You are on page 1of 9

Vaccine 29 (2011) 7276–7284

Contents lists available at ScienceDirect

Vaccine
journal homepage: www.elsevier.com/locate/vaccine

Next generation dengue vaccines: A review of candidates in preclinical


development夽
Julia Schmitz a , John Roehrig b , Alan Barrett c , Joachim Hombach a,∗
a
Initiative for Vaccine Research, World Health Organization, Geneva, Switzerland
b
Arboviral Diseases Branch, Centers for Disease Control and Prevention, Fort Collins, CO, USA
c
Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, USA

a r t i c l e i n f o a b s t r a c t

Article history: Dengue represents a major public health problem of growing global importance. In the absence of spe-
Available online 21 July 2011 cific dengue therapeutics, strategies for disease control have increasingly focused on the development
of dengue vaccines. While a licensed dengue vaccine is not yet available, several vaccine candidates
Keywords: are currently being evaluated in clinical trials and are described in detail in accompanying articles. In
Dengue vaccines addition, there are a large variety of candidates in preclinical development, which are based on diverse
Dengue
technologies, ensuring a continued influx of innovation into the development pipeline. Potentially, some
Dengue virus
of the current preclinical candidates may become next generation dengue vaccines with superior prod-
uct profiles. This review provides an overview of the various technological approaches to dengue vaccine
development and specifically focuses on candidates in preclinical development.
© 2011 World Health Organization.. Published by Elsevier Ltd. All rights reserved.

1. Introduction of dengue immunity, challenges to vaccine development and the


various technologies used, while the second part provides a more
Dengue is a mosquito-borne flavivirus disease that has spread detailed description of specific vaccine projects in preclinical devel-
to most tropical and many subtropical areas and creates a signif- opment.
icant burden of disease and economic costs in endemic countries
[1–4]. As specific dengue therapeutics are not available and disease
prevention is limited to vector control measures, the development 2. Overview of dengue vaccine development
of a dengue vaccine would represent a major advance in the control
of the disease (reviewed in [5]). While no licensed dengue vaccine 2.1. Dengue immunity and challenges to vaccine development
currently exists, vaccine development has progressed considerably
in recent years. Several candidates are currently undergoing either The disease dengue is caused by four dengue viruses (DENV),
clinical evaluation or preclinical development. These have been DENV-1 to DENV-4. Due to their serological and genetic relatedness
reviewed recently [6–9]. they are considered four serotypes of DENV. Multiple DENV can
This review focuses on dengue vaccine candidates in preclin- co-circulate in endemic areas (reviewed in [10]). Infection by one
ical development. It is based on published and unpublished data serotype confers lasting protection against re-infection by the same
presented by vaccine developers and researchers at the WHO/IVI serotype, but only transient protection against secondary infec-
Informal Consultation on Next Generation Dengue Vaccines and tion by one of the three heterologous serotypes (reviewed in [9]).
Diagnostics (1–2 November 2010, Atlanta). Additional preclinical Dengue vaccine development efforts therefore aim for a tetrava-
candidates from the published literature are included in the review lent vaccine which simultaneously provides long-term protection
to illustrate the wide range of strategies applied to dengue vaccine against all DENV serotypes.
development. The first part of the review presents an overview The mechanism of protective immunity against dengue is not
fully understood. There is considerable evidence for a major role
of antibody-mediated DENV neutralization in protection against
infection and disease [9]. However, it is unclear what quantity of
夽 Disclaimer: Two of the authors (JH, JS) are staff members of the World Health neutralizing antibody is needed for protective immunity, and it is
Organization. The authors alone are responsible for the views expressed in this pub- increasingly likely that the contribution of neutralizing antibodies
lication and they do not necessarily represent the decisions, policy or views of the
World Health Organization.
to protection will not become known until efficacy trials of candi-
∗ Corresponding author. Tel.: +41 22 791 4531; fax: +41 22 791 4860. date vaccines have been completed. In addition, contributions of
E-mail address: hombachj@who.int (J. Hombach). other immune mechanisms such as cytotoxic T cell responses are

0264-410X/$ – see front matter © 2011 World Health Organization.. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.vaccine.2011.07.017
J. Schmitz et al. / Vaccine 29 (2011) 7276–7284 7277

less clear (reviewed in [9,11,12]). Further research is needed to bet- tective efficacy of vaccine candidates in NHPs therefore measure
ter define and validate immune correlates of protection for vaccine prevention or reduction of viremia upon viral challenge [28].
development [11].
Another challenge for vaccine development is the potentially 2.2. Technological approaches to dengue vaccine development
detrimental role of immune enhancement in dengue pathogene-
sis [5]. Severe disease is most commonly observed in secondary, A wide range of vaccine technologies has been applied to
heterologous DENV infections. Antibody-dependent enhancement dengue vaccine development, including live attenuated virus (LAV),
(ADE) of infection has been proposed as the primary mechanism purified inactivated virus (PIV), recombinant subunits, virus-like
of dengue immunopathogenesis [13,14]. In vitro studies suggest particles (VLPs), and plasmid or viral vectors. Each of these
that the capacity of DENV antibodies to contribute to neutraliza- approaches has its advantages and challenges.
tion or enhancement of infection is determined by multiple factors, LAV vaccines have been shown to produce robust, lasting
including antibody specificity, antibody affinity, antibody titre and and broad immunity, including humoral and cellular immune
epitope accessibility (reviewed in [15,16]). responses [34]. In addition, their production cost tends be lower
The potential risk of immune enhancement of infection and dis- than for many other vaccine technologies. However, it has often
ease underscores the importance of developing dengue vaccines proven difficult to achieve a level of attenuation which opti-
which produce balanced, long-lasting immunity to all four DENV mally balances low reactogenicity (e.g. avoidance of clinical dengue
serotypes. A tetravalent vaccine eliciting protective, neutralizing symptoms) with sufficiently high immunogenicity. In addition, LAV
antibody responses against all serotypes should address theoretical vaccines should replicate well in cell culture in order to facilitate
concerns about vaccine-induced ADE. However, vaccine-induced their production, while their transmissibility by mosquitoes should
ADE might again become problematic as antibody titres wane post- be prevented or strongly reduced. Attenuation strategies for live
vaccination. dengue vaccines include serial cell passage (e.g. in Primary Dog Kid-
DEN virions are composed of a lipid envelope modified by the ney cells). The first tetravalent dengue vaccine was produced in this
insertion of envelope (E) proteins and premembrane/membrane way [35]. Additionally, introduction of selected mutations or cre-
(prM/M) proteins (depending on the maturation state [17]), sur- ation of chimeric viruses by recombinant DNA technology can be
rounding a nucleocapsid composed of capsid (C) proteins and the used. Success of these strategies is however often limited by lack
viral RNA genome (reviewed in [18]). Human antibodies raised of a detailed understanding of the molecular determinants of vir-
against the DEN virion are mostly targeted at the E and prM ulence due to unavailability of animal models, as described above.
proteins [19]. Research to determine the most suitable target epi- There are also concerns about the genetic stability of LAV vaccines,
topes for vaccines is ongoing, and while considerable progress including the possibility of reversion to a more virulent pheno-
has been made in the characterization of the humoral immune type and the theoretical risk of recombination between vaccine
response to DENV, important knowledge gaps still exist. There and wild-type viruses. Furthermore, the need to achieve a bal-
is evidence suggesting that anti-E antibodies have higher type- anced immune response against all four DENV serotypes presents
specific neutralization capacity and lower ADE potential than a particular challenge for multi-component LAV vaccines. Repli-
anti-prM antibodies. Moreover, most potent DENV neutralizing cation interference between the different serotypes of DENV in
antibodies identified so far recognize epitopes in domain III of tetravalent formulations has been observed, resulting in a reduc-
the E protein (EDIII) which is involved in binding of DENV to tion of neutralizing antibodies to some serotypes, when compared
cell receptors [19–27]. The conundrum of the human antibody to monovalent formulations. LAV vaccines represent the majority
response to DENV infection is that most antibodies are specific for of dengue vaccine candidates in clinical evaluation [36–47], and
domain II of the E protein (EDII) [19], which contains a variety of additional candidates are in preclinical development [48–53].
serotype and flavivirus cross-reactive epitopes. The contribution of Nonliving vaccines can have some advantages over LAV
the human antibody bias to EDII to sensitization for ADE has not vaccines, including reduced potential for reactogenicity and bet-
been determined. Because of its antigenic nature, EDIII is consid- ter suitability for immunocompromised individuals. Moreover,
ered an antigenic target of particular relevance for dengue vaccine achievement of a balanced immune response is facilitated by lack of
development. New vaccine strategies may have to be developed replication competition between tetravalent vaccine components.
to enhance the human anti-EDIII response. Antibodies directed However, immune responses to nonliving vaccines tend to be less
against the C protein and against non-structural proteins, such as broad, potent and durable compared to LAV vaccines. Adjuvants
NS1, have also been detected in DENV-infected individuals, but have been shown to improve immunogenicity of nonliving vac-
their role in dengue immunity or immunopathogenesis remains cines, but safety concerns raised in relation to some adjuvants need
unclear [9]. to be addressed [54]. Also, access to adjuvants has proven difficult
Dengue vaccine development has been hampered by the lack for many vaccine developers.
of an adequate animal model for the disease [28]. Normal mice PIV dengue vaccines are killed wild type virions, containing all
do not display significant viremia or disease when infected with DENV structural proteins and viral RNA. This permits induction of
human DENV isolates. Intracerebral challenge of mice with cer- an immune response to the three DENV structural proteins. PIV
tain mouse-adapted DENV strains produces a paralysis phenotype, dengue vaccines have not yet reached clinical evaluation, but sev-
and this intracerebral challenge model has been used to evalu- eral candidates are in preclinical development [55,56].
ate protective efficacy of vaccine candidates in mice. Furthermore, Other nonliving vaccines express particular DENV antigens,
immunocompromised mouse models have been developed which which allows for targeting of the immune response to antigens
show some of the characteristics of human disease and can also be with particular desirable properties (e.g. induction of potent neu-
used as an in vivo system for studying ADE [29–31]. However, con- tralizing antibodies). Recombinant subunit dengue vaccines have
cerns remain about whether the full spectrum of dengue immunity been produced in a variety of protein expression systems, including
and disease can be modeled in this immunocompromised setting. bacterial, yeast, insect and mammalian cells. Most dengue sub-
Humanized mice that lack a murine antibody response but demon- unit vaccines express truncated versions of the E protein that have
strate some symptoms that re-capitulate human infection are also deleted transmembrane domains, or simply express EDIII alone.
being investigated [32,33]. Non-human primates (NHPs) are sus- While subunit vaccines often show low reactogenicity, achieve-
ceptible to infection by DENV and develop viremia, but do not ment of sufficient immunogenicity usually requires the use of
show significant clinical signs of infection. Studies to address pro- adjuvants. Various recombinant dengue subunit vaccines are in
7278 J. Schmitz et al. / Vaccine 29 (2011) 7276–7284

