345 in younger adults, women of child-bearing potential and older adults were assessed, and from Phase II (NCT05127434) when the efficacy of a single dose of mRNA-1345 vaccine in the prevention of a first episode of RSV-associated lower respiratory tract disease as compared with a placebo was assessed. The mRNA-1345 vaccine is also studied in children who are (RSV)-seropositive, as a Phase I of clinical trial (NCT04528719). In the paediatric group, another mRNA vaccine is studied. This is the mRNA-1365 vaccine developed by Sanofi. Phase I trials have recently begun aim to assess the safety and immunogenicity of mRNA-1365, an mRNA vaccine targeting RSV and human metapneumovirus (hMPV), and mRNA-1345 in infants aged 5 months to <24 months (NCT05743881).
Currently, several other experimental vaccines are under clinical trials, including recombinant subunit vaccines, live attenuated vaccines and viral vector-based vaccines (see Table 3). The latter may soon be approved for use.

5. Conclusions

RSV prevention has been a challenge for many years. Recently licenced new passive (Nirsevimab) and active (AREXEVY and ABRYSVO) prevention options and the results from maternal immunisation demonstrate that there is hope for broader medical choices, which will be available for paediatric groups and adults.

Author Contributions

Conceptualization, E.K., H.C., P.Z. and A.M.; methodology, E.K.; investigation, E.K.; resources, E.K.; writing—original draft preparation, E.K.; writing—review and editing, P.Z.; visualization, E.K. and P.Z.; supervision, H.C. and A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

BCGBacillus Calmette–Guérin
CDCCenters for Disease Control and Prevention
CHOChinese hamster ovary
COVID-19Coronavirus disease 2019
EMAEuropean Medicines Agency
FDAFood and Drug Administration
hMPVhuman metapneumovirus
IgG1Immunoglobulin G subclass 1
kbpKilo-base pairs
LNPlipid nanoparticle
LRTLower respiratory tract
mAbMonoclonal antibody
MA-LRTIMedically attended LRT illness
mRNAMessenger ribonucleic acid
ORFOpen reading frame
RSVRespiratory syncytial virus
RNARibonucleic acid
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
SVLPSynthetic virus-like particle
TdapDiphtheria, tetanus, pertussis vaccine with lower concentration of diphtheria and pertussis toxoids
URTUpper respiratory tract
VLPVirus-like particle.

References

  1. Resch, B. Burden of respiratory syncytial virus infection in young children. World J. Clin. Pediatr. 2012, 1, 8–12. [Google Scholar] [CrossRef]
  2. Ambrosch, A.; Luber, D.; Klawonn, F.; Kabesch, M. Focusing on severe infections with the respiratory syncytial virus (RSV) in adults: Risk factors, symptomatology and clinical course compared to influenza A/B and the original SARS-CoV-2. J. Clin. Virol. 2023, 161, 105399, corrected in J. Clin. Virol. 2023, 12, 105443. [Google Scholar] [CrossRef] [PubMed]
  3. Prasad, N.; Walker, T.A.; Waite, B.; Wood, T.; Trenholme, A.A.; Baker, M.G.; McArthur, C.; Wong, C.A.; Grant, C.C.; Huang, Q.S.; et al. Respiratory Syncytial Virus-Associated Hospitalizations Among Adults with Chronic Medical Conditions. Clin. Infect. Dis. 2021, 73, e158–e163. [Google Scholar] [CrossRef] [PubMed]
  4. Ke, Z.; Dillard, R.S.; Chirkova, T.; Leon, F.; Stobart, C.C.; Hampton, C.M.; Strauss, J.D.; Rajan, D.; Rostad, C.A.; Taylor, J.V.; et al. The Morphology and Assembly of Respiratory Syncytial Virus Revealed by Cryo-Electron Tomography. Viruses 2018, 10, 446. [Google Scholar] [CrossRef] [PubMed]
