Respiratory syncytial virus vaccines: the time has finally come (2023)

Vaccine

Bundle 34, number 30,

June 24, 2016

, page 3535-3541

Author links open overlay panel

Abstract

Respiratory syncytial viruscauses significantPublic healthstrain, particularly in very young infants and the frail elderly. Inheritance of RSV enhanced disease (ERD) from whole formalin-inactivatedRSV-vaccineand complex biologyvirusand newborns have delayed the development of effective vaccines. However, new insight into the factors associated withERDand breakthroughs in understanding the structure of the fusion antigen (F)glycoproteinhas increased optimism that vaccine development is possible. This has led to the investment of time and resources by industry, regulatory agencies, governments and non-profit organizations to develop the necessary infrastructure to make advanced clinical development of RSV vaccine candidates a reality.

Section excerpt

Epidemiology

Respiratory syncytial virus (RSV) is the most common cause of hospitalization in children under 5 years of age [1]. In developing countries, RSV also causes significant mortality in children under 1 year of age [2]. All children are infected by the age of 3, and people are infected many times during their lifetime [3]. In otherwise healthy children over 5 years of age and adults, RSV usually causes an upper respiratory tract syndrome, sometimes complicated by sinusitis and otitis media [4], [5]. IN

Pathology

RSV infects the ciliated epithelium of the upper and lower respiratory tract. The bronchiolar epithelium is particularly susceptible to infection, and type I pneumocytes in the alveoli are also frequently infected, resulting in a high incidence (~20%) of children with hypoxemia, even in those without other significant symptoms. In children with severe RSV disease, the main feature of the pathology is thought to be airway obstruction. Several autopsy studies of fatal RSV infection show that the small airways can

Purpose of vaccination

The primary clinical goal of RSV vaccine is to prevent severe lower respiratory tract disease in young infants. End points will include prevention of hospitalization or medical management of lower respiratory tract infection (MALRI) in industrialized countries, prevention of mortality and hospitalization or MALRI in developing countries and, if possible, development of a clinical severity index as continuous variable of disease severity. Secondary objectives are: (1) medical prevention;

Virology

RSV is a pneumovirus from the Paramyxoviridae family. It has a single-stranded, negative-sense RNA genome of about 15 kilobases with 10 genes separated by stop/start sequences that encode 11 known proteins. There are two main subtypes of RSV, characterized mainly by variations in the G-glycoprotein [22]. Subtypes tend to alternate in dominance from year to year, but they circulate together and are not exclusive to a single era. Although there is genetic variation between characteristic strains

Surface proteins

There are three proteins in the lipid envelope that are exposed on the surface of the virus, including SH (small hydrophobic), G and F (fusion) [24]. SH is a 64–65 amino acid pentameric ion channel analog of the M2 protein of influenza virus. G is a highly glycosylated integral membrane type II protein 298 amino acids in length with a molecular weight of ca. 90 kDa. Its main characteristics include a high serine/threonine content of close to 30%, resulting in a high level of O-glycosylation and approximately 10%

Correlations of immunity

The basis for frequent re-infections with RSV is not known. The fact that there is relatively little genetic variation suggests that it relies on other mechanisms of immune evasion. One possibility is that induction of immunity at the initial event is ineffective and permanently alters the ability to achieve sustained immunity. Initial exposure often occurs in very young children in the presence of maternal antibodies and in hosts with a tendency toward lower IFN activity. These

Disease caused by vaccines

Vaccine-induced illness syndrome has been associated with immunization of antigen-naive infants with formalin-inactivated whole RSV (FI-RSV) formulated in alum [57] , [58] , [59] , [60] . Although the exact mechanism behind RSV disease (ERD) of FI-RSV is unknown, two major immunological phenomena are associated with this syndrome based on pathological evaluation from initial cases [61], [62] and extensive mouse studies. cotton rats, non-human primates (NHP) and cattle

The vaccine is coming

Since the FI-RSV vaccine episode, approaches with intranasally administered live attenuated or chimeric live vectors have progressed to antigen-naive infants. However, in recent years there has been a large increase in the number and variety of RSV vaccine candidates (Table 1). Live attenuated and chimeric live vectors are not associated with ERD and are generally safe and well tolerated [73]. Some of the more recent designs have shown higher levels of immunogenicity

F structure and implications for vaccine development

Solving the crystal structure of the trimeric F-glycoprotein in its prefusion conformation is a recent discovery that led to RSV vaccine development [29], [86], [87]. Prefusion F (pre-F) surfaces target antibodies from several different competitive binding moieties, five of which are currently published [27], [28]. Only two of these sites exist on the common surface that remains in the F (post-F) trimer after fusion, which is the end product of the bulk

Capital

RSV is a major cause of morbidity and mortality and is a high priority for vaccine development. Despite the fact that humans are frequently reinfected with RSV despite the lack of significant genetic variation and the heritability of FI-RSV ERD, there is optimism that a vaccine solution to prevent severe disease is possible. This is based on new insights into virus morphology, the atomic-level structure of F-glycoprotein in pre- and post-F conformational states, virus mechanisms

Acknowledgments

This project was funded by intramural fundingCenter for Vaccine Research, National Institute of Allergy and Infectious Diseases, NIH. The author is the named inventor of patents for RSV vaccines involving F subunit proteins and vector gene delivery of F. I thank Jason Gallo and Kaitlyn Morabito for thoughtful editorial comments on the manuscript.