preclinical development [57–64], and the most advanced candidate being studied in the context of heterologous prime-boost strategies
[65] is being evaluated in phase 1 clinical trials. [64,92].
VLP vaccines are virus-like particles that do not contain replica- A subunit vaccine candidate developed by VaxInnate is based
tive genetic material, but permit presentation of antigens in a on fusion of DENV EDIII to a proprietary form of flagellin (STF2D),
repetitive, ordered array similar to the virion structure, which is which serves as an adjuvant. STF2D contains the toll-like receptor
thought to increase immunogenicity [66]. Several dengue VLP vac- 5 (TLR5) activation domain from Salmonella typhimurium flagellin
cine candidates are currently in preclinical development [67–70]. [93]. This approach links adaptive and innate immunity and is
DNA vaccines and virus-vectored vaccines allow for in vivo believed to mimic natural infection where antigen and TLR lig-
expression of antigens, often in the form of DENV VLPs. One of and are contained in a single pathogen. Physical linkage of TLR
the advantages of DNA vaccines is their thermostability, which ligand to antigen has been shown to be more specific and effi-
avoids cold-chain requirements for vaccine storage and transport. cient than co-administration, requiring lower doses of adjuvant.
However, DNA vaccines often face challenges in terms of adequate STF2D fusion proteins can be expressed and purified in E. coli. In
cellular uptake and expression. Improved DNA vaccine delivery sys- immunogenicity studies in mice, monovalent EDIII-STF2D fusion
tems have shown some success in addressing these issues [71]. proteins were shown to elicit neutralizing DENV antibodies and
Various DNA dengue vaccines are currently in preclinical develop- provide partial protection against viral challenge. Bivalent DENV-
ment [72–81]. The most advanced DNA dengue vaccine candidate 1/3 and DENV-2/4 constructs were then generated by fusing the
has progressed to phase 1 clinical trials [82]. EDIII domains of two different serotypes to separate attachment
Virus-vectored dengue vaccines express DENV antigens from a points on STF2D. Subcutaneous immunization of mice with three
viral vaccine vector. Viral vectors allow for efficient infection of doses (3 ␮g each) of a DENV-1/3 EDIII-STF2D fusion protein resulted
target cells. Moreover, several well-characterized vaccine vector in robust and balanced titres of neutralizing antibodies against
platform technologies exist, some of which are used for licensed DENV-1 and DENV-3. It was found that the choice of antigen attach-
vaccines with a well-known safety record [83]. However, there ment points (C-terminal fusion versus insertion via replacement
are concerns that pre-existing immunity to viral vaccine vectors of the flagellin D3 domain) affects immunogenicity. Longevity of
could reduce the effectiveness of some virus-vectored dengue the immune response is yet to be studied. Additional bivalent con-
vaccines. Several virus-vectored dengue vaccine candidates are structs are being generated and tested, with the aim of combining
currently in preclinical development, including candidates based two suitable bivalent vaccine candidates to a tetravalent vaccine.
on non-replicating vectors [84,85], single-cycle vectors [86,87] and In a development approach by the International Centre for
replication-competent vectors [88]. Genetic Engineering and Biotechnology (ICGEB) in India, a tetrava-
Heterologous prime-boost approaches, which use different vac- lent chimeric EDIII fusion protein was generated, which consists of
cine types for the priming and boosting steps of an immunization the EDIII domains of DENV-1,-2,-3,-4 joined by flexible peptide link-
schedule, may allow for optimization of immune responses by com- ers [59]. This single tetravalent approach avoids the complexities
bining advantages of different vaccine technologies in a unique related to producing tetravalent mixtures of monovalent vaccine
order of vaccination. However, the increased complexity of vaccina- components. The tetravalent chimeric EDIII protein was expressed
tion schedules could make heterologous prime-boost approaches in Pichia pastoris. Immunization of mice with the tetravalent vac-
more difficult to implement in the context of immunization cine candidate adjuvanted with montanide resulted in neutralizing
programmes. Several heterologous prime-boost approaches are antibody responses against all DENV serotypes.
currently evaluated in preclinical studies [89,90]. A different approach to the development of a single tetrava-
lent subunit vaccine was taken at the Taiwanese National Health
Research Institutes (NHRI). A consensus EDIII amino acid sequence
3. Dengue vaccine candidates in preclinical was derived by sequence analysis of different DENV-1-4 strains,
development and the consensus EDIII protein was expressed in E. coli. The single
tetravalent vaccine candidate adjuvanted with alum induced neu-
3.1. Recombinant subunit vaccines tralizing antibody responses against all DENV serotypes in mice
[61]. This vaccine candidate was developed further by fusion of
Several recombinant subunit vaccine candidates have been the consensus EDIII with a fragment of the Ag473 lipoprotein of N.
developed by the Pedro Kourí Tropical Medicine Institute (IPK) in meningitidis. The consensus EDIII-Ag473 fusion protein was found
collaboration with the Center for Genetic Engineering and Biotech- to induce higher levels of neutralizing antibodies in mice than con-
nology (CIGB) in Cuba (Table 1). One approach is based on fusion sensus EDIII formulated in alum [60]. The intrinsic adjuvant activity
of DENV EDIII to the carrier protein p64k of Neisseria meningitidis. of the EDIII-Ag473 lipo-immunogen appears to be due to stim-
The EDIII-p64k fusion protein is expressed in E. coli. Monovalent ulation of innate immunity through the TLR2 signaling pathway
vaccine candidates for all DENV serotypes were shown to induce [94].
neutralizing antibodies and protect against viral challenge in mice.
DENV-1 and DENV-2 monovalent candidates have also been eval- 3.2. DNA vaccines
uated in NHPs. Monkeys were immunized subcutaneously with
four doses of the monovalent vaccine (50–100 ␮g protein per dose, A tetravalent DNA vaccine candidate developed by Inovio Phar-
formulated in Freund’s adjuvant). The monovalent vaccine can- maceuticals consists of a DNA plasmid vector that expresses a single
didates were found to be immunogenic and provided protection ORF comprising the EDIII domains of all four DENV serotypes sep-
against viral challenge [58,91]. Adjuvants suitable for human use arated by proteolytic cleavage sites [80]. In vivo expression of a
are under evaluation, including N. meningitidis serogroup A cap- single tetravalent EDIII fusion protein, which is subsequently pro-
sular polysaccharide (CPS-A) adsorbed on aluminium hydroxide cessed into monovalent EDIII proteins by cellular proteases, aims
[63]. In another approach, a DENV EDIII-capsid fusion protein was to ensure that the four EDIII domains are presented in equal ratios
expressed in E. coli and mixed in vitro with oligodeoxynucleotides in order to facilitate a balanced immune response. A synthetic con-
to obtain particulated aggregates. The aggregated DENV-2 EDIII- sensus sequence for the EDIII domain of each DENV serotype was
capsid fusion protein induced both humoral and cellular immune derived by sequence comparison of approximately 15 different
responses in NHPs and provided partial protection against viral strains. Sequences were also human codon optimized to improve
challenge [62]. The different subunit vaccine candidates are also expression levels. Administration of the vaccine is via an improved
J. Schmitz et al. / Vaccine 29 (2011) 7276–7284 7279

Table 1
Dengue vaccine candidates in preclinical development.