  5. Liljeroos, L.; Krzyzaniak, M.A.; Helenius, A.; Butcher, S.J. Architecture of respiratory syncytial virus revealed by electron cryotomography. Proc. Natl. Acad. Sci. USA 2013, 110, 11133–11138. [Google Scholar] [CrossRef]
  6. Fearns, R.; Collins, P.L. Role of the M2-1 transcription antitermination protein of respiratory syncytial virus in sequential transcription. J. Virol. 1999, 73, 5852–5864. [Google Scholar] [CrossRef] [PubMed]
  7. Bermingham, A.; Collins, P.L. The M2-2 protein of human respiratory syncytial virus is a regulatory factor involved in the balance between RNA replication and transcription. Proc. Natl. Acad. Sci. USA 1999, 96, 11259–11264. [Google Scholar] [CrossRef] [PubMed]
  8. Collins, P.L.; Fearns, R.; Graham, B.S. Respiratory Syncytial Virus: Virology, Reverse Genetics, and Pathogenesis of Disease. Curr. Top. Microbiol. Immunol. 2013, 372, 3–38. [Google Scholar] [CrossRef]
  9. Teng, M.N.; Collins, P.L. Altered growth characteristics of recombinant respiratory syncytial viruses which do not produce NS2 protein. J. Virol. 1999, 73, 466–473. [Google Scholar] [CrossRef]
  10. Yang, P.; Zheng, J.; Wang, S.; Liu, P.; Xie, M.; Zhao, D. Respiratory syncytial virus nonstructural proteins 1 and 2 are crucial pathogenic factors that modulate interferon signaling and Treg cell distribution in mice. Virology 2015, 485, 223–232. [Google Scholar] [CrossRef]
  11. Roberts, S.R.; Lichtenstein, D.; Ball, L.A.; Wertz, G.W. The membrane-associated and secreted forms of the respiratory syncytial virus attachment glycoprotein G are synthesized from alternative initiation codons. J. Virol. 1994, 68, 4538–4546. [Google Scholar] [CrossRef] [PubMed]
  12. Bukreyev, A.; Yang, L.; Fricke, J.; Cheng, L.; Ward, J.M.; Murphy, B.R.; Collins, P.L. The secreted form of respiratory syncytial virus G glycoprotein helps the virus evade antibody-mediated restriction of replication by acting as an antigen decoy and through effects on Fc receptor-bearing leukocytes. J. Virol. 2008, 82, 12191–12204. [Google Scholar] [CrossRef] [PubMed]
  13. Collins, P.L.; Mottet, G. Oligomerization and post-translational processing of glycoprotein G of human respiratory syncytial virus: Altered O-glycosylation in the presence of brefeldin A. J. Gen. Virol. 1992, 73, 849–863. [Google Scholar] [CrossRef] [PubMed]
  14. Wertz, G.W.; Collins, P.L.; Huang, Y.; Gruber, C.; Levine, S.; Ball, L.A. Nucleotide sequence of the G protein gene of human respiratory syncytial virus reveals an unusual type of viral membrane protein. Proc. Natl. Acad. Sci. USA 1985, 82, 4075–4079. [Google Scholar] [CrossRef] [PubMed]
  15. Ball, L.A.; Young, K.K.; Anderson, K.; Collins, P.L.; Wertz, G.W. Expression of the major glycoprotein G of human respiratory syncytial virus from recombinant vaccinia virus vectors. Proc. Natl. Acad. Sci. USA 1986, 83, 246–250. [Google Scholar] [CrossRef] [PubMed]
  16. Anderson, L.J.; Hierholzer, J.C.; Tsou, C.; Hendry, R.M.; Fernie, B.F.; Stone, Y.; McIntosh, K. Antigenic characterization of respiratory syncytial virus strains with monoclonal antibodies. J. Infect. Dis. 1985, 151, 623–633. [Google Scholar] [CrossRef] [PubMed]
  17. Cane, P.A. Molecular epidemiology of respiratory syncytial virus. Rev. Med. Virol. 2001, 11, 103–116. [Google Scholar] [CrossRef]
  18. Mufson, M.A.; Orvell, C.; Rafnar, B.; Norrby, E. Two distinct subtypes of human respiratory syncytial virus. J. Gen. Virol. 1985, 66, 2111–2124. [Google Scholar] [CrossRef]
