Elsevier

Vaccine

Volume 28, Issue 3, 8 January 2010, Pages 632-649
Vaccine

Review
Preclinical and clinical development of YFV 17D-based chimeric vaccines against dengue, West Nile and Japanese encephalitis viruses

https://doi.org/10.1016/j.vaccine.2009.09.098Get rights and content

Abstract

Dengue viruses (DENV), West Nile virus (WNV) and Japanese encephalitis virus (JEV) are major global health and growing medical problems. While a live-attenuated vaccine exists since decades against the prototype flavivirus, yellow fever virus (YFV), there is an urgent need for vaccines against dengue or West Nile diseases, and for improved vaccines against Japanese encephalitis. Live-attenuated chimeric viruses were constructed by replacing the genes coding for Premembrane (prM) and Envelope (E) proteins from YFV 17D vaccine strain with those of heterologous flaviviruses (ChimeriVax™ technology). This technology has been used to produce vaccine candidates for humans, for construction of a horse vaccine for West Nile fever, and as diagnostic reagents for dengue, Japanese encephalitis, West Nile and St. Louis encephalitis infections. This review focuses on human vaccines and their characterization from the early stages of research through to clinical development. Phenotypic and genetic properties and stability were examined, preclinical evaluation through in vitro or animal models, and clinical testing were carried out. Theoretical environmental concerns linked to the live and genetically modified nature of these vaccines have been carefully addressed. Results of the extensive characterizations are in accordance with the immunogenicity and excellent safety profile of the ChimeriVax™-based vaccine candidates, and support their development towards large-scale efficacy trials and registration.

Introduction

Several members of the Flavivirus genus are serious threats to human and/or animal health. Among them, Dengue virus (DENV), West Nile virus (WNV) and Japanese encephalitis virus (JEV) represent a major and growing medical problem [1].

First, the number of DENV infections in endemic areas has continued to increase over the past two decades, and more than 100 million dengue infections resulting in 24,000 deaths are now estimated annually with children bearing the bulk of the disease burden. There are four serotypes of DENV and infection by each of them can lead to clinical manifestations ranging from self-limiting dengue fever to severe dengue hemorrhagic fever (DHF) and fatal dengue shock syndrome (DSS). Over one hundred countries are affected with over three billion people at risk (for a review see [2]). This increased incidence and extended geographical reach of dengue have made the development of an effective vaccine an international health priority.

WNV is a zoonotic pathogen which first appeared in the Western Hemisphere in 1999 in New York City, before spreading rapidly across North America, causing disease in wild birds, horses, and humans. The virus has also been isolated in the West Indies and neutralizing antibody-positive birds and horses have been identified in Mexico, Jamaica, and the Dominican Republic; West Nile virus also appeared recently in South America, including Brazil and Argentina. The spectrum of disease extends from a mild febrile exanthem to fatal encephalitis; neuroinvasive disease occurs in ∼30% of reported WNV cases. Around 30,000 cases and more than 1000 deaths have been attributed to WNV infection since its introduction [3], [4]. There is no effective drug treatment against this infection. While the introduction of vaccines for horses represents a potent tool to control the veterinary disease, a human vaccine against WNV may represents an important approach to the prevention and control of this emerging disease (for a review see [5]), although recent epidemiological data show a sharp decline in the number WNV cases in the past years.

Finally, JEV is the main cause of viral encephalitis in Asia and thus constitutes a major threat for approximately three billion people living in endemic regions. JEV has also spread to previously unaffected areas of Australia [6], [7]. It causes an estimated 50,000 cases of disease and 10,000 deaths per year, mostly in children [8]. Acute encephalitis occurs in about 1–20 cases/1000 infections, being lethal in 29% of cases. A high proportion of survivors have neurological and psychiatric sequelae. Different vaccines are available: mouse brain-derived inactivated vaccine, and cell-culture derived, inactivated or live vaccines, yet none has been prequalified by WHO at this time (for a review see [9], [10]). The introduction of inactivated JE vaccines in Japan in the mid-1950s lead to a dramatic drop of reported cases but raised concerns regarding adverse events (AEs) following immunization (Acute Disseminated EncephaloMyelitis, ADEM, reported in 1 per 50,000–1,000,000 cases). An inactivated-vaccine produced on primary hamster kidney (PHK) cells has also been used in China from the 1970s to the 1990s, before being replaced by the live-attenuated vaccine SA14-14-2. This vaccine has been successfully used for massive immunization campaigns in China and India, in which it has been given to >100 million children and provides 80–96% protection after a single dose. First attempts made in the 1980s to adapt this virus in a more acceptable cell substrate for vaccine production have lead to a decrease immunogenicity and development of such a vaccine was discontinued [11]. The SA14-14-2 strain has been adapted to growth on Vero for use as a two dose inactivated vaccine (IXIARO®, Intercell, Vienna) which has recently been approved by the US FDA for adult travelers and military personnel.