Technological Vaccine developer Details Development stage


approach

Recombinant subunit IPK/CIGB EDIII-p64k fusion proteins and EDIII-capsid fusion Monovalent candidates evaluated
vaccines proteins expressed in E. coli [58,62–64] in NHPs
VaxInnate Bivalent EDIII-STF2D fusion proteins expressed in Bivalent candidate evaluated in
E. coli mice
ICGEB Tetravalent chimeric EDIII expressed in P. pastoris Tetravalent candidate evaluated in
[59] mice
NHRI Tetravalent consensus EDIII-Ag473 fusion protein Tetravalent candidate evaluated in
expressed in E. coli [60,61,94] mice

DNA vaccines Inovio Pharmaceuticals Tetravalent chimeric EDIII with inbuilt cleavage Tetravalent candidate evaluated in
sites expressed from plasmid vector [80] NHPs
Kobe University prM/E expressed from plasmid vector [78] Tetravalent candidate evaluated in
mice
CDC prM/E expressed from plasmid vector [72,75] Tetravalent candidate evaluated in
NHPs
NMRC Tetravalent “shuffled” prM/E expressed from Tetravalent candidates evaluated
plasmid vector [76] in NHPs

VLP vaccines Cytos Biotechnology EDIII chemically coupled to Qß VLPs expressed in Tetravalent candidate evaluated in
E. coli mice
ICGEB EDIII-HBsAg VLPs or ectoE-based VLPs expressed Monovalent candidates generated
in P. pastoris
Kobe University prM/E VLPs expressed in insect cells (Sf9) [69] Monovalent candidate evaluated in
mice

Virus-vectored vaccines ICGEB Tetravalent chimeric EDIII expressed from Tetravalent candidate evaluated in
replication-deficient adenovirus vector [85] mice
GenPhar/NMRC Bivalent prM/E expressed from Tetravalent candidate evaluated in
replication-deficient adenovirus vector [84] NHPs
UNC EDIII, E85 or prM/E expressed from single-cycle Monovalent candidates evaluated
VEE virus vector [86] in NHPs
UTMB prM/E expressed from single-cycle West Nile virus Monovalent candidate evaluated in
vector [87] mice
Themis Bioscience/Institut Pasteur Tetravalent EDIII and DENV-1 ectoM expressed Tetravalent candidate evaluated in
from live attenuated measles virus vector [88] NHPs

Purified inactivated virus NMRC Psoralen-inactivated DENV [56] Monovalent candidate evaluated in
vaccines NHPs
GSK/WRAIR/FIOCRUZ Purified inactivated DENV [55] Tetravalent candidate evaluated in
NHPs

Live attenuated virus FIOCRUZ DENV prM/E expressed from live attenuated Monovalent candidates evaluated
vaccines chimeric YF 17D/DEN virus [48–50] in NHPs
Chiang Mai University/Mahidol DEN/DEN chimeric viruses [51,52] Monovalent candidates evaluated
University/NSTDA/BioNet-Asia in mice

Heterologous prime-boost NMRC prM/E expressed from plasmid vector (prime) and Monovalent candidates evaluated
approaches from single cycle VEE virus vector (boost) [89] in NHPs
NMRC/WRAIR Purified inactivated DENV or plasmid vector Tetravalent candidates evaluated
expressing prM/E (prime) and live attenuated in NHPs
DENV (boost) [90]

Abbreviations: CDC: Centers for Disease Control and Prevention, USA; CIGB: Center for Genetic Engineering and Biotechnology, Cuba; FIOCRUZ: Oswaldo Cruz Foundation,
Brazil; GSK: GlaxoSmithKline Biologicals; ICGEB: International Centre for Genetic Engineering and Biotechnology, India; IPK: Pedro Kourí Tropical Medicine Institute, Cuba;
NHRI: National Health Research Institutes, Taiwan; NMRC: Naval Medical Research Center, USA; NSTDA: National Science and Technology Development Agency, Thailand;
UNC: University of North Carolina at Chapel Hill, USA; UTMB: University of Texas Medical Branch, USA; WRAIR: Walter Reed Army Institute of Research, USA.

DNA delivery system based on electroporation-enhanced trans- free jet injector. Mice were protected against viral challenge 3
fection, suitable for both intradermal and intramuscular routes. weeks after the last dose, and persistence of neutralizing antibodies
Electroporation-based DNA immunization is currently being tested was observed during a 30-week follow-up [95]. Immunogenicity
in phase 1 clinical trials for HIV and influenza prevention and in of the tetravalent DNA vaccine in mice was further increased by
phase 2 clinical trials for hepatitis C, HPV 16/18 and AML/CML can- co-immunization with DENV-2 VLPs generated by co-expression
cer therapeutic studies. Immunization of mice with the tetravalent of prM/E in cell culture. Immunogenicity of the DNA vaccine was
dengue vaccine candidate, using intramuscular electroporation of also found to be improved by co-immunization with an inactivated
three doses (10 ␮g DNA) at biweekly intervals, resulted in neutral- Japanese encephalitis vaccine [78].
izing antibody titres against all four DENV serotypes. Immunization A similar tetravalent DNA vaccine candidate based on prM/E
of NHPs was also shown to elicit robust tetravalent antibody expression has been developed by the U.S. Centers for Disease Con-
responses. Further studies to assess DENV cross-reactive versus trol and Prevention (CDC). Transfection of the recombinant plasmid
serotype-specific antibody responses are in progress. vectors into cultured cells has been shown to result in secretion of
Another tetravalent DNA vaccine candidate developed at Kobe prM/E containing VLPs, which have an antigenic structure similar
University is composed of a mixture of four plasmid vectors, each to DEN virions [72,75]. Immunogenicity of a tetravalent mixture
of which expresses the prM and E proteins (prM/E) of one DENV of four monovalent DNA vaccines was evaluated in NHPs (two
serotype. Two doses of the tetravalent vaccine (100 ␮g per dose, at dose schedule one month apart; 500 ␮g per monovalent compo-
three week intervals) were administered to mice using a needle- nent; administration by intramuscular injection). The tetravalent
7280 J. Schmitz et al. / Vaccine 29 (2011) 7276–7284