  19. Yu, J.M.; Fu, Y.-H.; Peng, X.-L.; Zheng, Y.-P.; He, J.-S. Genetic diversity and molecular evolution of human respiratory syncytial virus A and B. Sci. Rep. 2021, 11, 12941–12951. [Google Scholar] [CrossRef]
  20. Tabatabai, J.; Prifert, C.; Pfeil, J.; Grulich-Henn, J.; Schnitzler, P. Novel respiratory syncytial virus (RSV) genotype ON1 predominates in Germany during winter season 2012-13. PLoS ONE 2014, 9, e109191-10. [Google Scholar] [CrossRef]
  21. Rahombanjanahary, N.H.R.; Rybkina, K.; Randriambolamanantsoa, T.H.; Helisoa Razafimanjato, H.; Heraud, J.-M. Genetic diversity and molecular epidemiology of respiratory syncytial virus circulated in Antananarivo, Madagascar, from 2011 to 2017: Predominance of ON1 and BA9 genotypes. J. Clin. Virol. 2020, 129, 104506. [Google Scholar] [CrossRef] [PubMed]
  22. Gonzalez-Reyes, L.; Ruiz-Arguello, M.B.; Garcia-Barreno, B. Cleavage of the human respiratory syncytial virus fusion protein at two distinct sites is required for activation of membrane fusion. Proc. Natl. Acad. Sci. USA 2001, 98, 9859–9864. [Google Scholar] [CrossRef] [PubMed]
  23. McLellan, J.S.; Chen, M.; Leung, S.; Graepel, K.W.; Du, X.; Yang, Y.; Zhou, T.; Baxa, U.; Yasuda, E.; Beaumont, T.; et al. Structure of RSV fusion glycoprotein trimer bound to a prefusion-specific neutralizing antibody. Science 2013, 340, 1113–1117. [Google Scholar] [CrossRef] [PubMed]
  24. Gilman, M.S.A.; Furmanova-Hollenstein, P.; Pascual, G.; van ‘t Wout, A.B.; Langedijk, J.P.M. Transient opening of trimeric prefusion RSV F proteins. Nat. Commun. 2019, 10, 2105–2117. [Google Scholar] [CrossRef] [PubMed]
  25. McLellan, J.S.; Yang, Y.; Graham, B.S.; Kwong, P.D. Structure of the respiratory syncytial virus fusion glycoprotein in the post-fusion conformation reveals preservation of neutralizing epitopes. J. Virol. 2011, 85, 7788–7796. [Google Scholar] [CrossRef] [PubMed]
  26. McLellan, J.S.; Chen, M.; Joyce, M.G.; Sastry, M.; Stewart-Jones, G.B.E.; Yang, Y.; Zhang, B.; Chen, L.; Srivatsan, S.; Zheng, A.; et al. Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science 2013, 342, 592–598. [Google Scholar] [CrossRef] [PubMed]
  27. McLellan, J.S.; Chen, M.; Chang, J.-S.; Yang, Y.; Kim, A.; Graham, B.S.; Kwong, P. Structure of a major antigenic site on the respiratory syncytial virus fusion glycoprotein in complex with neutralizing antibody 101F. J. Virol. 2010, 84, 12236–12244. [Google Scholar] [CrossRef]
  28. Melero, J.A.; Mas, V.; McLellan, J.S. Structural, antigenic and immunogenic features of respiratory syncytial virus glycoproteins relevant for vaccine development. Vaccine 2017, 35, 461–468. [Google Scholar] [CrossRef]
  29. Liang, B.; Ngwuta, J.O.; Surman, S.; Kabatova, B.; Liu, X.; Lingemann, M.; Liu, X.; Yang, L.; Herbert, R.; Swerczek, J.; et al. Improved Prefusion Stability, Optimized Codon Usage, and Augmented Virion Packaging Enhance the Immunogenicity of Respiratory Syncytial Virus Fusion Protein in a Vectored-Vaccine Candidate. J. Virol. 2017, 91, e00189-17. [Google Scholar] [CrossRef]
  30. Liang, B.; Matsuoka, Y.; Nouën, C.L.; Liu, X.; Herbert, R.; Swerczek, J.; Santos, C.; Paneru, M.; Collins, P.L.; Buchholz, U.J.; et al. A Parainfluenza Virus Vector Expressing the Respiratory Syncytial Virus (RSV) Prefusion F Protein Is More Effective than RSV for Boosting a Primary Immunization with RSV. J. Virol. 2020, 95, e01512-20. [Google Scholar] [CrossRef]