Report(92)

  • R.Lausanneet al.

    Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010.

    Lanceta

    (2012)

  • T.G.Boyceet al.

    Rates of hospitalization for respiratory syncytial virus infection among children on Medicaid

    J Pediatr

    (2000)

  • R.T.Steinet al.

    Respiratory syncytial virus in early life and risk of wheezing and allergy at age 13

    Lanceta

    (1999)

  • J.E.Johnsonet al.

    Histopathology of fatal untreated human respiratory syncytial virus infection

    Moderni Pathol: Off J US Can Acad Pathol Inc

    (2007)

    (Video) Respiratory Syncytial Virus (RSV) - in 2 mins!

  • K.Modjarradet al.

    World Health Organization hearing on the report on respiratory syncytial virus vaccine development from the World Health Organization meeting on 23-24 March 2015.

    Vaccine

    (2016)

  • J.S.McLellan

    Neutralizing epitopes on the respiratory syncytial virus fusion glycoprotein

    Curr Opin Virol

    (2015)

  • W.P.Glezenet al.

    Risk of respiratory syncytial virus infection for infants from low-income families in relation to age, sex, ethnicity and maternal antibody level

    J Pediatr

    (1981)

  • K.L.O'Brienet al.

    Efficacy of motavizumab in preventing respiratory syncytial virus disease in healthy Native American infants: a randomized, double-blind, placebo-controlled phase 3 study

    Lancet Infect Dis

    (2015)

  • B.S.Grahamet al.

    New antigens for RSV vaccines

    Curr Opin Immunol

    (2015)

  • M.Connorset al.

    Cotton rats preimmunized with chimeric RSV FG glycoprotein develop enhanced lung pathology when challenged with RSV, a phenomenon not seen after immunization with RSV vaccinia or RSV recombinants

    Vaccine

    (1992)

  • T.R.Johnsonet al.

    A genetic vaccine against respiratory syncytial virus provides protection without worsening the disease

    Mol Ther

    (2014)

  • T.Camping houseet al.

    Efficacy and Safety of Intranasal Virosomal Respiratory Syncytial Virus Monophosphoryl Lipid A Vaccine in Mice and Cotton Rats

    Vaccine

    (2013)

  • C.B.Halet al.

    Burden of respiratory syncytial virus infection in young children

    N Engl J Med

    (2009)

  • W.P.Glezenet al.

    Risk of primary infection and re-infection with respiratory syncytial virus

    I'm J Dis Child

    (1986)

  • C.B.Halet al.

    Respiratory syncytial virus infections in previously healthy adults

    Clin Infect Dis

    (2001)

  • W.J.Halet al.

    Respiratory syncytial virus infection in adults: clinical, virological and serial pulmonary function studies

    Ann Trainee Med

    (1978)

  • MI.Hercet al.

    Respiratory syncytial virus-induced acute lung injury in adult bone marrow transplant patients: clinical approach and literature review

    Medicine (Baltimore)

    (1989)

  • FROM.El Salibiet al.

    Quantitative effects of palivizumab and donor-derived T cells on chronic respiratory syncytial virus infection, lung disease, and fusion glycoprotein amino acid sequences in patients before and after bone marrow transplantation.

    Clin Infect Dis

    (2004)

  • A.R.Falseyet al.

    Respiratory syncytial virus infection in the elderly and high-risk adults

    N Engl J Med

    (2005)

  • C.B.Halet al.

    Clinical and epidemiological features of respiratory syncytial virus

    Curr Top Microbiol Immunol

    (2013)

  • H.Zhouet al.

    Hospital admissions associated with influenza and respiratory syncytial virus in the United States, 1993–2008.

    Clin Infect Dis

    (2012)

  • F D.Martinez

    Respiratory syncytial virus bronchiolitis and the pathogenesis of asthma in children

    Pediatr Infect Dis J

    (2003)

  • M.O.Whiteet al.

    Respiratory syncytial virus and recurrent wheezing in healthy preterm infants

    N Engl J Med

    (2013)

  • andMiyairiet al.

    Human genetic factors and the severity of respiratory syncytial virus disease

    Clin Microbiol Rev

    (2008)

  • EH.Choiet al.

    Human genetics and respiratory syncytial virus disease: current knowledge and future approaches

    Curr Top Microbiol Immunol

    (2013)

    (Video) Respiratory Syncytial Virus (RSV) - Clinical Presentation

  • C.B.Halet al.

    Respiratory syncytial virus infections within families

    N Engl J Med

    (1976)

  • P.K.Munywokiet al.

    Source of respiratory syncytial virus infection in infants: a household cohort study in rural Kenya

    J Infect Dis

    (2014)

  • M.Baguelinet al.

    Estimating the optimal target population for influenza vaccination programs: an evidence synthesis and modeling study

    PLoS Med

    (2013)

  • P.L.Collinset al.

    Respiratory syncytial virus: virology, reverse genetics and disease pathogenesis

    Curr Top Microbiol Immunol

    (2013)

  • T.L.Goweret al.

    RhoA signaling is required for respiratory syncytial virus-induced syncytium formation and nematode morphology

    J Virol

    (2005)

  • J.S.McLellanet al.

    Structure and function of respiratory syncytial virus surface glycoproteins

    Curr Top Microbiol Immunol

    (2013)

  • J.O.Excavationet al.