vaccine was found to stimulate a balanced immunity that lasted expression of EDIII alone does not result in VLP formation, mosaic
for 10 months and protected from viral challenge. Pre-existing VLPs composed of varying ratios of EDIII-HBsAg and HBsAg could
antibodies against DENV or other flaviviruses were shown to be generated by co-expression of the EDIII-HBsAg fusion protein
improve immunogenicity of the vaccine. Moreover, there is evi- with 1–4 copies of HBsAg. In a second approach, a fusion pro-
dence suggesting that the safety and immunogenicity of the vaccine tein consisting of the E protein ectodomain (ectoE) and 30 amino
candidates could be further improved by manipulating antigenic residues of the prM protein was expressed in P. pastoris, which
determinants of the E protein. In order to reduce the potential for also resulted in VLP formation. Structural characterization of the
ADE by cross-reactive antibodies, flavivirus group cross-reactive VLPs, their functional evaluation for immunogenicity, and evalu-
epitopes and DENV serocomplex cross-reactive epitopes in the ation of prime-boost protocols combining VLP vaccine candidates
E protein were mapped with monoclonal antibody panels and with rAd5 vectored vaccine candidates are planned.
mutated to reduce cross-reactivity, resulting in the formation of In a VLP vaccine development approach employed at Kobe
cross-reactivity reduced VLPs in cell culture [96]. When evaluated University, prM/E is expressed from a plasmid vector transfected
in mice, cross-reactivity reduced DENV-1 and DENV-2 VLP vac- into cell culture-based expression systems and secreted VLPs are
cines continued to elicit high levels of neutralizing antibodies, but purified. A DENV-2 VLP vaccine candidate was found to be immuno-
showed reduced ADE potential compared to wild type vaccines genic and protective in mice [103]. Expression of VLPs by transient
both in vitro (K562 cell-based assay) and in vivo (AG129 mouse transfection of insect cells (Sf9 cell line) resulted in 10–100-fold
model [29,97]). Immunogenicity of the cross-reactivity reduced improved yields compared to mammalian expression systems
vaccines was further improved by incorporating a CD4 epitope from [69]. Immunogenicity of the DENV-2 VLP vaccine candidate in
the transmembrane domain of West Nile virus E protein into the mice could be improved by co-immunization with a DNA vaccine
expression construct. expressing prM/E, which also functions as a CpG adjuvant.
In a single tetravalent approach developed at the U.S. Naval
Medical Research Center (NMRC), a prM/E construct containing a 3.4. Virus-vectored vaccines
chimeric “shuffled” E protein is expressed from a plasmid vector
[76]. Various chimeric E proteins containing sequence portions of A virus-vectored dengue vaccine developed by ICGEB is based
all DENV serotypes were derived by DNA shuffling technology. The on expression of a tetravalent chimeric EDIII fusion protein from
shuffled constructs were screened for antigen expression in vitro a replication-deficient adenovirus vaccine vector [85]. Advantages
and for immunogenicity in mice. Three selected constructs were of adenovirus vaccine vectors include a large insert capacity, effi-
evaluated as single tetravalent DNA vaccine candidates in NHPs. cient delivery and high expression levels of antigens in a variety of
Two of these candidates were found to induce tetravalent neutral- cells, and a well-documented human safety record. Immunization
izing antibody responses in the majority of animals and to provide of mice with two doses of the single component tetravalent dengue
partial protection against viral challenge. vaccine candidate (administered 30 days apart by the intraperi-
toneal route) resulted in balanced neutralizing antibody responses
3.3. VLP vaccines against all four DENV serotypes. Of note, prior immunity to the ade-
novirus vaccine vector did not impair the potency of the dengue
A VLP dengue vaccine candidate developed by Cytos Biotechnol- vaccine candidate. Presence of antibodies against the vector in the
ogy is based on chemical coupling of recombinant EDIII to carrier sera of immunized mice enabled uptake of the vector into U937
VLPs derived from bacteriophage Q␤ [98]. A similar approach has human monocytic cells, which are not susceptible to infection in the
recently been applied to the development of a West Nile virus vac- absence of antibodies against the vector. This effect is presumably
cine [99]. About 60–90 EDIII molecules can be coupled to one VLP mediated by the Fc receptor pathway [104]. The results suggest that
carrier, resulting in the formation of a highly repetitive, ordered low levels of pre-existing antibodies against the adenovirus vec-
array of antigen, which is expected to increase immunogenicity. tor may in fact even facilitate uptake of the virus-vectored dengue
Q␤ VLPs also contain E. coli-derived ssRNA, which has been shown vaccine into antigen-presenting cells.
to be required for IgG isotype switching in mice immunized with Another virus-vectored vaccine approach developed by Gen-
Q␤ VLPs [100,101] and is used as a built-in adjuvant to improve the Phar in collaboration with the NMRC is based on expression
antibody response to Q␤ VLP-based vaccines. Q␤ VLPs can be eco- of DENV prM/E from a replication-deficient complex adenovirus
nomically produced in E. coli, resulting in 2 g/l culture of GMP grade vaccine vector capable of accommodating multiple large antigen
material. Q␤ VLP based vaccine candidates are in advanced clinical inserts [84]. The tetravalent vaccine is composed of two bivalent
evaluation for other target antigens. Four monovalent dengue vac- vaccines, each of which expresses prM and E proteins of two dif-
cine candidates were generated by coupling the EDIII domain of ferent DENV serotypes. NHPs were immunized with two doses of
the different DENV serotypes to the carrier VLP. Immunogenicity tetravalent vaccine 8 weeks apart, followed by viral challenge at
studies in mice were performed to compare monovalent vaccines 3–6 months. Neutralizing antibody responses were induced against
individually and in a tetravalent formulation (three doses at 10 all serotypes. Viremia from challenge with DENV-1 and DENV-3
day intervals; 50 ␮g per dose of monovalent vaccine and 200 ␮g was blocked, while viremia from challenge with DENV-2 and DENV-
per dose of tetravalent vaccine; subcutaneous administration with 4 was significantly reduced. Further development of the vaccine
alum). The neutralizing antibody response observed to DENV-4 constructs, including optimization of all prM and E gene sequences
was weaker than for the other serotypes. However, an improved for human codon usage, resulted in an improved tetravalent for-
formulation with increased representation of the DENV-4 monova- mulation, which induced a more balanced immune response to all
lent component induced a potent neutralizing antibody response four serotypes. The candidate vaccine is scheduled to enter clinical
against all four serotypes. evaluation shortly.
VLP vaccine candidates have also been developed by the ICGEB. A virus-vectored vaccine candidate developed at the University
In a first approach, DENV E was expressed as a fusion protein with of North Carolina at Chapel Hill (UNC) is based on expression of
Hepatitis B virus surface antigen (HBsAg) in P. pastoris [67]. HBsAg DENV antigens from a single-cycle Venezuelan equine encephalitis
expressed in P. pastoris has been shown to assemble into highly (VEE) virus vaccine vector [86]. Packaging technologies based on
immunogenic VLPs. Furthermore, high protein yields (up to 7 gram helper RNAs allow for the production of infectious single-cycle VEE
HBsAg per liter culture [102]) make P. pastoris an attractive sys- virus particles also referred to as virus replicon particles (VRPs). VEE
tem for affordable production of protein-based vaccines. While VRPs have been shown to infect human dendritic cells and express
J. Schmitz et al. / Vaccine 29 (2011) 7276–7284 7281