  31. Zhang, L.; Durr, E.; Galli, J.D.; Cosmi, S.; Cejas, P.J.; Luo, B.; Touch, S.; Parmet, P.; Fridman, A.; Espeseth, A.S.; et al. Design and characterization of a fusion glycoprotein vaccine for Respiratory Syncytial Virus with improved stability. Vaccine 2018, 36, 8119–8130. [Google Scholar] [CrossRef] [PubMed]
  32. Patel, N.; Massare, M.J.; Tian, J.H.; Guebre-Xabier, M.; Lu, H.; Zhou, H.; Maynard, E.; Scott, D.; Ellingsworth, L.; Glenn, G.; et al. Respiratory syncytial virus prefusogenic fusion (F) protein nanoparticle vaccine: Structure, antigenic profile, immunogenicity, and protection. Vaccine 2019, 37, 6112–6124. [Google Scholar] [CrossRef] [PubMed]
  33. Li, Y.; Wang, X.; Blau, D.M.; Caballero, M.T.; Feikin, D.R.; Gill, C.J.; Madhi, S.A.; Omer, S.B.; Simões, E.A.F.; Campbell, H.; et al. Respiratory Virus Global Epidemiology Network; Harish Nair; RESCEU investigators. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in children younger than 5 years in 2019: A systematic analysis. Lancet 2022, 399, 2047–2064. [Google Scholar] [CrossRef] [PubMed]
  34. Meert, K.; Heidemann, S.; Lieh-Lai, M.; Sarnaik, A.P. Clinical characteristics of respiratory syncytial virus infections in healthy versus previously compromised host. Pediatr. Pulmonol. 1989, 7, 167–170. [Google Scholar] [CrossRef] [PubMed]
  35. Binns, E.; Tuckerman, J.; Licciardi, P.V.; Wurzel, D. Respiratory syncytial virus, recurrent wheeze and asthma: A narrative review of pathophysiology, prevention and future directions. J. Paediat. Child Health 2022, 58, 1741–1746. [Google Scholar] [CrossRef] [PubMed]
  36. Falsey, A.R.; Walsh, E.E. Respiratory Syncytial Virus Infection in Elderly Adults. Drugs Aging 2005, 22, 577–587. [Google Scholar] [CrossRef] [PubMed]
  37. Kachikis, A.B.; Cho, H.; Englund, J.A. Respiratory Syncytial Virus-An Update for Prenatal and Primary Health Providers. Obstet. Gynecol. Clin. N. Am. 2023, 50, 421–437. [Google Scholar] [CrossRef]
  38. Chuang, Y.-C.; Lin, K.-P.; Wang, L.-A.; Yeh, T.-K.; Liu, P.-Y. The Impact of the COVID-19 Pandemic on Respiratory Syncytial Virus Infection: A Narrative Review. Infect. Drug Resist. 2023, 16, 661–675. [Google Scholar] [CrossRef]
  39. Gastaldi, A.; Donà, D.; Barbieri, E.; Giaquinto, C.; Bont, J.; Baraldi, E. COVID-19 Lesson for Respiratory Syncytial Virus (RSV): Hygiene Works. Children 2021, 8, 1144. [Google Scholar] [CrossRef]
  40. Torres-Fernandez, D.; Casellas, A.; Mellado, M.J.; Calvo, C.; Bassat, Q. Acute bronchiolitis and respiratory syncytial virus seasonal transmission during the COVID-19 pandemic in Spain: A national perspective from the pediatric Spanish Society (AEP). J. Clin. Virol. 2021, 145, 105027. [Google Scholar] [CrossRef]
  41. Bardsley, M.; Morbey, R.A.; Hughes, H.E.; Beck, C.R.; Watson, C.H.; Zhao, H.; Ellis, J.; Smith, G.E.; Elliot, A.J. Epidemiology of respiratory syncytial virus in children younger than 5 years in England during the COVID-19 pandemic, measured by laboratory, clinical, and syndromic surveillance: A retrospective observational study. Lancet Infect. Dis. 2023, 23, 56–66. [Google Scholar] [CrossRef] [PubMed]
  42. Rao, S.; Armistead, I.; Messacar, K.; Alden, N.B.; Schmoll, E.; Austin, E.; Dominguez, S.R. Shifting Epidemiology and Severity of Respiratory Syncytial Virus in Children During the COVID-19 Pandemic. JAMA Pediatr. 2023, 15, e231088. [Google Scholar] [CrossRef] [PubMed]