While a live-attenuated vaccine exists since decades against the prototype flavivirus, i.e. the YFV, there is thus still a need for vaccines against dengue or West Nile diseases, and for improved vaccines against JEV which protect after a single dose.

Several vaccine candidates against these diseases exist or are being developed, including live, vectored and recombinant preparations (for reviews see [5], [12], [13], [14], [15], [16]). Sanofi Pasteur has developed chimeric vaccines using the ChimeriVax technology, originated at St Louis University, where the first YFV 17D-based chimera was constructed in 1999 by Chambers et al for Japanese encephalitis virus [17] and further developed as a vaccine virus by Guirakhoo et al. [18] at Acambis inc., now a part of Sanofi Pasteur. The first YFV 17D/dengue chimera was constructed by Guirakhoo et al in 2000 [19]. These vaccines, and later on other chimeric flaviviruses, were constructed by replacing the genes for YF vaccine (YFV 17D 204) Premembrane (prM) and Envelope (E) proteins, with those of heterologous flaviviruses [17], [18], [19], [20], [21], [22], [23], [24], [25], [26] (see Fig. 1).

Characteristics of different ChimerVax-based vaccines are summarized in Table 1. PrM and E genes were either derived from wild type (wt) viruses without modification (e.g. dengue and veterinary West Nile vaccines), or from empirically derived attenuated vaccine (e.g. JEV strain SA14-14-2 for JEV vaccine) or by introduction of specific attenuating mutations into the WT E by site directed mutagenesis (e.g. NY99 strain for development of a human West Nile vaccine). In contrast to neurotropic flaviviruses such as TBEV and JEV, where residues involved in virulence had been previously identified to lie within the envelope proteins, no such residues were known for non-neurotropic viruses such as dengue. Therefore for construction of chimeric vaccine viruses for non-neurotropic viruses such as dengue, it was envisioned that the wild type E sequences linked to the YFV 17D backbone should be sufficient for attenuation of the chimeric viruses. This was indeed turned out to be true as was shown by preclinical and clinical testing (see below).

All chimeric vaccines are immunogenic and safe in humans, and are either waiting registration (JEV) or are undergoing large-scale efficacy studies (DEN) [27]. The particular – live attenuated and chimeric – nature of theses vaccines required however an extensive preclinical and clinical characterization, from the early stages of research to clinical development. In fact, the licensure of new vaccines is more challenging now than it was more than half a century ago, in particular in the case of live-attenuated vaccines, despite decades of proven efficacy and good to excellent safety record of long-established vaccines such as live polio and YF vaccines. The situation is even more complex for chimeric vaccine candidates, which are genetically modified organisms (GMO), and they have to comply with additional and specific regulations. The different tools, assays and strategies developed to address these points will be presented in this review, focusing to some extent on dengue vaccines, which generated diverse and in-depth evaluation because of their particular complexity. As presented in Fig. 2, we will first address vaccine characterization (phenotypic and genetic properties/stability), followed by preclinical in vitro or animal evaluation, and then clinical testing. Finally, we will address the theoretical issues linked to the live GMO nature of the ChimeriVax-based vaccines.

Section snippets

Phenotypic and genotypic characterization of chimeric vaccine candidates

Compared to live-attenuated vaccines obtained by empirical passages, ChimeriVax-based vaccines are rationally designed, starting from cDNA to plaque-purified clonal vaccines. Further scale-up of viral vaccine production from laboratory scale to GMP manufacture requires multiple rounds of virus amplification, which have to be strictly controlled in order to avoid the loss of critical attenuation properties. The seed lot system (primary or master seed lot [MSL] and secondary/working seed lot

Preclinical evaluation of safety and immunogenicity

Before entering clinical trials, all candidate vaccines must be tested for safety and immunogenicity in preclinical studies. Such evaluations can be conducted both in vitro on primary or transformed cells – including human cells – and in vivo in animal models, in particular non-human primates (NHP). In the case of arboviral vaccines, studies with potential vectors also have to be documented. Results obtained in these models can be correlated with safety and immunogenicity of the candidate