high levels of recombinant antigens, inducing both innate and lent inactivated virus vaccine candidate together with different
adaptive immune responses. Different monovalent vaccine candi- proprietary adjuvant systems (AS01, AS03, AS04). Two doses of
dates expressing DENV prM/E proteins, C-terminally truncated E adjuvanted vaccine were given 30 days apart, followed by viral
proteins (E81, E85 [81% or 85% of E]) or EDIII were evaluated for challenge 4 months later. All immunized animals were found to
immunogenicity in mice. Vaccine candidates expressing truncated be protected against viral challenge. The highest neutralizing anti-
E proteins were found to elicit higher neutralizing antibody titres body responses were obtained with the AS03 adjuvant system.
than candidates expressing prM/E. Immunogenicity and protec- Clinical trials of the adjuvanted PIV vaccine are planned to start
tive efficacy of the monovalent candidates were evaluated further in 2012, with a first assessment of protective efficacy envisaged for
in NHP studies. Three doses (108 IU each) of a monovalent vac- 2014–15.
cine candidate expressing either DENV-3 E85 or DENV-3 prM/E
were administered subcutaneously at weeks 0, 7, and 25, and viral 3.6. Live attenuated virus vaccines
challenge was performed at week 41. Higher neutralizing anti-
body titres were observed with the DENV-3 E85 based vaccine, A live attenuated dengue vaccine candidate developed by
which also completely protected all monkeys against viral chal- FIOCRUZ uses the live attenuated yellow fever vaccine 17DD sub-
lenge, while immunization with the DENV-3 prM/E based vaccine strain as a genetic backbone. Chimeric YF 17D/DEN viruses were
resulted in only partial protection. Neutralizing antibodies against constructed by replacing the YFV prM/E genes with those of DENV
DENV-3 E85 were serotype-specific and largely directed against [106]. Monovalent dengue vaccine candidates for different DENV
EDIII epitopes. Immunogenicity and protective efficacy studies of a serotypes were generated and evaluated in NHPs. In attenua-
tetravalent E85 based vaccine candidate in NHPs are in progress. tion studies, the monovalent vaccines produced only low levels
Another approach taken at the University of Texas Medical of viremia and showed reduced neurotropism compared to the
Branch (UTMB) is based on a single-cycle, capsid-gene deleted West YF 17D vaccine. Neutralizing antibody responses and protection
Nile virus vaccine vector [105]. Infectious single-cycle WNV par- against viral challenge were also shown [48–50]. DEN viruses used
ticles are produced by packaging technologies, which supply the for the construction of chimeric YF 17D/DEN viruses in this vac-
capsid protein in trans. Infected target cells are unable to produce cine project include strains isolated from Latin American patients,
viral progeny, but the single replication cycle results in efficient while the DENV strains that provided the basis for a tetravalent
production of antigen including secretion of VLPs from infected dengue vaccine candidate developed by Sanofi Pasteur, which is
cells. A DENV-2 vaccine candidate was generated by replacing the currently in advanced clinical evaluation [44–47,107], were iso-
prM/E genes of the WNV vaccine vector with the prM/E genes of lated from Asian patients. Different sets of chimeric YF 17D/DEN
DENV-2 [87]. The vaccine was tested for potency and efficacy in a viruses are thus available for the development of a tetravalent live
mouse model of dengue pathogenesis (AG129 mice [97]). A single attenuated dengue vaccine.
intraperitoneal immunization of mice produced a dose-dependent Another live attenuated vaccine candidate has been devel-
DENV-2 neutralizing antibody response and resulted in a protective oped in collaboration between Chiang Mai University, Mahidol
effect against viral challenge. University, the Thai National Science and Technology Develop-
A virus-vectored dengue vaccine candidate developed by ment Agency (NSTDA) and BioNet-Asia [51,52]. DEN/DEN chimeric
Themis Bioscience and Institut Pasteur is based on expression of viruses were constructed which contain the prM/E coding region of
a single tetravalent DENV antigen construct from a live attenuated recent dengue clinical isolates in the genetic background of atten-
measles virus vaccine vector [88]. This vaccine vector technology uated DENV. Monovalent vaccine candidates have been evaluated
allows for integration of large antigen inserts and has been shown for neurovirulence and immunogenicity in mice. Safety and efficacy
to induce strong neutralizing antibodies and cellular immune studies in NHPs are planned.
responses even in the presence of pre-existing immunity to measles
virus. The dengue vaccine candidate expresses a construct contain- 3.7. Heterologous prime-boost approaches
ing the EDIII domains of DENV-1-4 as well as the DENV-1 M protein
ectodomain (ectoM). The single component tetravalent vaccine- A heterologous prime-boost approach developed by the NMRC
induced neutralizing antibodies against all DENV serotypes in mice. uses a DNA vaccine candidate and a virus-vectored vaccine candi-
Inclusion of ectoM in the vaccine construct was found to provide an date [89]. The monovalent DNA vaccine, which has been evaluated
adjuvant activity. Following further improvements of the vaccine in a phase 1 clinical trial, consists of a plasmid vector expressing
construct, immunogenicity and efficacy studies have been initiated DENV-1 prM/E and is administered intramuscularly with a needle-
in NHPs. Establishment of a GMP process and initiation of clinical free Biojector system [82]. The virus-vectored vaccine expresses
trials are planned in 2011 and 2012, respectively. the same DENV-1 prM/E sequence from a single-cycle VEE virus
vector. A three-dose prime-boost regimen was evaluated in NHPs,
3.5. Purified inactivated virus vaccines which consists of two doses of the DNA vaccine at days 0 and 28
followed by intramuscular injection of the virus-vectored vaccine
A purified psoralen-inactivated DENV-1 vaccine candidate has at day 117. When compared to homologous three dose schedules
been developed by the NMRC [56]. Following immunogenicity of either the DNA or the virus-vectored vaccines, the heterologous
studies in mice, the monovalent candidate adjuvanted in alum was prime-boost approach was found to improve neutralizing antibody
evaluated in NHPs using a three-dose schedule (intradermal injec- responses and protection against viral challenge [89].
tion, biweekly intervals). Immunized monkeys showed a DENV-1 Another heterologous prime-boost approach developed by the
neutralizing antibody response and reduced duration of viremia NMRC and the WRAIR uses non-replicating vaccines for priming,
upon viral challenge on day 132, suggesting that the vaccine can- followed by boosting with a LAV vaccine candidate [90]. Two non-
didate provides partial protection in NHPs. replicating vaccines were evaluated: a tetravalent DNA vaccine
PIV vaccine candidates are also being developed by Glaxo- composed of plasmid vectors expressing DENV prM/E [82] and a
SmithKline (GSK) in collaboration with the Walter Reed Army tetravalent PIV vaccine consisting of formalin-inactivated DENV
Institute of Research (WRAIR) and the Oswaldo Cruz Foundation adjuvanted with alum [55]. The tetravalent LAV vaccine candidate,
(FIOCRUZ) [55]. Different approaches for virus inactivation and which was developed by GSK and WRAIR using serial passage of
purification and various adjuvant systems are currently under clinical DENV isolates in PDK cells, has reached phase 2 clinical trials
evaluation. A pilot study was carried out in NHPs to test a tetrava- [42]. To determine which of the two non-replicating vaccine candi-
7282 J. Schmitz et al. / Vaccine 29 (2011) 7276–7284

