Elsevier

Current Opinion in Virology

Volume 11, April 2015, Pages 70-75
Current Opinion in Virology

Neutralizing epitopes on the respiratory syncytial virus fusion glycoprotein

https://doi.org/10.1016/j.coviro.2015.03.002Get rights and content

Highlights

  • X-ray crystal structures of prefusion and postfusion RSV F have been determined.

  • Antibodies specific for the prefusion conformation are extremely potent.

  • Select antibodies neutralize both RSV and human metapneumovirus.

  • Structure-based vaccine design has produced promising immunogens.

Respiratory syncytial virus (RSV) is a leading cause of pneumonia and bronchiolitis, but despite decades of research a safe and effective vaccine has remained elusive. The viral fusion glycoprotein (RSV F) plays an obligatory role in the entry process and is the major target of neutralizing antibodies, making it an attractive target for vaccine development. This review will summarize the recently determined structures of RSV F in the prefusion and postfusion conformations and describe the location and properties of neutralizing epitopes on RSV F, including the newly identified prefusion-specific epitopes. The influence of these findings on vaccine development will also be discussed, with a focus on the rational design and optimization of vaccine antigens.

Introduction

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract diseases such as pneumonia and bronchiolitis [1]. It is an extremely infectious paramyxovirus, infecting almost all children within their first two years of life [2]. The very young are most susceptible to severe disease, and in the U.S. the peak age of hospitalization is 1–2 months of age [3, 4]. In a recent analysis of global mortality, it was estimated that RSV is responsible for 6.7% of all deaths for children between 1 month and 1 year of age [5]. RSV infection elicits neutralizing antibodies and an effective T cell response, but immunity to the virus wanes over time, leading to reinfections throughout life [6]. Although disease severity is mild for older children and healthy adults, RSV causes substantial morbidity and mortality in elderly people [7, 8].

An effective RSV vaccine is urgently needed, and more than 40 are currently in development covering a broad range of modalities, including live-attenuated and protein subunit vaccines [9, 10]. Of the three proteins on the virion surface, only the fusion glycoprotein (RSV F) is absolutely required for infection, and it is the main target of neutralizing antibodies in human sera [11••, 12]. RSV F is also the target of palivizumab, a monoclonal antibody that passively protects infants from severe disease [13]. Therefore, the elicitation of potent, neutralizing antibodies against RSV F is a major goal for most vaccine developers [14].

In the last few years, there have been new developments in RSV research that have significantly advanced our understanding of the structure, antigenicity, and immunogenicity of RSV F. These include the elucidation of the X-ray crystal structures of RSV F in the prefusion and postfusion conformations, the isolation of extremely potent neutralizing antibodies that target novel epitopes, and the rational design of vaccine antigens. The goal of this review is to summarize these recent advances and highlight how this knowledge is being used to inform vaccine development.

Section snippets

RSV F structure

RSV F is a class I fusion glycoprotein that, like influenza HA and HIV-1 env, is synthesized as a precursor (F0) that requires proteolytic cleavage for activation [15]. The mature protein contains three copies of two polypeptides (F2 and F1) that are held together by two disulfide bonds. After initially folding into a metastable prefusion conformation, RSV F undergoes a dramatic rearrangement that results in a stable postfusion conformation, leading to fusion of the viral and cellular membranes.

Epitopes shared on prefusion and postfusion F

There are two major neutralizing epitopes, antigenic sites II and IV, found on both the prefusion and postfusion conformations of RSV F (Figure 2). Antibodies against both sites fail to block virus attachment but they neutralize infection by blocking fusion of the viral and cellular membranes [24].

Antibodies that recognize antigenic site II include murine 47F and 1129 [25, 26], with humanization of the latter resulting in the licensed monoclonal antibody palivizumab [27] and its more potent

Epitopes unique to prefusion F

By passing human sera over immobilized postfusion RSV F, Melero and colleagues found that the majority of the neutralizing activity was not retained on the column. This demonstrated not only that prefusion-specific epitopes exist, but also that they are the main target of neutralizing antibodies elicited by natural infection in humans [11••].

Antibodies D25, AM22 and 5C4 were the first antibodies shown to be prefusion-specific. D25 and AM22 were isolated by AIMM Therapeutics from immortalized B

Conclusions

RSV causes substantial morbidity and mortality in infants and the elderly, and an effective vaccine has yet to be developed. Recent advances in our understanding of the structure, function and antigenicity of RSV F, as well as the isolation and characterization of novel RSV F-directed antibodies, have created new opportunities for passive prophylaxis and vaccine development. Antibodies that specifically recognize the prefusion conformation of RSV F are especially potent, and elicitation of such

Conflict of interest

Dr. Jason McLellan is an inventor on several patent applications related to the use of motavizumab-epitope scaffolds, antibody 5C4 and prefusion RSV F.

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

The author would like to thank José Melero and members of the McLellan laboratory for helpful comments on the manuscript. Dr. McLellan was supported in part by NIAID Grant 1R43AI112124.

References (42)

  • C.B. Hall et al.

    Clinical and epidemiologic features of respiratory syncytial virus

    Curr Top Microbiol Immunol

    (2013)
  • W.P. Glezen et al.

    Risk of primary infection and reinfection with respiratory syncytial virus

    Am J Dis Child

    (1986)
  • C.B. Hall

    The burgeoning burden of respiratory syncytial virus among children

    Infect Disord Drug Targets

    (2012)
  • C.B. Hall et al.

    Respiratory syncytial virus-associated hospitalizations among children less than 24 months of age

    Pediatrics

    (2013)
  • S.M. Varga et al.

    The adaptive immune response to respiratory syncytial virus

    Curr Top Microbiol Immunol

    (2013)
  • K. Widmer et al.

    Rates of hospitalizations for respiratory syncytial virus, human metapneumovirus, and influenza virus in older adults

    J Infect Dis

    (2012)
  • E.E. Walsh et al.

    Respiratory syncytial virus infection in adult populations

    Infect Disord Drug Targets

    (2012)
  • R. Rudraraju et al.

    Respiratory syncytial virus: current progress in vaccine development

    Viruses

    (2013)
  • M. Magro et al.

    Neutralizing antibodies against the preactive form of respiratory syncytial virus fusion protein offer unique possibilities for clinical intervention

    Proc Natl Acad Sci U S A

    (2012)
  • P.L. Collins et al.

    Progress in understanding and controlling respiratory syncytial virus: still crazy after all these years

    Virus Res

    (2011)
  • L.J. Anderson et al.

    Strategic priorities for respiratory syncytial virus (RSV) vaccine development

    Vaccine

    (2013)
  • Cited by (88)

    • Vaccines

      2023, Haschek and Rousseaux's Handbook of Toxicologic Pathology: Volume 2: Toxicologic Pathology in Safety Assessment, Fourth Edition
    View all citing articles on Scopus
    View full text