    Prefusion F-Specific Antibodies Quantify RSV Neutralizing Activity in Human Sera

    Sci Transl Med

    (2015)

  • M.Thinet al.

    Neutralizing antibodies against the hyperactive form of respiratory syncytial virus fusion protein provide unique opportunities for clinical intervention

    Proc Natl Acad Sci USA

    (2012)

  • LADY.Gilmanet al.

    Characterization of a fusion-specific antibody that recognizes a cleavage-dependent quaternary epitope on the RSV fusion glycoprotein

    PLOS Pathog

    (2015)

  • J.S.McLellanet al.

    Structure of the major antigenic site of respiratory syncytial virus fusion glycoprotein in complex with neutralizing antibody 101F

    J Virol

    (2010)

  • J.S.McLellanet al.

    Structural basis for respiratory syncytial virus neutralization by motavizumab

    Nat Struct Mol Biol

    (2010)

  • J.S.McLellanet al.

    Structure of a ternary fused RSV glycoprotein bound to perfusion-specific neutralizing antibody

    Science

    (2013)

  • SI.Garcia-Barrenoet al.

    Marked differences in the antigenic structure of human respiratory syncytial virus F and G glycoproteins

    J Virol

    (1989)

  • J.A.Beeleret al.

    Neutralization of respiratory syncytial virus F-glycoprotein epitopes: effect of mutation on fusion function

    J Virol

    (1989)

  • andMartinezet al.

    Antigenic structure of human respiratory syncytial virus G glycoprotein and the importance of hypermutation events in the generation of antigenic variants

    J Gen Virol

    (1997)

  • H.Plotnicky-Gilquinet al.

    Identification of multiple protective epitopes (prototopes) in the central conserved region of the prototypical human respiratory syncytial virus G protein

    J Virol

    (1999)

  • E.J.Collaret al.

    Potent high-affinity antibodies for the treatment and prophylaxis of respiratory syncytial virus derived from B cells of infected patients

    J Immunol

    (2009)

  • S.M.Johnsonet al.

    Respiratory syncytial virus uses CX3CR1 as a receptor in primary cultures of human airway epithelium

    PLOS Pathog

    (2015)

  • SI.Shipset al.

    Protection and mechanism of action of a novel human respiratory syncytial virus vaccine candidate based on the extracellular domain of a small hydrophobic protein

    EMBO Mol Med

    (2014)

  • G.A.Princet al.

    Quantitative aspects of passive immunity to respiratory syncytial virus infection in cotton rat pups

    J Virol

    (1985)

  • J.R.A big houseet al.

    Respiratory syncytial virus (RSV) infection in preterm infants and protective effects of RSV immunoglobulin (RSVIG). Respiratory syncytial virus immunoglobulin study group

    Pediatrics

    (1995)

  • Reported by (67)

    • Antibody-dependent enhancement (ADE) and the role of the complement system in disease pathogenesis

      2022, Molecular immunology

      Antibody-dependent enhancement (ADE) is associated with severe disease outcomes in several viral infections, including respiratory infections.In vitroand in vivo studies have shown that the antibody response to SARS-CoV and MERS-CoV can exacerbate infection through ADE. Recently, in SARS CoV-2, in vitro studies and structural analyzes show risk of disease severity through ADEs. This phenomenon is partly attributed to non-neutralizing antibodies or antibodies at sub-neutralizing levels. These antibodies lead to the deposition of antigen-antibody complexes and the spread of a chronic inflammatory process that destroys the affected tissue. In addition, antigen-antibody complexes can increase viral internalization into cells via the Fc-gamma receptor (FcγR) and lead to further viral replication. Thus, ADE occurs through two mechanisms. 1. Antibody-mediated replication and 2. Enhanced immune activation. Antibody-mediated effector functions are primarily driven by complement activation, and the first complement in the cascade is complement 1q (C1q), which binds to the virus-antibody complex. Lack of circulating plasma C1q levels, an independent predictor of mortality in high-risk patients including diabetes, has been reported to be associated with severe viral infections. Complement-mediated ADE occurs in many viral infections such as dengue fever, West Nile virus, measles, RSV, human immunodeficiency virus (HIV), and Ebola virus. This review examines ADEs in viral infections and in vitro evidence for ADEs in coronaviruses. We describe the mechanisms of ADE and highlight the role of complement, particularly C1q, in increased disease outcome.

    • Induction of Th1 and Th2 in protection against SARS-CoV-2 by mucosal delivery of an adenovirus vaccine expressing a modified spike protein

      2022, Vaccine

      Excerpt from the quote:

      In summary, mucosal administration of AdCoV2 vaccine has the advantages of expressing full protection against SARS-CoV-2. Antibody-dependent enhancement (ADE) has been observed in coronaviruses, including SARS, MERS [36] and other human respiratory viral infections such as RSV [37] and measles [38,39]. Prototype SARS-CoV vaccines that induce a Th2-type immune response carry a risk of ADEs in animal models [40,41].