  43. Pruccoli, G.; Castagno, E.; Raffaldi, I.; Denina, M.; Barisone, E.; Baroero, L.; Timeus, F.; Rabbone, I.; Monzani, A.; Terragni, G.A.; et al. The Importance of RSV Epidemiological Surveillance: A Multicenter Observational Study of RSV Infection during the COVID-19 Pandemic. Viruses 2023, 15, 280. [Google Scholar] [CrossRef] [PubMed]
  44. Rodríguez-Fernández, R.; González-Martínez, F.; Perez-Moreno, J.; González-Sánchez, M.I.; de la Mata Navazo, S.; Toledo Del Castillo, B.; Lozano, J.S.; Espinosa, L.V.; Manso Perez, S.; Berlanga, M.M.; et al. Clinical Phenotype of Respiratory Syncytial Virus Bronchiolitis before and during the Coronavirus Disease 2019 Pandemic. Am. J. Perinatol. 2022. [Google Scholar] [CrossRef] [PubMed]
  45. Rząd, M.; Kanecki, K.; Lewtak, K.; Tyszko, P.; Szwejkowska, M.; Goryński, P.; Nitsch-Osuch, A. Human Respiratory Syncytial Virus Infections among Hospitalized Children in Poland during 2010-2020: Study Based on the National Hospital Registry. J. Clin. Med. 2022, 11, 6451. [Google Scholar] [CrossRef]
  46. Miyama, T.; Kakimoto, K.; Iritani, N.; Nishio, T.; Ukai, T.; Satsuki, Y.; Yamanaka, Y.; Nishida, Y.; Shintani, A.; Motomura, K. Exploring the threshold for the start of respiratory syncytial virus infection epidemic season using sentinel surveillance data in Japan. Front. Public Health 2023, 11, 1062726. [Google Scholar] [CrossRef]
  47. Hamid, S.; Winn, A.; Parikh, R.; Jones, J.M.; McMorrow, M.; Prill, M.M.; Silk, B.J.; Scobie, H.M.; Hall, A.J. Seasonality of Respiratory Syncytial Virus—United States, 2017–2023. Morb. Mortal. Wkly. Rep. 2023, 72, 355–361. [Google Scholar] [CrossRef]
  48. Langel, S.N.; Blasi, M.; Permar, S.R. Maternal immune protection against infectious diseases. Cell Host Microbe 2022, 30, 660–674. [Google Scholar] [CrossRef]
  49. European Medicines Agency. Available online: https://www.ema.europa.eu/en/documents/product-information/synagis-epar-product-information_en.pdf (accessed on 28 October 2023).
  50. Caserta, M.T.; O’Leary, S.T.; Munoz, F.M.; Ralston, S.L. Palivizumab Prophylaxis in Infants and Young Children at Increased Risk of Hospitalization for Respiratory Syncytial Virus Infection. Pediatrics 2023, 152, e2023061803. [Google Scholar] [CrossRef]
  51. Simões, E.A.; Forleo-Neto, E.; Geba, G.P.; Kamal, M.; Yang, F.; Cicirello, H.; Houghton, M.R.; Rideman, R.; Zhao, Q.; Benvin, S.L.; et al. Suptavumab for the prevention of medically attended respiratory syncytial virus infection in preterm infants. Clin. Infect. Dis. 2021, 73, e4400–e4408. [Google Scholar] [CrossRef]
  52. Carbonell-Estrany, X.; Simões, E.A.; Dagan, R.; Hall, C.B.; Harris, B.; Hultquist, M.; Connor, E.M.; Losonsky, G.A.; Motavizumab Study Group. Motavizumab for prophylaxis of respiratory syncytial virus in high-risk children: A noninferiority trial. Pediatrics 2010, 125, e35–e51. [Google Scholar] [CrossRef]
  53. Hammitt, L.L.; Dagan, R.; Yuan, Y.; Baca Cots, M.; Bosheva, M.; Madhi, S.A.; Muller, W.J.; Zar, H.J.; Brooks, D.; Grenham, A.; et al. Nirsevimab for Prevention of RSV in Healthy Late-Preterm and Term Infants. N. Engl. J. Med. 2022, 386, 837–846. [Google Scholar] [CrossRef]
  54. Domachowske, J.; Madhi, S.A.; Simões, E.A.F.; Atanasova, V.; Cabañas, F.; Furuno, K.; Garcia-Garcia, M.L.; Grantina, I.; Nguyen, K.A.; Brooks, D.; et al. Safety of Nirsevimab for RSV in Infants with Heart or Lung Disease or Prematurity. N. Engl. J. Med. 2022, 386, 892–894. [Google Scholar] [CrossRef] [PubMed]