Clinical evaluation

The three different chimeric vaccine candidates against DENV, JEV and WNV have now been tested extensively in clinical trials, in volunteers ranging from children to the elderly. All results obtained so far demonstrate their excellent safety profile, with low/absent viremia of short duration [41], [62]. Satisfactory immunogenicity is obtained after a single dose (JEV, WNV) or after 2–3 doses (DENV). The following paragraphs will describe the main findings and conclusions obtained from these

Evaluation of theoretical safety/GMO issues

It was important to address the theoretical issues linked to both the live and GMO nature of ChimeriVax-based vaccines throughout their development. The four most frequently raised concerns were specifically addressed: transmission by arthropod vectors, reversion to virulence, recombination with a circulating virus, and risks of viscerotropism. A summary of each of these potential risks follows and the main points are summarized in Table 3.

Dengue-specific issue: antibody-dependent enhancement (ADE)

As stated previously, the most often proposed markers of protection are seroneutralizing antibodies, as measured for instance by the PRNT50, although titres required for protection are yet to be determined [59], [111]. Cross-protection between DENV serotypes is limited and antibody dependent enhancement (ADE) has been proposed to constitute one mechanism leading to severe dengue disease, e.g. DHF or DSS [65], [66]: pre-existing non-neutralizing heterotypic antibodies, instead of being

Conclusions

The ChimeriVax candidates developed against DENV, WNV and JEV demonstrated satisfactory safety and immunogenicity characteristics in in vitro and in vivo pre-clinical tests. We tried however to consider potential issues linked to these chimeric vaccines, unlikely as their occurrences may be. The utilization of the infectious clone and recombinant technologies facilitated their rapid evaluation.

Clinical data generated in humans with the chimeric dengue vaccines demonstrated expected safety and

Acknowledgements

The authors would like to acknowledge Simon Jones for editorial assistance and Claude Meric for critical reading of the manuscript.

References (123)

  • K.H. Eckels et al.

    Japanese encephalitis virus live-attenuated vaccine, Chinese strain SA14-14-2; adaptation to primary canine kidney cell cultures and preparation of a vaccine for human use

    Vaccine

    (1988)
  • T.P. Monath et al.

    Safety testing for neurovirulence of novel live, attenuated flavivirus vaccines: infant mice provide an accurate surrogate for the test in monkeys

    Biologicals

    (2005)
  • D.W. Beasley et al.

    Protection against Japanese encephalitis virus strains representing four genotypes by passive transfer of sera raised against ChimeriVax-JE experimental vaccine

    Vaccine

    (2004)
  • A.S. De Groot et al.

    Analysis of ChimeriVax Japanese encephalitis virus envelope for T-cell epitopes and comparison to circulating strain sequences

    Vaccine

    (2007)
  • T.P. Monath et al.

    Recombinant, chimaeric live, attenuated vaccine (ChimeriVax) incorporating the envelope genes of Japanese encephalitis (SA14-14-2) virus and the capsid and nonstructural genes of yellow fever (17D) virus is safe, immunogenic and protective in non-human primates

    Vaccine

    (1999)
  • B. Guy et al.

    Cell-mediated immunity induced by chimeric tetravalent dengue vaccine in naive or flavivirus-primed subjects

    Vaccine

    (2008)
  • T.P. Monath et al.

    Clinical proof of principle for ChimeriVax: recombinant live, attenuated vaccines against flavivirus infections

    Vaccine

    (2002)
  • J. Hombach et al.

    Report on a WHO consultation on immunological endpoints for evaluation of new Japanese encephalitis vaccines. WHO, Geneva. 2–3 September 2004

    Vaccine

    (2005)
  • J.S. Mackenzie et al.

    Emerging flaviviruses: the spread and resurgence of Japanese encephalitis, West Nile and dengue viruses

    Nat Med

    (2004)
  • N.P. Lindsey et al.

    Neuroinvasive disease incidence in the United States, 2002–2006

    Vector-Borne Zoonotic Dis

    (2008)
  • K.O. Murray et al.

    Clinical investigation of hospitalized human cases of West Nile virus infection in Houston, Texas, 2002–2004

    Vector-Borne Zoonotic Dis

    (2008)
  • T.P. Monath

    Prospects for development of a vaccine against the West Nile virus

    Ann N Y Acad Sci

    (2001)
  • A.F. van den Hurk et al.

    Ecology and geographical expansion of Japanese encephalitis virus

    Annu Rev Entomol

    (2009)
  • A.F. van-den-Hurk et al.