      A series of recombinant full-length or membrane-truncated human adenoviruses expressing SARS-CoV-2 (S) spike protein (AdCoV2-S or AdCoV2-SdTM) were tested for efficacy against SARS-CoV-2over himintranasal (i.n.) or subcutaneous (s.c.) immunization in a rodent model. Mucosal administration of adenovirus (Ad) vaccines could induce anti-SARS-CoV-2 IgG and IgA in serum and mucosa as indicated by vaginal lavage (vw) and bronchoalveolar lavage fluid (BALF). AdCoV2-S s.c. induced anti-SARS-CoV-2 serum IgG but not IgA in vw and BALF. Use of AdCoV2-S i.n. was able to induce higher levels of anti-SARS-CoV-2 binding and neutralizing antibodies than s.c. Injection. AdCoV2-SdTM i.n. elicits a lower antibody response than AdCoV2-S i.n. Induced anti-S antibody response by AdCoV2-S via i.n. or s.c. are unaffected by pre-existing anti-Ad serum antibodies. Novelly, S-specific IgG1 representing a Th2-mediated humoral response was induced predominantly in sera immunized with Ad i.n., in contrast to more IgG2a representing a Th1-mediated cellular response found in sera immunized with Ad s.c. Activation of S-specific IFN-ɣ and IL-4 in Th1 and Th2 cells in the spleen was observed in AdCoV2-S i.n. and s.c. group, which indicates activation of Th1 and Th2 immunity. AdCoV2-S and AdCoV2-SdTM significantly prevented weight loss and reduced pulmonary viral load in hamsters. Pneumonia reduction was observed with AdCoV-S via i.n. or s.c.-immunized hamsters after SARS-CoV-2 infection. It was associated with Th1 cytokine, but no inflammatory cytokine secretions were found in AdCoV-S i.n. -immunized BALF. These results demonstrate that intranasal administration of the AdCoV2-S vaccine is safe and potent for the prevention of SARS-CoV-2 infections.

    • Viral vaccines: proteins prefer prolines

      2021, Cell Host and Microbe

      Excerpt from the quote:

      The proposed mechanism involves non-NAb induction, resulting in immune complex deposition and complement activation (Browne et al., 2020; Graham, 2020). Therefore, vaccines against RSV and, by extension, others based on class I fusion proteins are best based on trimers that are in the prefusion conformation (Graham, 2016; Graham et al., 2019). But therein lies the problem: the metastable nature of the ternary primer means that it is very difficult to make into a vaccine, especially when expressed as a recombinant protein.

      Most viral vaccines rely on the induction of neutralizing antibodies (NAbs) against viral envelope or spike glycoproteins. Many viral surface proteins exist as trimers that transition from a profusion state when NAb core epitopes are exposed to a post-fusion form in which virus-cell fusion is no longer possible. For optimal vaccine performance, these viral proteins are often engineered to improve the stability and presentation of these NAb epitopes. The method involves the structure-directed introduction of proline residues at key positions that maintain the trimer in the prefusion conformation. We examine how this technique emerged during the development of the HIV-1 Env vaccine and its subsequent broader application to other viral vaccines, including SARS-CoV-2.

    See all cited articles in Scopus
    (Video) Respiratory Syncytial Virus (RSV) Infection: The Causes and Diagnosis | Merck Manual Quick Facts

    Featured Articles (6)

    • Research article

      New avenues in the treatment of human epidermal growth factor receptor 2-negative breast cancer

      New Horizons in Translational Medicine, Volume 2, Number 2, 2015, pp. 27-38

      Breast cancer remains the leading cause of new cancer cases in women and is responsible for the largest number of cancer-related deaths among women worldwide. The goals of breast cancer treatment are to maintain or improve quality of life, prolong survival, and increase disease-free progression. Most cases of breast cancer are estrogen receptor (ER) positive and human epidermal growth factor receptor-2 (HER-2) negative, and current treatment guidelines recommend multiple lines of endocrine therapy followed by chemotherapy in patients with locally recurrent or metastatic disease. Resistance to current therapies increases the need for new treatment options. Translational research and preclinical data have provided insights to identify new signaling pathways for new drugs, and a growing number of biologically targeted agents are currently being developed to identify new therapies. An alternative approach to improving patient benefit is to increase the efficacy and safety of existing agents by changing their administration or pharmacokinetics (ie, adding albumin to paclitaxel), as well as to identify new combination therapies. An interesting combination therapy is the addition of a 130 nm albumin-bound paclitaxel formulation (capture-paclitaxel) for currently approved therapies or targeted drugs in development. This review focuses on a number of key agents being investigated for the treatment of HER-2-negative breast cancer and the use of these agents as combination therapy to achieve sustained disease control.

      • Next to the bed

      • ○ New treatment options are needed for breast cancer patients with HER-2 negative disease and hormone receptor positive or negative disease who develop resistance to current therapies. Newer insights into molecular pathways may soon expand treatment options for all patients with HER-2 negative breast cancer.

      • Bench

      • ○ Several rationally designed combinations of biologically targeted agents and next-generation chemotherapy agents are currently being investigated to prolong disease control and overcome treatment resistance in patients with HER-2-negative breast cancer.