  55. Simões, E.A.F.; Madhi, S.A.; Muller, W.J.; Atanasova, V.; Bosheva, M.; Cabañas, F.; Cots, M.B.; Domachowske, J.; Garcia-Garcia, M.L.; Grantina, I.; et al. Efficacy of nirsevimab against respiratory syncytial virus lower respiratory tract infections in preterm and term infants, and pharmacokinetic extrapolation to infants with congenital heart disease and chronic lung disease: A pooled analysis of randomised controlled trials. Lancet Child Adolesc. Health 2023, 7, 180–189. [Google Scholar] [CrossRef] [PubMed]
  56. Drysdale, S. Efficacy of nirsevimab against RSV lower respiratory tract infection hospitalization in infants: Preliminary data from the HARMONIE phase 3b trial. In Proceedings of the 41st Annual Meeting of the European Society for Paediatric Infectious Diseases in Lisbon, Online, 8–12 May 2023. [Google Scholar]
  57. Aliprantis, A.O.; Wolford, D.; Caro, L.; Maas, B.M.; Ma, H.; Montgomery, D.L.; Sterling, L.M.; Hunt, A.; Cox, K.S.; Vora, K.A.; et al. A Phase 1 Randomized, Double-Blind, Placebo-Controlled Trial to Assess the Safety, Tolerability, and Pharmacokinetics of a Respiratory Syncytial Virus Neutralizing Monoclonal Antibody MK-1654 in Healthy Adults. Clin. Pharmacol. Drug Dev. 2021, 10, 556–566. [Google Scholar] [CrossRef] [PubMed]
  58. Murphy, B.R.; Walsh, E.E. Formalin-inactivated respiratory syncytial virus vaccine induces antibodies to the fusion glycoprotein that are deficient in fusion-inhibiting activity. J. Clin. Microbiol. 1988, 26, 1595–1597. [Google Scholar] [CrossRef] [PubMed]
  59. Leroux-Roels, I.; Davis, M.G.; Steenackers, K.; Essink, B.; Vandermeulen, C.; Fogarty, C.; Andrews, C.P.; Kerwin, E.; David, M.-P.; Fissette, L.; et al. Safety and Immunogenicity of a Respiratory Syncytial Virus Prefusion F (RSVPreF3) Candidate Vaccine in Older Adults: Phase 1/2 Randomized Clinical Trial. J. Infect. Dis. 2023, 227, 761–772. [Google Scholar] [CrossRef]
  60. Papi, A.; Ison, M.G.; Langley, J.M.; Lee, D.-G.; Leroux-Roels, I.; Martinon-Torres, F.; Schwarz, T.F.; van Zyl-Smit, R.N.; Campora, L.; Dezutter, N.; et al. Respiratory Syncytial Virus Prefusion F Protein Vaccine in Older Adults. N. Engl. J. Med. 2023, 16, 595–608. [Google Scholar] [CrossRef]
  61. Schmoele-Thoma, B.; Zareba, A.M.; Jiang, Q.; Maddur, M.S.; Danaf, R.; Mann, A.; Eze, K.; Fok-Seang, J.; Kabir, G.; Catchpole, A.; et al. Vaccine Efficacy in Adults in a Respiratory Syncytial Virus Challenge Study. N. Engl. J. Med. 2022, 386, 2377–2386. [Google Scholar] [CrossRef]
  62. Walsh, E.E.; Pérez Marc, G.; Zareba, A.M.; Falsey, A.R.; Jiang, Q.; Patton, M.; Polack, F.P.; Llapur, C.; Doreski, P.A.; Ilangovan, K.; et al. Efficacy and Safety of a Bivalent RSV Prefusion F Vaccine in Older Adults. N. Engl. J. Med. 2023, 388, 1465–1477. [Google Scholar] [CrossRef]
  63. Kampmann, B.; Madhi, S.A.; Munjal, I.; Simões, E.A.F.; Pahud, B.A.; Llapur, C.; Baker, J.; Pérez Marc, G.; Radley, D.; Shittu, E.; et al. Bivalent Prefusion F Vaccine in Pregnancy to Prevent RSV Illness in Infants. N. Engl. J. Med. 2023, 388, 1451–1464. [Google Scholar] [CrossRef] [PubMed]
  64. Simões, E.A.F.; Center, K.J.; Tita, A.T.N.; Swanson, K.A.; Radley, D.; Houghton, J.; McGrory, S.B.; Gomme, E.; Anderson, M.; Roberts, J.P.; et al. Prefusion F Protein-Based Respiratory Syncytial Virus Immunization in Pregnancy. N. Engl. J. Med. 2022, 386, 1615–1626. [Google Scholar] [CrossRef] [PubMed]
Table 1. RSV F epitopes and their main characteristic.