dates was more effective for priming, a DNA-DNA-LAV vaccination Drs. Martin Bachmann (Cytos Biotechnology), Gwong-Jen Chang
schedule (at -30, 0 and 60 days) was compared to a PIV-LAV vac- (CDC), John Dong (GenPhar), Bruce Innis (GSK), Niranjan Y. Sardesai
cination schedule (at 0 and 60 days) in NHPs. Both regimens were (Inovio Pharmaceuticals) and David B. Weiner (University of Penn-
found to elicit neutralizing antibody responses. However, priming sylvania), Alan Shaw (VaxInnate), Sathyamangalam Swaminathan
with the DNA vaccine provided only partial protection against viral (ICGEB), Erich Tauber (Themis Bioscience) and Laura White (UNC).
challenge 6 months after the last dose, as evidenced by the obser- Financial support of the consultation from the Bill and Melinda
vation of breakthrough viremia. By contrast, priming with one dose Gates Foundation is kindly acknowledged.
of PIV vaccine followed by a LAV vaccine booster dose two months
later resulted in complete protection against challenge with any of
the four DENV serotypes [90]. References
While heterologous prime-boost strategies may offer advan-
tages over other vaccination approaches, recent investigations with [1] Gubler DJ. Epidemic dengue/dengue hemorrhagic fever as a public health,
social and economic problem in the 21st century. Trends Microbiol
West Nile virus vaccine candidates suggest that the order of anti-
2002;10:100–3.
gens used in a heterologous prime-boost approach was important [2] Suaya JA, Shepard DS, Siqueira JB, Martelli CT, Lum LC, Tan LH, et al. Cost of
in terms of the specificity and magnitude of the immune response dengue cases in eight countries in the Americas and Asia: a prospective study.
[108]. Similar results can be expected for dengue vaccines. Am J Trop Med Hyg 2009;80:846–55.
[3] World Health Organization. Dengue guidelines for diagnosis, treatment,
prevention and control. Geneva: World Health Organization; 2009. Available
at: http://whqlibdoc.who.int/publications/2009/9789241547871 eng.pdf
4. Conclusions [accessed 21 June 2011].
[4] Shepard DS, Coudeville L, Halasa YA, Zambrano B, Dayan GH. Economic impact
Efforts for dengue vaccine development have faced multiple of dengue illness in the Americas. Am J Trop Med Hyg 2011;84:200–7.
[5] Whitehorn J, Farrar J. Dengue. Br Med Bull 2010;95:161–73.
challenges, including the need to induce a balanced and last- [6] Durbin AP, Whitehead SS. Dengue vaccine candidates in development. Curr
ing immunity against four DENV serotypes, uncertainties around Top Microbiol Immunol 2010;338:129–43.
immune correlates of protection, potential immune enhancement [7] Swaminathan S, Batra G, Khanna N. Dengue vaccines: state of the art. Expert
Opin Ther Pat 2010;20:819–35.
of disease, and lack of suitable animal model. However, consider- [8] Guzman MG. Dengue vaccines: new developments. Drugs Future
able progress has been made in recent years, which has resulted in 2011;36:45–62.
an advanced dengue vaccine pipeline, with a lead candidate enter- [9] Murphy BR, Whitehead SS. Immune response to dengue virus and prospects
for a vaccine. Annu Rev Immunol 2011;29:587–619.
ing phase 3 clinical trials [44–47] and several other candidates in
[10] Halstead SB. Dengue. Lancet 2007;370:1644–52.
earlier stages of clinical evaluation [36–43,65,82]. The majority of [11] Hombach J, Cardosa MJ, Sabchareon A, Vaughn DW, Barrett AD. Scientific
dengue vaccine candidates currently in clinical development are consultation on immunological correlates of protection induced by dengue
vaccines. Vaccine 2007;25:4130–9.
based on the same immunization regimen (namely a tetravalent
[12] Thomas SJ, Hombach J, Barrett A. Scientific consultation on cell medi-
LAV) derived using different technologies. However, some uncer- ated immunity (CMI) in dengue and dengue vaccine development. Vaccine
tainties around the utilization of LAVs remain, including potential 2009;27:355–68.
imbalances in immunity due to immune interference. [13] Halstead SB. Neutralization and antibody-dependent enhancement of dengue
viruses. Adv Virus Res 2003;60:421–67.
A large number of diverse dengue vaccine candidates are in [14] Halstead SB. Antibodies determine virulence in dengue. Ann N Y Acad Sci
preclinical development. These preclinical candidates ensure a con- 2009;1171(Suppl. 1):E48–56.
tinued influx of innovation into the vaccine pipeline, which is [15] Pierson TC, Fremont DH, Kuhn RJ, Diamond MS. Structural insights into
the mechanisms of antibody-mediated neutralization of flavivirus infection:
critical for maximizing the chances of success for dengue vac- implications for vaccine development. Cell Host Microbe 2008;4:229–38.
cine development. One possible scenario is that a first generation [16] Dowd KA, Pierson TC. Antibody-mediated neutralization of flaviviruses: a
of licensed dengue vaccines will arise from candidates currently reductionist view. Virology 2011;411:306–15.
[17] Junjhon J, Edwards TJ, Utaipat U, Bowman VD, Holdaway HA, Zhang W, et al.
in clinical development, while some of the current preclinical Influence of pr-M cleavage on the heterogeneity of extracellular dengue virus
stage candidates could become next generation dengue vaccines particles. J Virol 2010;84:8353–8.
licensed at a later stage. Next generation dengue vaccines could [18] Perera R, Kuhn RJ. Structural proteomics of dengue virus. Curr Opin Microbiol
2008;11:369–77.
serve to complement existing vaccines. For example, prime-boost
[19] Beltramello M, Williams KL, Simmons CP, Macagno A, Simonelli L, Quyen NT,
approaches combining live and non-replicating vaccines may allow et al. The human immune response to dengue virus is dominated by highly
for balancing the relative advantages and shortcomings of these cross-reactive antibodies endowed with neutralizing and enhancing activity.
Cell Host Microbe 2010;8:271–83.
technologies. Furthermore, next generation dengue vaccines could
[20] Crill WD, Roehrig JT. Monoclonal antibodies that bind to domain III of dengue
have a superior product profile compared to existing vaccines, virus E glycoprotein are the most efficient blockers of virus adsorption to Vero
including efficacy, dose-scheduling, and stability. There are indi- cells. J Virol 2001;75:7769–73.
cations that candidates which are currently in advanced clinical [21] Roehrig JT. Antigenic structure of flavivirus proteins. Adv Virus Res
2003;59:141–75.
development may require long, multi-dose immunization sched- [22] Brien JD, Austin SK, Sukupolvi-Petty S, O’Brien KM, Johnson S, Fremont DH,
ules (e.g. three doses over a 12 month period). Next generation et al. Genotype-specific neutralization and protection by antibodies against
dengue vaccines could have more compressed schedules requir- dengue virus type 3. J Virol 2010;84:10630–43.
[23] Dejnirattisai W, Jumnainsong A, Onsirisakul N, Fitton P, Vasanawathana S,
ing fewer doses, or characteristics that make them more suitable Limpitikul W, et al. Cross-reacting antibodies enhance dengue virus infection
for certain target groups (e.g. particular age groups, immunocom- in humans. Science 2010;328:745–8.
promised individuals, travelers). Finally, next generation dengue [24] Rodenhuis-Zybert IA, van der Schaar HM, da Silva Voorham JM, van der
Ende-Metselaar H, Lei HY, Wilschut J, et al. Immature dengue virus: a veiled
vaccines could generate much-welcomed competition, leading to pathogen? PLoS Pathog 2010;6:e1000718.
more affordable and cost-effective vaccines, which would facilitate [25] Shrestha B, Brien JD, Sukupolvi-Petty S, Austin SK, Edeling MA, Kim T, et al. The
their use particularly in endemic developing countries. development of therapeutic antibodies that neutralize homologous and het-
erologous genotypes of dengue virus type 1. PLoS Pathog 2010;6:e1000823.
[26] Sukupolvi-Petty S, Austin SK, Engle M, Brien JD, Dowd KA, Williams KL, et al.
Structure and function analysis of therapeutic monoclonal antibodies against
Acknowledgements
dengue virus type 2. J Virol 2010;84:9227–39.
[27] Wahala WM, Donaldson EF, de Alwis R, Accavitti-Loper MA, Baric RS, de Silva
Participants of the WHO/IVI Informal Consultation on Next Gen- AM. Natural strain variation and antibody neutralization of dengue serotype
eration Dengue Vaccines and Diagnostics (1–2 November 2010, 3 viruses. PLoS Pathog 2010;6:e1000821.
[28] Cassetti MC, Durbin A, Harris E, Rico-Hesse R, Roehrig J, Rothman A,
Atlanta) are thanked for sharing unpublished data. Preclinical stage et al. Report of an NIAID workshop on dengue animal models. Vaccine
dengue vaccine candidates were presented at the consultation by 2010;28:4229–34.
J. Schmitz et al. / Vaccine 29 (2011) 7276–7284 7283