      (Video) Respiratory Syncytial Virus (RSV) - Pathophysiology
    • Research article

      Differential regulation of polysaccharide-specific antibody responses to isolated polysaccharides, conjugate vaccines, and intact gram-positive versus gram-negative extracellular bacteria

      Cjepivo, bind 34, number 30, 2016, pp. 3542-3548

      Bacterial polysaccharides are important virulence agents and are key targets in a number of approved antibacterial vaccines. Their main characteristics are high molecular weight and the expression of repeated antigenic epitopes that mediate multivalent cross-linking of B cell receptors. Furthermore, since most of these antigens cannot bind to MHC-II, they fail to recruit CD4+ T-cell help and are called T-cell-independent antigens. In this review, I will discuss a number of studies from my laboratory that have highlighted the importance of understanding polysaccharide-specific antibody responses in the context in which PS is expressed (ie, isolated as a component of conjugate vaccines and naturally expressed by intact bacteria). . We show that polysaccharide-mediated multivalent cross-linking of B cell receptors uniquely determines the qualitative response of B cells to subsequent stimuli, but is insufficient by itself to induce antibody secretion or class switching. For these latter events to occur, other signals must act in concert with the primary signals emanating from B cell receptors. Coexpression of polysaccharides and proteins in intact bacteria promotes the recruitment of helper CD4+ T cells to a PS-associated IgG response, unlike isolated polysaccharides. In addition, the particulate nature of extracellular bacteria imparts properties to the polysaccharide-specific IgG response that make it immunologically distinct from soluble conjugate vaccines. Finally, fundamental biochemical and/or structural differences distinguishing Gram-positive and Gram-negative bacteria appear to play a critical role in the differential regulation of polysaccharide-related IgG responses in these groups of pathogens. These studies have several implications for the understanding and future design of polysaccharide-based vaccines.

    • Research article

      Introduction to the selected research issue presented at the 2015 National Institute of Infectious Diseases Annual Vaccine Research Conference.

      Cjepivo, bind 34, number 30, 2016, pp. 3522-3524

    • Research article

      Mucosal vaccines against respiratory syncytial virus

      Current Opinion in Virology, svezak 6, 2014., str. 78-84

      Respiratory syncytial virus (RSV) is the leading cause of severe respiratory disease in infants, young children, the immunocompromised, and the elderly worldwide. Natural RSV infection in young children does not induce long-term immunity, and individuals remain susceptible to recurrent RSV infections throughout life. Because RSV infection is confined to the respiratory tract, an RSV vaccine should induce upper and lower airway mucosal immunity to most effectively prevent reinfection with RSV. Although there is no safe and effective vaccine against RSV, significant progress has been made recently in basic RSV vaccine research and development. This review will discuss recent progress in identifying a novel neutralizing antigenic site in the RSV fusion protein (F), understanding the importance of mucosal immune responses in RSV infection, and developing new mucosal vaccination strategies.

    • Research article

      Development and clinical use of novel antibodies for the prevention and treatment of respiratory syncytial virus infections

      Cjepivo, Volume 35, Number 3, 2017, pp. 496-502

      Respiratory syncytial virus (RSV) remains a major cause of morbidity and mortality in infants and young children, immunocompromised patients, and the elderly. Despite the high disease burden, an effective and safe vaccine is lacking, although several candidates are currently under development. Current therapy for RSV infection remains largely supportive, and RSV-specific prophylaxis options are limited to palivizumab. In recent years, new therapeutic options have emerged, including nanosomes, polyclonal, and monoclonal antibodies, and several products are in preclinical and phase I, -II, or -III clinical trials. An important target for antiviral drug development is the surface fusion glycoprotein (F), which is essential for viral infectivity and pathogenesis. Solving the structures of two conformations of the RSV F protein, the prefusion and postfusion forms, revolutionized RSV research. It is now known that F diffusion is far superior in inducing neutralizing antibodies. In this section, we will review the stages of development and the availability of different antibodies directed against RSV for the prevention and also for the treatment of acute RSV infections. Some of these newer RSV drugs have demonstrated increased potency, are being explored through alternative routes of administration, have improved pharmacokinetic profiles with extended half-lives, and may reduce design and manufacturing costs. Management strategies will require targeting not only high-risk populations (including adults or immunocompromised patients) but also previously healthy children, who in fact represent the majority of children hospitalized with RSV infection. Longitudinal follow-up of treated patients is necessary to determine the effect of these strategies on acute disease, as well as their potential long-term benefit on lung morbidity.

    • Research article

      Nebulized ALX-0171 for respiratory syncytial virus lower respiratory tract infection in hospitalized children: a double-blind, randomized, placebo-controlled phase 2b trial

      The Lancet Respiratory Medicine, bind 9, nummer 1, 2021, s. 21-32

      Respiratory syncytial virus (RSV) is the most common cause of severe lower respiratory tract infections with a high global health burden. There are no effective treatments. ALX-0171 is a novel trivalent nanomaterial with antiviral properties against RSV. Our objective was to evaluate the safety and antiviral activity of nebulized ALX-0171 in children hospitalized with lower respiratory tract RSV infection.