Table 1. RSV F epitopes and their main characteristic.
Site NameRSV F ConformationCharacteristicProduct
θpre-Fα-helix from F1 (aa 196–210) and a strand from F2 (aa 62–69), D25-specific antibody, AM22-specific antibodyNirsevimab,
RSM01 (under clinical development)
Ipost-Flocated at F2 subunit, Pro389, 131-2a-
IIpre-F and post-Fhelix-turn-helix motif from F1 (aa 253–278), conserved epitope, Mota-specific antibodyPalivizumab,
Motavizumab (discontinued)
IIIpre-F and post-Ftwo anti-parallel β-strands, MPE8-specific antibody-
IVpre-F and post-Flinear epitope (aa 422–436), 101F-specific antibody, does not blocking virus attachment.Clesrovimab (under clinical development)
Vpre-Fα2-α3 and β3-β4, located between site Ø and III on the RSV pre-F, AM14-specific antibodySuptavumab (discontinued)
Abbreviations: RSV—respiratory syncytial virus.
Table 2. RSV immunoprophylaxis by target populations. Monoclonal antibodies are categorised into sequences of clinical phases (I, II, III) for two different population groups: paediatric and adults.
Table 2. RSV immunoprophylaxis by target populations. Monoclonal antibodies are categorised into sequences of clinical phases (I, II, III) for two different population groups: paediatric and adults.
PopulationPhase IPhase IIPhase IIIMarket Approved
Children--Clesrovimab (MK-1654) fully human mAb (IgG1) binding RSV F protein site IV, half-life: 73–88 days, single injectionPalivizumab Humanized mAb (IgG1) binding RSV F protein site II, half-life: 20 days, monthly intramuscular injection
TNM001 human anti-RSV mAb injection--Nirsevimab fully human mAb (IgG1) binding RSV F protein site Ø, YTE mutation of the Fc, half-life: 65–70 days, single intramuscular injection
AdultsRSM01 Humanized mAb (IgG1) binding RSV F protein site Ø, half-life: 65–70 days, single injection---
Abbreviations: IgG1—immunoglobulin G subclass 1; mAb—monoclonal antibody; RSV—respiratory syncytial virus.
Table 3. RSV vaccines by target populations. Vaccine candidates are categorised into sequences of clinical phases (I, II, III) for different population groups: paediatric, maternal and adults/elderly. Various type of vaccines are marked with colours: blue refers to recombinant subunit vaccine; green: live attenuated; orange: mRNA; red: recombinant viral vector; grey: virus-like particle (VLP).
Table 3. RSV vaccines by target populations. Vaccine candidates are categorised into sequences of clinical phases (I, II, III) for different population groups: paediatric, maternal and adults/elderly. Various type of vaccines are marked with colours: blue refers to recombinant subunit vaccine; green: live attenuated; orange: mRNA; red: recombinant viral vector; grey: virus-like particle (VLP).