[29] Williams KL, Zompi S, Beatty PR, Harris E. A mouse model for studying dengue [57] Simmons M, Murphy GS, Hayes CG. Short report: antibody responses of mice
virus pathogenesis and immune response. Ann N Y Acad Sci 2009;1171(Suppl. immunized with a tetravalent dengue recombinant protein subunit vaccine.
1):E12–23. Am J Trop Med Hyg 2001;65:159–61.
[30] Balsitis SJ, Williams KL, Lachica R, Flores D, Kyle JL, Mehlhop E, et al. Lethal [58] Bernardo L, Izquierdo A, Alvarez M, Rosario D, Prado I, Lopez C, et al. Immuno-
antibody enhancement of dengue disease in mice is prevented by Fc modifi- genicity and protective efficacy of a recombinant fusion protein containing
cation. PLoS Pathog 2010;6:e1000790. the domain III of the dengue 1 envelope protein in non-human primates.
[31] Zellweger RM, Prestwood TR, Shresta S. Enhanced infection of liver sinu- Antiviral Res 2008;80:194–9.
soidal endothelial cells in a mouse model of antibody-induced severe dengue [59] Etemad B, Batra G, Raut R, Dahiya S, Khanam S, Swaminathan S, et al. An
disease. Cell Host Microbe 2010;7:128–39. envelope domain III-based chimeric antigen produced in Pichia pastoris elicits
[32] Kuruvilla JG, Troyer RM, Devi S, Akkina R. Dengue virus infection and immune neutralizing antibodies against all four dengue virus serotypes. Am J Trop Med
response in humanized RAG2(−/−)gamma(c)(−/−) (RAG-hu) mice. Virology Hyg 2008;79:353–63.
2007;369:143–52. [60] Chen HW, Liu SJ, Liu HH, Kwok Y, Lin CL, Lin LH, et al. A novel technology for
[33] Mota J, Rico-Hesse R. Humanized mice show clinical signs of dengue fever the production of a heterologous lipoprotein immunogen in high yield has
according to infecting virus genotype. J Virol 2009;83:8638–45. implications for the field of vaccine design. Vaccine 2009;27:1400–9.
[34] Lauring AS, Jones JO, Andino R. Rationalizing the development of live attenu- [61] Leng CH, Liu SJ, Tsai JP, Li YS, Chen MY, Liu HH, et al. A novel dengue vaccine
ated virus vaccines. Nat Biotechnol 2010;28:573–9. candidate that induces cross-neutralizing antibodies and memory immunity.
[35] Bhamarapravati N, Sutee Y. Live attenuated tetravalent dengue vaccine. Vac- Microbes Infect 2009;11:288–95.
cine 2000;18(Suppl. 2):44–7. [62] Valdes I, Bernardo L, Gil L, Pavon A, Lazo L, Lopez C, et al. A novel fusion protein
[36] Huang CY, Butrapet S, Tsuchiya KR, Bhamarapravati N, Gubler DJ, Kinney RM. domain III-capsid from dengue-2, in a highly aggregated form, induces a func-
Dengue 2 PDK-53 virus as a chimeric carrier for tetravalent dengue vaccine tional immune response and protection in mice. Virology 2009;394:249–58.
development. J Virol 2003;77:11436–47. [63] Valdes I, Hermida L, Martin J, Menendez T, Gil L, Lazo L, et al. Immunologi-
[37] Durbin AP, Whitehead SS, McArthur J, Perreault JR, Blaney Jr JE, Thumar B, et al. cal evaluation in nonhuman primates of formulations based on the chimeric
rDEN4delta30, a live attenuated dengue virus type 4 vaccine candidate, is safe, protein P64k-domain III of dengue 2 and two components of Neisseria menin-
immunogenic, and highly infectious in healthy adult volunteers. J Infect Dis gitidis. Vaccine 2009;27:995–1001.
2005;191:710–8. [64] Valdes I, Gil L, Romero Y, Castro J, Puente P, Lazo L, et al. The chimeric protein
[38] Durbin AP, McArthur J, Marron JA, Blaney Jr JE, Thumar B, Wanionek K, domain III-capsid of dengue virus serotype 2 (DEN-2) successfully boosts neu-
et al. The live attenuated dengue serotype 1 vaccine rDEN1Delta30 is tralizing antibodies generated in monkeys upon infection with DEN-2. Clin
safe and highly immunogenic in healthy adult volunteers. Hum Vaccin Vaccine Immunol 2011;18:455–9.
2006;2:167–73. [65] Clements DE, Coller BA, Lieberman MM, Ogata S, Wang G, Harada KE, et al.
[39] Durbin AP, McArthur JH, Marron JA, Blaney JE, Thumar B, Wanionek K, et al. Development of a recombinant tetravalent dengue virus vaccine: immuno-
rDEN2/4Delta30(ME), a live attenuated chimeric dengue serotype 2 vaccine genicity and efficacy studies in mice and monkeys. Vaccine 2010;28:2705–15.
is safe and highly immunogenic in healthy dengue-naive adults. Hum Vaccin [66] Jennings GT, Bachmann MF. The coming of age of virus-like particle vaccines.
2006;2:255–60. Biol Chem 2008;389:521–36.
[40] McArthur JH, Durbin AP, Marron JA, Wanionek KA, Thumar B, Pierro DJ, [67] Bisht H, Chugh DA, Raje M, Swaminathan SS, Khanna N. Recombinant
et al. Phase I clinical evaluation of rDEN4Delta30-200,201: a live attenuated dengue virus type 2 envelope/hepatitis B surface antigen hybrid protein
dengue 4 vaccine candidate designed for decreased hepatotoxicity. Am J Trop expressed in Pichia pastoris can function as a bivalent immunogen. J Biotech-
Med Hyg 2008;79:678–84. nol 2002;99:97–110.
[41] Simasathien S, Thomas SJ, Watanaveeradej V, Nisalak A, Barberousse C, Innis [68] Amexis G, Young NS. Parvovirus B19 empty capsids as antigen carriers
BL, et al. Safety and immunogenicity of a tetravalent live-attenuated dengue for presentation of antigenic determinants of dengue 2 virus. J Infect Dis
vaccine in flavivirus naive children. Am J Trop Med Hyg 2008;78:426–33. 2006;194:790–4.
[42] Sun W, Cunningham D, Wasserman SS, Perry J, Putnak JR, Eckels KH, et al. [69] Kuwahara M, Konishi E. Evaluation of extracellular subviral particles of
Phase 2 clinical trial of three formulations of tetravalent live-attenuated dengue virus type 2 and Japanese encephalitis virus produced by Spodoptera
dengue vaccine in flavivirus-naive adults. Hum Vaccin 2009;5:33–40. frugiperda cells for use as vaccine and diagnostic antigens. Clin Vaccine
[43] Wright PF, Durbin AP, Whitehead SS, Ikizler MR, Henderson S, Blaney JE, et al. Immunol 2010;17:1560–6.
Phase 1 trial of the dengue virus type 4 vaccine candidate rDEN4Delta30-4995 [70] Liu W, Jiang H, Zhou J, Yang X, Tang Y, Fang D, et al. Recombinant dengue virus-
in healthy adult volunteers. Am J Trop Med Hyg 2009;81:834–41. like particles from Pichia pastoris: efficient production and immunological
[44] Guy B, Guirakhoo F, Barban V, Higgs S, Monath TP, Lang J. Preclinical and properties. Virus Genes 2010;40:53–9.
clinical development of YFV 17D-based chimeric vaccines against dengue, [71] Kutzler MA, Weiner DB. DNA vaccines: ready for prime time? Nat Rev Genet
West Nile and Japanese encephalitis viruses. Vaccine 2010;28:632–49. 2008;9:776–88.
[45] Morrison D, Legg TJ, Billings CW, Forrat R, Yoksan S, Lang J. A novel tetravalent [72] Chang GJ, Hunt AR, Holmes DA, Springfield T, Chiueh TS, Roehrig JT, et al.
dengue vaccine is well tolerated and immunogenic against all 4 serotypes in Enhancing biosynthesis and secretion of premembrane and envelope proteins
flavivirus-naive adults. J Infect Dis 2010;201:370–7. by the chimeric plasmid of dengue virus type 2 and Japanese encephalitis
[46] Guy B, Almond J, Lang J. Dengue vaccine prospects: a step forward. Lancet virus. Virology 2003;306:170–80.
2011;377:381–2. [73] Wu SF, Liao CL, Lin YL, Yeh CT, Chen LK, Huang YF, et al. Evaluation of protective
[47] Poo J, Galan F, Forrat R, Zambrano B, Lang J, Dayan GH. Live-attenuated tetrava- efficacy and immune mechanisms of using a non-structural protein NS1 in
lent dengue vaccine in dengue-naive children, adolescents, and adults in DNA vaccine against dengue 2 virus in mice. Vaccine 2003;21:3919–29.
Mexico City: randomized controlled phase 1 trial of safety and immunogenic- [74] Mota J, Acosta M, Argotte R, Figueroa R, Mendez A, Ramos C. Induction of
ity. Pediatr Infect Dis J 2011;30:E9–17. protective antibodies against dengue virus by tetravalent DNA immunization
[48] Galler R, Marchevsky RS, Caride E, Almeida LF, Yamamura AM, Jabor AV, et al. of mice with domain III of the envelope protein. Vaccine 2005;23:3469–76.
Attenuation and immunogenicity of recombinant yellow fever 17D-dengue [75] Purdy DE, Chang GJ. Secretion of noninfectious dengue virus-like particles
type 2 virus for rhesus monkeys. Braz J Med Biol Res 2005;38:1835–46. and identification of amino acids in the stem region involved in intracellular
[49] Mateu GP, Marchevsky RS, Liprandi F, Bonaldo MC, Coutinho ES, Dieudonne retention of envelope protein. Virology 2005;333:239–50.
M, et al. Construction and biological properties of yellow fever 17D/dengue [76] Raviprakash K, Apt D, Brinkman A, Skinner C, Yang S, Dawes G, et al. A chimeric
type 1 recombinant virus. Trans R Soc Trop Med Hyg 2007;101:289–98. tetravalent dengue DNA vaccine elicits neutralizing antibody to all four virus
[50] Trindade GF, Marchevsky RS, Fillipis AM, Nogueira RM, Bonaldo MC, Acero PC, serotypes in rhesus macaques. Virology 2006;353:166–73.
et al. Limited replication of yellow fever 17DD and 17D-Dengue recombinant [77] Costa SM, Azevedo AS, Paes MV, Sarges FS, Freire MS, Alves AM. DNA vaccines
viruses in rhesus monkeys. An Acad Bras Cienc 2008;80:311–21. against dengue virus based on the ns1 gene: the influence of different sig-
[51] BioNet-Asia. BioNet-Asia to enter development of a dengue vaccine. nal sequences on the protein expression and its correlation to the immune
Press release 2011. Available at: http://www.bionet-asia.com/feb2111.html response elicited in mice. Virology 2007;358:413–23.
[accessed 21 June 2011]. [78] Imoto J, Konishi E. Dengue tetravalent DNA vaccine increases its immuno-
[52] National Science and Technology Development Agency. Chimeric dengue vac- genicity in mice when mixed with a dengue type 2 subunit vaccine or an
cine licensing agreement. Press release 2011. Available at: http://www.nstda. inactivated Japanese encephalitis vaccine. Vaccine 2007;25:1076–84.
or.th/eng/index.php/research-themes/health-and-medicine/item/177-chim [79] De Paula SO, Lima DM, de Oliveira Franca RF, Gomes-Ruiz AC, da Fonseca BA. A
eric-dengue-vaccine-licensing-agreement [accessed 21 June 2011]. DNA vaccine candidate expressing dengue-3 virus prM and E proteins elicits
[53] Smith KM, Nanda K, Spears CJ, Ribeiro M, Vancini R, Piper A, et al. Struc- neutralizing antibodies and protects mice against lethal challenge. Arch Virol
tural mutants of dengue virus 2 transmembrane domains exhibit host-range 2008;153:2215–23.
phenotype. Virol J 2011;8:289. [80] Ramanathan MP, Kuo YC, Selling BH, Li Q, Sardesai NY, Kim JJ, et al. Develop-
[54] Leroux-Roels G. Unmet needs in modern vaccinology: adjuvants to improve ment of a novel DNA SynCon tetravalent dengue vaccine that elicits immune
the immune response. Vaccine 2010;28(Suppl. 3):C25–36. responses against four serotypes. Vaccine 2009;27:6444–53.
[55] Putnak JR, Coller BA, Voss G, Vaughn DW, Clements D, Peters I, et al. An evalu- [81] Lima DM, de Paula SO, Franca RF, Palma PV, Morais FR, Gomes-Ruiz AC, et al. A
ation of dengue type-2 inactivated, recombinant subunit, and live-attenuated DNA vaccine candidate encoding the structural prM/E proteins elicits a strong
vaccine candidates in the rhesus macaque model. Vaccine 2005;23: immune response and protects mice against dengue-4 virus infection. Vaccine
4442–52. 2011;29:831–8.
[56] Maves RC, Ore RM, Porter KR, Kochel TJ. Immunogenicity and protective effi- [82] Beckett CG, Tjaden J, Burgess T, Danko JR, Tamminga C, Simmons M, et al.
cacy of a psoralen-inactivated dengue-1 virus vaccine candidate in Aotus Evaluation of a prototype dengue-1 DNA vaccine in a Phase 1 clinical trial.
nancymaae monkeys. Vaccine 2011;29:2691–6. Vaccine 2011;29:960–8.
7284 J. Schmitz et al. / Vaccine 29 (2011) 7276–7284