      This double-blind, randomized, placebo-controlled phase 2b trial was conducted in 50 pediatric hospital wards in 16 countries. Previously healthy children aged 28 days to less than 24 months hospitalized with acute severe RSV lower respiratory tract infection were randomized into three consecutive safety cohorts (3:1) to receive nebulized ALX-0171 (cohort 1 received 3 mg/kg , cohort 2 received 6 mg/kg and cohort 3 received 9 mg/kg) or placebo once daily for 3 days using online randomization in a sequential safety arm (first part of 12, then four ). In the parallel part of the study, participants (cohort 4) were randomized (parallel 1:1:1:1) to receive nebulized ALX-0171 3 mg/kg, 6 mg/kg, 9 mg/kg or placebo ( block of eight limited by randomization). Study drug coverage was achieved with two consecutive nebulizations (each with either ALX-0171 or placebo) depending on assigned treatment group. The primary outcome was to assess the time for RSV viral load to fall below a measurable threshold, as measured by the mid-orbital nasal swab plaque assay. Safety, clinical efficacy, pharmacokinetics, viral load by RT-qPCR and immunogenicity were secondary outcomes. The analysis, including the primary outcome, was done on a modified intention-to-treat basis (participants who received at least one dose of the study drug as prescribed), and safety was assessed in all children who received at least one dose of the study drug as treated. This trial is registered in EudraCT, 2016-001651-49.

      Between January 10, 2017 and April 26, 2018, 175 children (median age 4.8 months [IQR 2.0-10.8]) received at least one dose of study drug (45 received 3 mg/kg ALX -a -0171, 43 received 6 mg/kg ALX-0171, 45 received 9 mg/kg ALX-0171, and 42 received placebo; modified intent-to-treat population) starting a mean of 3.3 days (SD 1.1) from symptom onset . The median time for viral load to fall below the measurable limit in the plaque assay was significantly faster for the 3 mg/kg group (median 14.2 hours [IQR 5.0–28.0]), the 6 mg/kg (5·1 h ) [4.7–28.5]), and the 9 mg/kg group (5.1 h [4.6–5.9]) than the placebo group (46 ,1 h [25,2–116; 7 ) ]; hazard ratio [HR] for all ALX-0171 groupscompare toplacebo 2.6 [1.7–3.9]; p<0.0001). The median time for viral load to fall below the limit of quantification by RT-qPCR was 95.9 hours (IQR 26.7 to not estimable) for the placebo group (n=35) versus 49.4 hours (25, 1 to 351.4) for all ALX-0171 groups (n=118). Clinical outcomes were not improved with ALX-0171 compared with placebo, with no difference in time to clinical response (oxygen saturation >92% for 4 hours on room air and adequate oral nutrition) in the ALX-0171 and placebo groups drug (median 43·8 hours [IQR 21·7–68.5]compare to47.9 hours [22.5-76.4]; HR 1.1 [95% CI 0.8–1.6]) or change in total weight score from baseline to 5 h post-dose on day 2 (–4 [IQR –6 to –2]compare to−4 [–6 to −1]; difference in least squares mean -0.45 [95% CI -1.39 to 0.49]). Day 2 serum concentrations of ALX-0171 exceeded the concentration estimated to provide complete neutralization of RSV in the lung at the 6 mg/kg and 9 mg/kg doses. Treatment-emergent anti-drug antibodies were detected on day 14 in 46 (34%) of 135 patients receiving ALX-0171 and ten (26%) of 39 patients receiving placebo. Serious adverse events were reported in five (13%) of 40 children in the placebo group and ten (7%) of 135 children in all ALX-0171 groups, leading to study drug discontinuation in three children (two in 3 mg/kg group and one in the 6 mg/kg group). 13 of 15 serious adverse events (three of four in the 3 mg/kg group, two of three in the 6 mg/kg group, three of three in the 9 mg/kg group, and five of five in the placebo) for worsening respiratory status and none were considered to be related to the study drug.

      Antiviral drugs against RSV may not be able to improve the clinical course when RSV infection of the lower respiratory tract is established. Future studies of RSV antivirals should focus on early intervention and more accurate measurement of objective outcomes before significant lower respiratory tract inflammation occurs.

      Ablynx, a Sanofi company.

      (Video) Respiratory Syncytial Virus (RSV): Not Just a Little Kids’ Virus, Explained in 60 Seconds

    See the full text

    Published by Elsevier Ltd.

    FAQs

    Has there ever been a vaccine for RSV? ›

    Today, the U.S. Food and Drug Administration approved Arexvy, the first respiratory syncytial virus (RSV) vaccine approved for use in the United States. Arexvy is approved for the prevention of lower respiratory tract disease caused by RSV in individuals 60 years of age and older.

    How long has the RSV vaccine been around? ›

    RSV vaccines

    RSV vaccine development began in the 1960s with an unsuccessful formalin-inactivated RSV (FI-RSV) vaccine that induced a severe – and in two cases lethal – lung inflammatory response during the first natural RSV infection after vaccination of RSV-naive infants.

    How long does the RSV vaccine last? ›

    So far, GSK found that protection from a single vaccination protects older people through an entire respiratory disease season, and potentially up to a year.

    What is the progress of the RSV vaccine? ›

    In March 2022 AstraZeneca and Sanofi announced that their long-acting antibody, nirsevimab, is 75 percent effective against cases of RSV that require medical care in infants younger than one year old with no history of RSV—and the protection lasts five months.

    Why hasn t there been a vaccine for RSV? ›

    Instead of protecting against RSV, the experimental vaccine made the children more likely to develop more severe illnesses if they got infected. Many children in the trial were hospitalized, and two young children died. “That was a disaster,” Levy said, “and it set the field back by 20 to 30 years.”

    Why did the RSV vaccine fail? ›

    Most notably, the adjuvants strongly activated type 1 T-helper (Th1) responses, which are essential in defending against viruses. These responses have been hard to stimulate in newborns. In fact, a major reason the earlier RSV vaccine failed was that it did not produce a Th1 response.