PopulationPhase IPhase IIPhase IIIMarket Approved
ChildrenPIV5-vectored RSV Vaccine (BLB-201) (NCT05655182) live attenuated chimeric, antigen: RSV fusion F, intranasal route
MV-012-968 (NCT04909021, NCT04444284), live, attenuated target organism, antigen: all viral proteins, intranasal route
LID/ΔM2-2/1030s (NCT04520659), live, attenuated target organism, antigen: all viral proteins except M2-2, intranasal route
RSV NS2/Δ1313/I1314L, RSV 6120/ΔNS2/1030s, RSV 276 (NCT03916185), live attenuated, antigen: all viral proteins except NS2, intranasal route
RSV6120/∆NS1, RSV6120/F1/G2/∆NS1 (NCT03596801), live attenuated, antigen: all viral proteins except NS2, intranasal route
mRNA-1345 (NCT04528719), antigen: RSVF
mRNA-1365
mRNA-1345
(NCT05743881)
human metapneumovirus (hMPV
antigen: RSVF
VAD00001 (NCT04491877), live, attenuated, antigen: all viral proteins, intranasal route-ABRYSVO® recombinant subunit, antigen: RSV preF A and RSV preF B
Pregnant-RSV (NCT04032093), recombinant subunit, antigen: RSV F, with aluminium salt
RSV vaccine given together with Tdap (NCT04071158), recombinant subunit, antigen: RSV F, with aluminium salt
RSVpreF (NCT04424316), recombinant subunit, antigen: RSV preFABRYSVO® recombinant subunit, antigen: RSV preF A and RSV preF B
Adults and ElderlyCPI-RSV-F Vaccine (NCT05281263) live attenuated chimeric, antigen: RSV fusion F, intranasal route
CodaVaxRSV (NCT04295070), live attenuated, antigen: codon deoptimized RSV, intranasal route
IVX-A12 (RSV/hMPV) (NCT05664334), bivalent combination respiratory syncytial virus (RSV)/human metapneumovirus (hMPV) virus-like particle (VLP) vaccine, with MF59 adjuvant
V-306 (NCT04519073), synthetic virus-like particle (SVLP) vaccine, with Pam2Cys adjuvant
mRNA-1345, mRNA-1045, mRNA-1230 (NCT05585632), multi-component, antigen: RSVF/Flu-HA/SARS-CoV-2S
mRNA LNP (NCT05639894), vaccine candidate formulated with two different lipid nanoparticles (LNPs) CL-0059 and CL-0137
rBCG-N-hRSV (NCT03213405) recombinant Mycobacterium bovis BCG vaccine that expresses the human RSV nucleoprotein (N)
BARS13 (NCT04681833) recombinant subunit, antigen: rRSV-G-protein, with CsA adjuvant
VN-0200 (NCT05547087), recombinant subunit, antigen: VAGA-9001a, with MABH-9002b adjuvant
RSVPreF (NCT04785612), recombinant subunit, antigen: RSV F
RSVPreF together with SIIV (NCT03529773), recombinant subunit, antigen: RSV F, with aluminium salt
MV-012-968 (NCT04690335), live, attenuated target organism, antigen: all viral proteins, intranasal route
mRNA-1345 (NCT05127434), antigen: RSVF
MVA-BN-RSV (NCT05238025) recombinant viral vector, antigen: two surface proteins and internal conserved protein
Ad26.RSV.PreF (NCT05070546, NCT05071313, NCT05083585, NCT05101486, NCT0524243, NCT05327816), live attenuated, antigen: RSV fusion F
mRNA-1345 (NCT05330975), antigen: RSVF, co-administered with a seasonal influenza vaccine (Afluria® Quadrivalent)
AREXEVY® recombinant subunit, antigen: RSVPreF3, with AS01E adjuvant
ABRYSVO® recombinant subunit, antigen: RSV preF A and RSV preF B
Abbreviations: BCG—Bacillus Calmette–Guérin; hMPV—human metapneumovirus; LNP—lipid nanoparticle; mRNA—messenger ribonucleic acid; RSV—respiratory syncytial virus; SVLP—synthetic virus-like particle; Tdap—diphtheria, tetanus, pertussis vaccine with lower concentration of diphtheria and pertussis toxoids; VLP—virus-like particle.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kopera, E.; Czajka, H.; Zapolnik, P.; Mazur, A. New Insights on Respiratory Syncytial Virus Prevention. Vaccines 2023, 11, 1797. https://doi.org/10.3390/vaccines11121797

AMA Style

Kopera E, Czajka H, Zapolnik P, Mazur A. New Insights on Respiratory Syncytial Virus Prevention. Vaccines. 2023; 11(12):1797. https://doi.org/10.3390/vaccines11121797

Chicago/Turabian Style

Kopera, Edyta, Hanna Czajka, Paweł Zapolnik, and Artur Mazur. 2023. "New Insights on Respiratory Syncytial Virus Prevention" Vaccines 11, no. 12: 1797. https://doi.org/10.3390/vaccines11121797

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop