Elsevier

Clinical Immunology

Volume 108, Issue 3, September 2003, Pages 241-247
Clinical Immunology

Regular article
Humoral, mucosal, and cellular immune responses to oral Norwalk virus-like particles in volunteers

https://doi.org/10.1016/S1521-6616(03)00120-7Get rights and content

Abstract

Norwalk virus-like particles (VLPs), made from recombinant capsid protein, are a promising vaccine. Thirty-six healthy adult volunteers received 250 μg (n = 10), 500 μg (n = 10), or 2000 μg (n = 10) of orally administered VLP or placebo (n = 6). All vaccinees developed significant rises in IgA anti-VLP antibody-secreting cells. Ninety percent who received 250 μg developed rises in serum anti-VLP IgG; neither the rates of seroconversion nor geometric mean titers increased at the higher doses. About 30–40% of volunteers developed mucosal anti-VLP IgA. Lymphoproliferative responses and IFN-γ production were observed transiently among those who received 250 μg or 500 μg but not 2000 μg of VLP. Studies to increase immunogenicity using a mucosal adjuvant are planned.

Introduction

Noroviruses are enteric viruses of the Caliciviridae family that are the major cause of epidemic gastroenteritis in the United States [1]. Norwalk virus (NV) is one strain within this group. Recent studies using new diagnostic assays developed with recombinant NV (rNV) particles have shown that the epidemiologic significance of NV infections has been previously greatly underestimated. New antibody prevalence studies indicate NV infections occur in young children and adults in developed countries more frequently than previously reported [2], [3], [4]. Norovirus infections are a particular concern in elderly residents of nursing homes [5]. In addition, NV caused illness among U.S. troops in Kuwait and has caused significant debilitating illness during recent military maneuvers in Somalia and Afghanistan, on aircraft carriers, and on pleasure cruise ships [6], [7], [8]. The virus is spread by food and water and from person-to-person contact [9], [10]. The clinical manifestations of NV infections include sudden onset of vomiting and/or diarrhea after a 24- to 48-h incubation period; symptoms typically last 12–24 h [7].

The successful cloning, sequencing, and expression of the major capsid protein of NV in insect cells using baculovirus recombinants led to the discovery that the capsid protein folds spontaneously into virus-like particles (VLPs) that lack nucleic acid [11], [12]. Such rNV particles may be produced in high concentrations (20 mg of purified particles per 2 × 108 infected cells), and these particles have desirable properties for use as a subunit vaccine [23]. Specifically, the particles are (i) stable following lyophilization and when exposed to acid (pH 2.5), (ii) highly immunogenic when injected parenterally into animals, (iii) immunogenic when given orally [13] or intranasally [14] to mice in the absence of adjuvant, and (iv) safe and immunogenic when given orally to antibody-positive volunteers [15]. Recombinant NV particles could be used to immunize against NV and as a carrier to deliver other vaccine antigens.

Norwalk VLPs have previously been investigated as an oral immunogen in volunteers. Seropositive volunteers studied at Baylor College of Medicine were given two oral doses of 100 μg of Norwalk VLPs, 3 weeks apart [15]. Even with this low dosage, 3 (60%) of 5 volunteers developed a rise in serum antibody after the first dose. After 250 μg of VLP with bicarbonate buffer, 15 (100%) of 15 volunteers developed a four-fold rise in ELISA antibody to rNV protein when measured 2 weeks after immunization. The seroconversion rate was encouraging, but the maximal titers achieved did not approach those after experimental infection [15].

The purpose of this study was to determine the safety and immune responses to escalating dosages of Norwalk VLPs given by the oral route in seropositive adult volunteers (the seroprevalence of NV antibody in the United States is at least 90%) in an attempt to increase the serum antibody levels to titers closer to those observed after experimental Norwalk virus infection. The local mucosal antibody responses in the gastrointestinal and genital tracts and the cellular immune responses to VLP were also evaluated.