    Has the RSV vaccine trial stopped? ›

    GlaxoSmithKline (GSK) last year stopped a late-stage clinical trial of its RSV vaccine in pregnant people over an elevated risk of premature birth and associated neonatal deaths in the babies born prematurely. In the GSK trial, premature birth was 38% more likely in the vaccinated pregnancies than in the placebo group.

    Is Pfizer making a vaccine for RSV? ›

    In February 2023, it was announced that the European Medicines Agency (EMA) accepted for review Pfizer's Marketing Authorization Application (MAA) under accelerated assessmentfor RSVpreF, as submitted for both older adults and maternal immunization to help protect infants against RSV.

    Is RSV vaccine a live vaccine? ›

    Live-attenuated respiratory syncytial virus (RSV) vaccines offer several advantages for immunization of infants and young children: (1) they do not cause vaccine-associated enhanced RSV disease; (2) they broadly stimulate innate, humoral, and cellular immunity, both systemically and locally in the respiratory tract; (3 ...

    Do adults need RSV vaccine? ›

    FDA approves world's first RSV vaccine, a shot for adults ages 60 and up. GSK's single-dose shot lowered the risk of severe illness by 94% in older adults. The Food and Drug Administration on Wednesday approved the world's first RSV vaccine: a shot for adults ages 60 and up, made by pharmaceutical giant GSK.

    Should seniors get RSV vaccine? ›

    The panel recommended that the agency approve both. The GSK vaccine was nearly 83 percent effective in preventing lower respiratory tract illness in adults 60 and older in a study of about 25,000 patients, according to data published in The New England Journal of Medicine.

    Is RSV vaccine safe? ›

    A vaccine against respiratory syncytial virus (RSV) was effective at preventing RSV-associated illness among older adults and the children of vaccinated mothers, based on interim results from 2 phase 3 trials.

    Is RSV vaccine FDA approved? ›

    The US Food and Drug Administration on Wednesday approved Pfizer's RSV vaccine for older adults, the second such shot approved for the common virus. Earlier in May, the agency approved the world's first RSV vaccine for older adults, made by GSK.

    What are the challenges with RSV vaccine? ›

    However, vaccine development for RSV faces several challenges unique to RSV: first is the young age of infection; second is the multiple mechanisms RSV uses to evade innate immunity; third is the lack of durable protective immunity induced by natural infection; fourth, a legacy of vaccine-enhanced disease dating back ...

    Do babies get shots to prevent RSV? ›

    There is no vaccine yet to prevent RSV infection, but scientists are working hard to develop one. And there is a medicine that can help protect some babies at high risk for severe RSV disease.

    When is RSV season over 2023? ›

    On March 1, 2023, the Health and Human Services Commission (HHSC) will end Synagis prophylaxis therapy for the season's respiratory syncytial virus (RSV) in all regions.

    Is RSV a Covid? ›

    What is the difference between the flu, COVID-19, and RSV? The flu, COVID-19, and respiratory syncytial virus (RSV) are all highly contagious respiratory infections caused by viruses: The flu by influenza virus, COVID-19 by SARS-CoV-2 virus, and RSV by respiratory syncytial virus.

    Can you get RSV twice? ›

    Infants and older adults may develop severe infections from RSV, such as pneumonia or bronchiolitis. Most kids get an RSV infection by age 2. However, you can get an RSV infection at any age and more than once in your lifetime.

    Why is RSV called syncytial? ›

    Its name is derived from the large cells known as syncytia that form when infected cells fuse. RSV is the single most common cause of respiratory hospitalization in infants, and reinfection remains common in later life: it is a notable pathogen in all age groups.

    How effective is the RSV vaccine? ›

    In an RSV challenge study involving healthy persons who were 18 to 50 years of age, the vaccine efficacy was 87% (95% confidence interval [CI], 54 to 96) against symptomatic RSV infection confirmed by any detectable viral RNA on at least 2 consecutive days.

    How long has SYNAGIS been out? ›

    For nearly 20 years SYNAGIS has been prescribed to high-risk infants and children to help reduce the risk of RSV-related hospitalizations.

    Why are more adults getting RSV? ›

    Older adults are at greater risk than young adults for serious complications from RSV because our immune systems weaken when we are older. Wash your hands often with soap and water for at least 20 seconds.

    Is RSV and whooping cough the same? ›

    Respiratory syncytial virus, or RSV, and pertussis, commonly called whooping cough, are not likely to cause serious health issues for otherwise healthy adults, but it's still very important for everyone to take precautions against both.

    Who is most at risk for RSV? ›

    Risk factors

    People at increased risk of severe or sometimes life-threatening RSV infections include: Infants, especially premature infants or babies who are 6 months or younger. Children who have heart disease that's present from birth (congenital heart disease) or chronic lung disease.

    Who should get RSV vaccine? ›

    Q: Who should get the RSV vaccine? A: Anyone 65 and older with any kind of lung disease, heart disease or other underlying conditions that may cause them to have problems when they get an infection should consider getting the RSV vaccine.

    How much does RSV vaccine cost? ›

    Outcomes Measure Incremental cost (2010 US dollars) per hospitalization for RSV infection avoided. Results The mean cost of palivizumab per dose ranged from $1661 for infants younger than 6 months of age to $2584 for children in their second year of life.

    How much is the SYNAGIS shot? ›

    The cost for Synagis intramuscular solution (100 mg/mL) is around $1,844 for a supply of 0.5 milliliters, depending on the pharmacy you visit. Quoted prices are for cash-paying customers and are not valid with insurance plans.
    ...
    Intramuscular Solution.
    QuantityPer unitPrice
    1 milliliter$3,473.75$3,473.75
    1 more row

    What does RSV look like in adults? ›

    Usually, as an adult, when you become ill with RSV you have mild cold-like symptoms such as a runny nose, sore throat, cough and a headache. But sometimes, and for some people, you can become so ill you need to be hospitalized. And each year in the United States thousands of older adults die of complications from RSV.

    Is there a shot to protect babies from RSV? ›

    In Pfizer's international study of nearly 7,400 pregnant women, maternal vaccination proved 82 percent effective at preventing severe RSV during babies' most vulnerable first three months of life. At age 6 months, it still was proving 69 percent protective against severe illness.

    Is there a vaccine to prevent RSV in babies? ›

    A drug called palivizumab (pah-lih-VIH-zu-mahb) is available to prevent severe RSV illness in certain infants and children who are at high risk for severe disease. This could include, for example, infants born prematurely or with congenital (present from birth) heart disease or chronic lung disease.

    What did RSV used to be called? ›

    RSV was first discovered in 1956 when researchers isolated a virus from a population of chimpanzees with respiratory illness. They named the virus CCA (Chimpanzee Coryza Agent).

    Who qualifies for RSV vaccine? ›

    Infants, especially those 6 months and younger. Children younger than 2 years old with chronic lung disease or congenital heart disease. Children with suppressed immune systems. Children who have neuromuscular disorders, including those who have difficulty swallowing or clearing mucus secretions.

    How much do SYNAGIS shots cost? ›

    The cost for Synagis intramuscular solution (100 mg/mL) is around $1,844 for a supply of 0.5 milliliters, depending on the pharmacy you visit. Quoted prices are for cash-paying customers and are not valid with insurance plans.
    ...
    Intramuscular Solution.
    QuantityPer unitPrice
    1 milliliter$3,473.75$3,473.75
    1 more row

    Is the RSV vaccine safe? ›

    A committee of advisers to the U.S. Food and Drug Administration (FDA) yesterday voted unanimously that a vaccine from Pfizer, given as an injection during pregnancy, is efficacious at protecting infants from severe respiratory syncytial virus (RSV) disease during the first 6 months of life.

    Is RSV caused by Covid? ›

    The flu, COVID-19, and respiratory syncytial virus (RSV) are all highly contagious respiratory infections caused by viruses: The flu by influenza virus, COVID-19 by SARS-CoV-2 virus, and RSV by respiratory syncytial virus. It is possible for a person to be infected with multiple viruses at the same time.

    How contagious is RSV? ›

    RSV is very contagious. It can spread through the droplets released into the air when an infected person coughs or sneezes, or if you touch a contaminated surface, such as counters or doorknobs (where it can live for hours). It can also spread through direct contact (kissing the face of a child with RSV, for example).

    Why are so many kids getting RSV? ›

    Many say the uptick in cases is likely because people weren't exposed much in the past two years with masking and social distancing during the COVID-19 pandemic. Here's some information for parents to help protect children from RSV and to know the first signs of severe infection.

    Why do adults not get RSV? ›

    Yes, adults can get RSV. In fact, you've probably had RSV several times in your life. As you get older and your immune system develops more antibodies, you're able to fight off RSV more efficiently. So you may have only developed a mild cold with your RSV infection.

    Why is there so much RSV right now? ›

    If you are suddenly hearing a lot about respiratory syncytial virus (RSV), it is because the virus has come earlier and hit harder than in most years, and is currently surging and straining children's hospitals across the US

    Videos

    1. Jan 12, 2022 ACIP Meeting - Respiratory Syncytial Virus Vaccines & Pneumococcal vaccines
    (Centers for Disease Control and Prevention (CDC))
    2. Respiratory Syncytial Virus (RSV)
    (Cleveland Clinic)
    3. Development of Respiratory Syncytial Virus and Universal Influenza Virus Vaccines
    (Labroots)
    4. WVU Medicine Tuesday Talks: Respiratory Syncytial Virus - RSV
    (WVU Medicine)
    5. Respiratory Syncytial Virus (RSV): Not Just a Little Kids’ Virus
    (Alliance for Aging Research)
    6. Oct 20, 2022 ACIP Meeting - Respiratory Syncytial Virus Vaccines
    (Centers for Disease Control and Prevention (CDC))

    References

    Top Articles
    Latest Posts
    Article information

    Author: Carlyn Walter

    Last Updated: 06/08/2023

    Views: 5485

    Rating: 5 / 5 (50 voted)

    Reviews: 81% of readers found this page helpful

    Author information

    Name: Carlyn Walter

    Birthday: 1996-01-03

    Address: Suite 452 40815 Denyse Extensions, Sengermouth, OR 42374

    Phone: +8501809515404

    Job: Manufacturing Technician

    Hobby: Table tennis, Archery, Vacation, Metal detecting, Yo-yoing, Crocheting, Creative writing

    Introduction: My name is Carlyn Walter, I am a lively, glamorous, healthy, clean, powerful, calm, combative person who loves writing and wants to share my knowledge and understanding with you.