    Domestic pigs and Japanese encephalitis virus infection, Australia

    Emerg Infect Dis

    (2008)
  • T.P. Monath

    Japanese encephalitis vaccines: current vaccines and future prospects

    Curr Topics Microbiol Immunol

    (2002)
  • K.V. Pugachev et al.

    New developments in flavivirus vaccines with special attention to yellow fever

    Curr Opin Infect Dis

    (2005)
  • S.S. Whitehead et al.

    Prospects for a dengue virus vaccine

    Nat Rev Microbiol

    (2007)
  • D.W. Beasley et al.

    Current use and development of vaccines for Japanese encephalitis

    Expert Opin Biol Ther

    (2008)
  • S. Delagrave et al.

    Research and development of chimeric flavivirus vaccines

  • T.J. Chambers et al.

    Yellow fever/Japanese encephalitis chimeric viruses: construction and biological properties

    J Virol

    (1999)
  • F. Guirakhoo et al.

    Recombinant chimeric yellow fever-dengue type 2 virus is immunogenic and protective in nonhuman primates

    J Virol

    (2000)
  • J. Arroyo et al.

    ChimeriVax-West Nile virus live-attenuated vaccine: preclinical evaluation of safety, immunogenicity, and efficacy

    J Virol

    (2004)
  • T.P. Monath et al.

    Chimeric yellow fever virus 17D-Japanese encephalitis virus vaccine: dose-response effectiveness and extended safety testing in rhesus monkeys

    J Virol

    (2000)
  • M.C. Bonaldo et al.

    The yellow fever 17D vaccine virus as a vector for the expression of foreign proteins: development of new live flavivirus vaccines

    Mem Inst Oswaldo Cruz

    (2000)
  • T.J. Chambers et al.

    Yellow fever virus/dengue-2 virus and yellow fever virus/dengue-4 virus chimeras: biological characterization, immunogenicity, and protection against dengue encephalitis in the mouse model

    J Virol

    (2003)
  • R.G. van der Most et al.

    Chimeric yellow fever/dengue virus as a candidate dengue vaccine: quantitation of the dengue virus-specific CD8 T-cell response

    J Virol

    (2000)
  • R. Galler et al.

    Attenuation and immunogenicity of recombinant yellow fever 17D-dengue type 2 virus for rhesus monkeys

    Braz J Med Biol Res

    (2005)
  • Guy B, Guirakhoo F, Watson M, Higgs S, Monath TP. Safety of flavivirus chimeric vaccines: answer to Ishikawa et al....
  • S. Lehrman

    Nature

    (1999)
  • Y. Zhang et al.

    Structure of immature flavivirus particles

    EMBO J.

    (2003)
  • S.B. Halstead et al.

    Selection of attenuated dengue 4 viruses by serial passage in primary kidney cells. III. Reversion to virulence by passage of cloned virus in fetal rhesus lung cells

    Am J Trop Med Hyg

    (1984)
  • W. Sirigulpanit et al.

    Substitution or deletion mutations between nt 54 and 70 in the 5’ non-coding region of dengue type 2 virus produce variable effects on virus viability

    J Gen Virol

    (2007)
  • K.V. Pugachev et al.

    High fidelity of yellow fever virus RNA polymerase

    J Virol

    (2004)
  • F. Guirakhoo et al.

    Safety and efficacy of chimeric yellow fever-dengue virus tetravalent vaccine formulations in nonhuman primates

    J Virol

    (2004)
  • B.R. Miller et al.

    Biological characterization of plaque-size variants of yellow fever virus in mosquitoes and mice

    Acta Virol

    (1988)
  • L.A. Vlaycheva et al.

    Neuroblastma cell-adapted yellow fever 17D virus: characterization of a viral variant associated with persistent infection and decreased virus spread

    J Virol

    (2002)
  • J. Arroyo et al.

    Molecular basis for attenuation of neurovirulence of a yellow fever virus/Japanese encephalitis virus chimera vaccine (ChimeriVax-JE)

    J Virol

    (2001)
  • T.P. Monath et al.

    A live, attenuated recombinant West Nile virus vaccine

    Proc Natl Acad Sci USA

    (2006)
  • S. Green et al.

    Immunopathological mechanisms in dengue and dengue hemorrhagic fever

    Curr Opin Infect Dis

    (2006)
  • D.H. Libraty et al.

    Human dendritic cells are activated by dengue virus infection: enhancement by gamma interferon and implications for disease pathogenesis

    J Virol

    (2001)
  • Cited by (246)

    View all citing articles on Scopus
    View full text