Section snippets

Volunteers

Volunteers were adults 18–40 years of age recruited from the Baltimore community who gave informed, signed consent. They were counseled and trained so that they understood the rationale for the study as well as the risks, benefits, and procedures involved. To ensure comprehension of the study and to document that informed consent had been elicited, the volunteers had to pass a written examination before participation. Prospective volunteers were carefully screened to ensure that they were in

Safety

In the 3 days after receiving VLP vaccine, no volunteer at any of the three doses experienced fever, diarrhea, or vomiting. Three volunteers who received 250 or 500 μg, but no volunteer who received 2000 μg, reported mild cramps after vaccination. Nausea was reported by one vaccinee (2000 μg) and one placebo recipient; headache and malaise were reported more commonly among placebo recipients.

Immune responses

The ASC responses are shown in Fig. 1. All 30 recipients of VLP vaccine, regardless of dosage, had a

Discussion

It is estimated that every American has one episode of gastroenteritis each year [22], and young children and the elderly are at particularly high risk [5], [23]. The widespread incidence of norovirus infections in different epidemiologic settings and the impact of illness has led to interest in developing a vaccine. Vaccine development has been promoted by the observation that immunity develops after infection, but immunity is short-term and homologous virus-specific [24], [25]. Recombinant NV

Acknowledgements

The investigators are grateful for the invaluable contributions of the staffs of the Adult Clinical Studies Section, the Cellular Immunology and Flow Cytometry Section, and Applied Immunology Section of the Center for Vaccine Development and, in particular, acknowledge the invaluable contribution of the volunteers from our community. This study was supported by the Enteric Pathogens Research Unit, NIAID contract NO1-AI65299.

References (31)

  • Centers for Disease Control and Prevention, Outbreak of acute gastroenteritis associated with Norwalk-like viruses...
  • F.E. Ahmed, Seafood Safety. National Academy Press, 30,...
  • A.Z. Kapikian, M.K. Estes, R.M. Chanock, in Fields Virology, Raven Press, New York, 1994, pp....
  • X. Jiang et al.

    Norwalk virus genome cloning and characterization

    Science

    (1990)
  • B.V. Venkataram Prasad et al.

    Structural studies of recombinant Norwalk capsids

    J. Infect. Dis.

    (2000)
  • Cited by (211)

    • Viral protein-based nanoparticles (part 2): Pharmaceutical applications

      2023, European Journal of Pharmaceutical Sciences
    • Immunity to enteric viruses

      2022, Immunity
      Citation Excerpt :

      Secretory IgA is among the most important indicators of protection against enteric viral infection and can potentially neutralize virus in the mucus layer before it reaches host cells, preventing infection altogether. Indeed, clearance of enteric viruses has been positively correlated to the presence of specific intestinal IgA (Atmar et al., 2011; Blutt et al., 2012; Cheuvart et al., 2014; Lindesmith et al., 2003; Nishio et al., 1992; Ramani et al., 2015; Tacket et al., 2003; Unicomb et al., 1996). In contrast, circulating antibodies have limited access to the gut mucosa and hence are less capable of limiting the initial mucosal phase of enteric infections, though they have been shown to offer protection against the systemic phase of certain enteric pathogens (Faden et al., 1990; Hird and Grassly, 2012; Zeng et al., 2016).

    • Phylogenetic and biological characterizations of a GI.3 norovirus

      2020, Infection, Genetics and Evolution
      Citation Excerpt :

      The study of human NoVs is impeded by the lack of simple and efficient culture methods and animal models (Jones et al., 2015). Current knowledge about the immune response to human NoVs has mainly been obtained from experimental human infections or vaccinations based on purified virions or virus-like particles (VLPs) (Graham et al., 1994; Lindesmith et al., 2015; Tacket et al., 2003). NoV VLPs, which are similar in morphology and antigenicity with wild virions, can be produced by in vitro expression of VP1 proteins using different expression systems (Huo et al., 2018; Jiang et al., 1992).

    View all citing articles on Scopus
    View full text