Elsevier

Vaccine

Volume 26, Issues 27–28, 25 June 2008, Pages 3480-3488
Vaccine

Sendai virus recombinant vaccine expressing hPIV-3 HN or F elicits protective immunity and combines with a second recombinant to prevent hPIV-1, hPIV-3 and RSV infections

https://doi.org/10.1016/j.vaccine.2008.04.022Get rights and content

Abstract

The human parainfluenza viruses (hPIVs) and respiratory syncytial virus (RSV) are the leading causes of serious respiratory illness in the human pediatric population. Despite decades of research, there are currently no licensed vaccines for either the hPIV or RSV pathogens. Here we describe the testing of hPIV-3 and RSV candidate vaccines using Sendai virus (SeV, murine PIV-1) as a vector. SeV was selected as the vaccine backbone, because it has been shown to elicit robust and durable immune activities in animal studies, and has already advanced to human safety trials as a xenogenic vaccine for hPIV-1. Two new SeV-based hPIV-3 vaccine candidates were first generated by inserting either the fusion (F) gene or hemagglutinin-neuraminidase (HN) gene from hPIV-3 into SeV. The resultant rSeV-hPIV3-F and rSeV-hPIV3-HN vaccines expressed their inserted hPIV-3 genes upon infection. The inoculation of either vaccine into cotton rats elicited binding and neutralizing antibody activities, as well as interferon-γ-producing T cells. Vaccination of cotton rats resulted in protection against subsequent challenges with either homologous or heterologous hPIV-3. Furthermore, vaccination of cotton rats with a mixture of rSeV-hPIV3-HN and a previously described recombinant SeV expressing the F protein of RSV resulted in protection against three different challenge viruses: hPIV-3, hPIV-1 and RSV. Results encourage the continued development of the candidate recombinant SeV vaccines to combat serious respiratory infections of children.

Introduction

The human parainfluenza viruses (hPIVs) and respiratory syncytial virus (RSV) are the leading causes of viral pneumonia in infants and children [1]. Among the hPIVs, the hPIV-3 subtype causes the most serious infections. In the United States, hPIV-3 epidemics occur annually during spring and summer months [1], [2]. Approximately 62% of humans are infected with hPIV-3 by age 1, more than 90% by age 2, and almost 100% by age 4 [3], [4].

Clinical observations have indicated that the first hPIV-3 infection is generally most severe. Re-infection with hPIV-3 occurs throughout life, but tends to result in more mild disease and is associated only infrequently with serious lower respiratory tract illness. The more mild disease is likely attributed to the larger airways of infected individuals and to the memory T-cell and B-cell activities elicited by first infections [1]. The production of an effective hPIV-3 vaccine is clearly desired as a means to combat the more serious infections of younger individuals.

Previous efforts to develop hPIV-3 vaccines have included studies of cold-adapted viruses [5], [6], [7] and bovine PIV-3 [8]. Challenges facing the advancement of cold-adapted vaccines have concerned the safety of vaccinated infants and their close contacts. In early studies, the frequency of adverse events and transmission rendered certain vaccine candidates unacceptable. However, one cold-adapted vaccine (HPIV3cp45) has met safety requirements and may continue to advance [9], [10], [11]. The main challenge facing the bovine PIV-3 strategy has been its limited antigenic relation to human PIV-3. The vaccine has appeared to be safe in humans, but has not generated protective immune responses. Researchers hope to remedy this situation by producing vaccines that recombine the hPIV-3 hemagglutinin-neuraminidase (HN) and fusion (F) genes with the bovine PIV-3 backbone [12], [13].

Here, we describe a new strategy for the development of hPIV-3 vaccines: the use of reverse genetics to create Sendai virus (SeV)-based vectors that express the hPIV-3 genes HN and F. SeV (mouse PIV-1) was chosen as the delivery vehicle for these vaccines, because of its ability to prevent hPIV-1 infections in non-human primates [14], [15], its natural host range restriction [16] and its safety profile in current clinical trials [16], [17]. The hPIV-3 HN and F genes were selected as target antigens because each encodes a viral membrane protein with known B-cell and T-cell immunogenicity [18], [19], [20], [21].

In this report, we show that the SeV-based hPIV-3 vaccines not only elicit robust immune responses, but also mediate protection against homologous and heterologous hPIV-3 infections in a cotton rat model. Further, we show that a vaccine formulated by mixing one of these candidate SeV-based hPIV-3 vaccines with a previously described SeV-based RSV vaccine [22], [23] protects cotton rats from challenges with three different respiratory viruses: hPIV-1, hPIV-3 and RSV.

Section snippets

Construct design

Replication-competent recombinant SeVs were rescued using a reverse genetics system, described previously [22], [23], [24], [25]. The full-length cDNA of SeV (Enders strain) was first cloned. To this end, Enders SeV RNA was extracted from purified stock virus and reverse transcription (RT)-PCR was performed. PCR products of each gene were cloned into pTF1 and then cloned into pUC19 to construct the full genome SeV Enders cDNA (pSV(E)). The SeV genome in this clone was straddled by a T7 promoter

Human PIV-3 F and HN proteins are expressed by cells infected with recombinant SeV vaccines

Recombinant SeVs were prepared by the insertion of hPIV-3 F or HN genes between the SeV F and HN genes of the full SeV Enders genome (Fig. 1, panels A–C). The viruses rSeV-hPIV3-F and rSeV-hPIV3-HN were subsequently rescued and sequenced (demonstrating precise maintenance of passenger gene sequences). To examine expression of passenger genes by new viruses, we infected Hep-2 cells with the recombinant SeVs and performed radio-immunoprecipitation experiments. As shown in Fig. 1 (panels D and E),

Discussion

This report describes two new recombinant SeV vaccines that express the hPIV-3 F (rSeV-hPIV3-F) and HN (rSeV-hPIV3-HN) proteins, respectively. We initiated studies by demonstrating PIV-3 protein expression by cells infected with the recombinant SeVs. We then employed a cotton rat model to show that each candidate vaccine elicited neutralizing B-cell and T-cell activities and protected animals against homologous and heterologous hPIV-3 challenges. These hPIV-3 results confirmed and supplemented

Acknowledgements

We thank Dr. Greg Prince (Virion Systems) for providing cotton rat antibody reagents. We thank Robert Sealy and Ruth Ann Scroggs for expert technical assistance. We thank Sharon Naron for critical editorial review. This work was supported by NIH NIAID grant P01 AI054955, NIH NCI grant P30-CA21765, and the American–Lebanese Syrian Associated Charities (ALSAC). We thank Dr. R. Hayden (St. Jude Children's Research Hospital, Memphis, TN) and the American Type Culture Collection (ATCC, Rockville,

References (51)

  • X. Zhan et al.

    Respiratory syncytial virus (RSV) fusion protein expressed by recombinant Sendai virus elicits B-cell and T-cell responses in cotton rats and confers protection against RSV subtypes A and B

    Vaccine

    (2007)
  • M. Sangster et al.

    Distinctive kinetics of the antibody-forming cell response to Sendai virus infection of mice in different anatomical compartments

    Virology

    (1995)
  • B. Voges et al.

    Recombinant Sendai virus induces T cell immunity against respiratory syncytial virus that is protective in the absence of antibodies

    Cell Immunol

    (2007)
  • A.M. Arvin et al.

    New viral vaccines

    Virology

    (2006)
  • R.A. Karron et al.

    Parainfluenza viruses

  • A.P. Durbin et al.

    Progress in the development of respiratory syncytial virus and parainfluenza virus vaccines

    Clin Infect Dis

    (2003)
  • W.P. Glezen et al.

    Parainfluenza virus type 3: seasonality and risk of infection and reinfection in young children

    J Infect Dis

    (1984)
  • M.S. Lee et al.

    Antibody responses to bovine parainfluenza virus type 3 (PIV3) vaccination and human PIV3 infection in young infants

    J Infect Dis

    (2001)
  • R.B. Belshe et al.

    Evaluation of a live attenuated, cold-adapted parainfluenza virus type 3 vaccine in children

    J Clin Micro

    (1992)
  • S.L. Hall et al.

    A cold-adapted mutant of parainfluenza virus type 3 is attenuated and protective in chimpanzees

    J Infect Dis

    (1993)
  • R.A. Karron et al.

    Evaluation of a live attenuated bovine parainfluenza type 3 vaccine in two- to six-month-old infants

    Pediatr Infect Dis J

    (1996)
  • R. Ranjit et al.

    Temperature-sensitive phenotype of the human parainfluenza virus type 3 candidate vaccine strain (cp45) correlates with a defect in the L gene

    J Virol

    (1996)
  • A.A. Haller et al.

    Expression of the surface glycoproteins of human parainfluenza virus type 3 by bovine parainfluenza virus type 3, a novel attenuated virus vaccine vector

    J Virol

    (2000)
  • S. Pennathur et al.

    Evaluation of attenuation, immunogenicity and efficacy of a bovine parainfluenza virus type 3 (PIV-3) vaccine and a recombinant chimeric bovine/human PIV-3 vaccine vector in rhesus monkeys

    J Gen Virol

    (2003)
  • R. Sealy et al.

    Antibody response to influenza infection of mice: different patterns for glycoprotein and nucleocapsid antigens

    Immunology

    (2003)
  • Cited by (45)

    • Evaluation of a novel immunogenic vaccine platform based on a genome replication-deficient Sendai vector

      2013, Vaccine
      Citation Excerpt :

      Thus, novel vaccine approaches could help to achieve optimal protection against infectious diseases. Viral vectors have emerged as potent and defined approaches with immunogenic characteristics [5–9] similar to live attenuated vaccines. However, live attenuated vectors still present similar safety concerns.

    • Role of N-linked glycosylation of the human parainfluenza virus type 3 hemagglutinin-neuraminidase protein

      2013, Virus Research
      Citation Excerpt :

      The human parainfluenza viruses (hPIV), belonging to the Paramyxoviridae family, are a major cause of croup and respiratory tract infections in infants and children. Among hPIV types 1–4, the hPIV-3 subtype is the most frequently detected virus causing most infections (Fry et al., 2006; Moscona, 2005; Reed et al., 1997; Watanabe et al., 2009; Zhan et al., 2008). Approximately 62% of humans are infected with hPIV-3 by age 1, 92% by age 2 and almost 100% by age 4 (Moscona, 2005; Zhan et al., 2008).

    • Sendai virus-based RSV vaccine protects African green monkeys from RSV infection

      2012, Vaccine
      Citation Excerpt :

      Constructs have expressed genes from pathogens such as human PIV-2, PIV-3, and RSV. In cotton rats, these vaccines can be used individually or in combination to protect against respiratory virus challenge [12–16]. When three vaccines were combined, expressing foreign genes from PIV-2, PIV-3 and RSV, four different pathogen infections could be prevented (hPIV-1, hPIV-2, hPIV-3 and RSV).

    View all citing articles on Scopus
    1

    Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, United States.

    2

    Early Development, Novartis Vaccines and Diagnostics, 350 Massachusetts Avenue, Cambridge, MA 02139, United States.

    3

    2645 NE Haskett PI, Mountain Home, ID 83647, United States.

    4

    Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, United States.

    View full text