[83] Liniger M, Zuniga A, Naim HY. Use of viral vectors for the development of [95] Konishi E, Kosugi S, Imoto J. Dengue tetravalent DNA vaccine inducing neutral-
vaccines. Expert Rev Vaccines 2007;6:255–66. izing antibody and anamnestic responses to four serotypes in mice. Vaccine
[84] Raviprakash K, Wang D, Ewing D, Holman DH, Block K, Woraratanadharm 2006;24:2200–7.
J, et al. A tetravalent dengue vaccine based on a complex adenovirus vector [96] Crill WD, Hughes HR, Delorey MJ, Chang GJ. Humoral immune responses of
provides significant protection in rhesus monkeys against all four serotypes dengue fever patients using epitope-specific serotype-2 virus-like particle
of dengue virus. J Virol 2008;82:6927–34. antigens. PLoS One 2009;4:e4991.
[85] Khanam S, Pilankatta R, Khanna N, Swaminathan S. An adenovirus type 5 [97] Johnson AJ, Roehrig JT. New mouse model for dengue virus vaccine testing. J
(AdV5) vector encoding an envelope domain III-based tetravalent antigen Virol 1999;73:783–6.
elicits immune responses against all four dengue viruses in the presence of [98] Spohn G, Keller I, Beck M, Grest P, Jennings GT, Bachmann MF. Active immu-
prior AdV5 immunity. Vaccine 2009;27:6011–21. nization with IL-1 displayed on virus-like particles protects from autoimmune
[86] White LJ, Parsons MM, Whitmore AC, Williams BM, de Silva A, Johnston RE. arthritis. Eur J Immunol 2008;38:877–87.
An immunogenic and protective alphavirus replicon particle-based dengue [99] Spohn G, Jennings GT, Martina BE, Keller I, Beck M, Pumpens P, et al. A VLP-
vaccine overcomes maternal antibody interference in weanling mice. J Virol based vaccine targeting domain III of the West Nile virus E protein protects
2007;81:10329–39. from lethal infection in mice. Virol J 2010;7:146.
[87] Suzuki R, Winkelmann ER, Mason PW. Construction and characterization of a [100] Jegerlehner A, Maurer P, Bessa J, Hinton HJ, Kopf M, Bachmann MF. TLR9 sig-
single-cycle chimeric flavivirus vaccine candidate that protects mice against naling in B cells determines class switch recombination to IgG2a. J Immunol
lethal challenge with dengue virus type 2. J Virol 2009;83:1870–80. 2007;178:2415–20.
[88] Brandler S, Ruffie C, Najburg V, Frenkiel MP, Bedouelle H, Despres P, et al. [101] Bessa J, Jegerlehner A, Hinton HJ, Pumpens P, Saudan P, Schneider P, et al.
Pediatric measles vaccine expressing a dengue tetravalent antigen elicits Alveolar macrophages and lung dendritic cells sense RNA and drive mucosal
neutralizing antibodies against all four dengue viruses. Vaccine 2010;28: IgA responses. J Immunol 2009;183:3788–99.
6730–9. [102] Gurramkonda C, Adnan A, Gabel T, Lunsdorf H, Ross A, Nemani SK, et al.
[89] Chen L, Ewing D, Subramanian H, Block K, Rayner J, Alterson KD, et al. A Simple high-cell density fed-batch technique for high-level recombinant pro-
heterologous DNA prime-Venezuelan equine encephalitis virus replicon par- tein production with Pichia pastoris: application to intracellular production
ticle boost dengue vaccine regimen affords complete protection from virus of Hepatitis B surface antigen. Microb Cell Fact 2009;8:13.
challenge in cynomolgus macaques. J Virol 2007;81:11634–9. [103] Konishi E, Fujii A. Dengue type 2 virus subviral extracellular particles pro-
[90] Simmons M, Burgess T, Lynch J, Putnak R. Protection against dengue virus by duced by a stably transfected mammalian cell line and their evaluation for a
non-replicating and live attenuated vaccines used together in a prime boost subunit vaccine. Vaccine 2002;20:1058–67.
vaccination strategy. Virology 2010;396:280–8. [104] Leopold PL, Wendland RL, Vincent T, Crystal RG. Neutralized adenovirus-
[91] Hermida L, Bernardo L, Martin J, Alvarez M, Prado I, Lopez C, et al. A recom- immune complexes can mediate effective gene transfer via an Fc
binant fusion protein containing the domain III of the dengue-2 envelope receptor-dependent infection pathway. J Virol 2006;80:10237–47.
protein is immunogenic and protective in nonhuman primates. Vaccine [105] Widman DG, Frolov I, Mason PW. Third-generation flavivirus vaccines
2006;24:3165–71. based on single-cycle, encapsidation-defective viruses. Adv Virus Res
[92] Valdes I, Hermida L, Gil L, Lazo L, Castro J, Martin J, et al. Heterologous prime- 2008;72:77–126.
boost strategy in non-human primates combining the infective dengue virus [106] Caufour PS, Motta MC, Yamamura AM, Vazquez S, Ferreira II, Jabor AV, et al.
and a recombinant protein in a formulation suitable for human use. Int J Infect Construction, characterization and immunogenicity of recombinant yellow
Dis 2010;14:e377–83. fever 17D-dengue type 2 viruses. Virus Res 2001;79:1–14.
[93] McDonald WF, Huleatt JW, Foellmer HG, Hewitt D, Tang J, Desai P, et al. A West [107] Guirakhoo F, Weltzin R, Chambers TJ, Zhang ZX, Soike K, Ratterree
Nile virus recombinant protein vaccine that coactivates innate and adaptive M, et al. Recombinant chimeric yellow fever-dengue type 2 virus is
immunity. J Infect Dis 2007;195:1607–17. immunogenic and protective in nonhuman primates. J Virol 2000;74:
[94] Leng CH, Chen HW, Chang LS, Liu HH, Liu HY, Sher YP, et al. A recombinant 5477–85.
lipoprotein containing an unsaturated fatty acid activates NF-kappaB through [108] Zlatkovic J, Stiasny K, Heinz FX. Immunodominance and functional activities
the TLR2 signaling pathway and induces a differential gene profile from a of antibody responses to inactivated West Nile virus and recombinant subunit
synthetic lipopeptide. Mol Immunol 2010;47:2015–21. vaccines in mice. J Virol 2011;85:1994–2003.

You might also like