You are on page 1of 10

Review

Progress towards a dengue vaccine


Daniel P Webster, Jeremy Farrar, Sarah Rowland-Jones

Lancet Infect Dis 2009; 9: 678–87 The spread of dengue virus throughout the tropics represents a major, rapidly growing public health problem with
See Newsdesk page 662 an estimated 2·5 billion people at risk of dengue fever and the life-threatening disease, severe dengue. A safe and
Department of Infectious effective vaccine for dengue is urgently needed. The pathogenesis of severe dengue results from a complex
Diseases and Microbiology, interaction between the virus, the host, and, at least in part, immune-mediated mechanisms. Vaccine development
John Radcliffe Hospital, Oxford,
has been slowed by fears that immunisation might predispose individuals to the severe form of dengue infection.
UK (D P Webster FRCPath);
Hospital for Tropical Diseases, A pipeline of candidate vaccines now exists, including live attenuated, inactivated, chimeric, DNA, and viral-vector
Wellcome Trust Major Overseas vaccines, some of which are at the stage of clinical testing. In this Review, we present what is understood about
Programme, Oxford University dengue pathogenesis and its implications for vaccine design, the progress that is being made in the development
Clinical Research Unit, Ho Chi
of a vaccine, and the future challenges.
Minh City, Vietnam
(Prof J Farrar FRCP); and Human
Immunology Unit, The Introduction genus.13 Aedes aegypti, which exploits peridomestic water
Weatherall Institute of Dengue virus infection represents a major, growing public containers as larval habitats, is by far the predominant
Molecular Medicine, University
health problem with an estimated 2·5 billion people at risk vector of dengue, although Aedes albopictus can also
of Oxford, Oxford, UK
(Prof S Rowland-Jones FRCP) of infection in tropical and subtropical countries, mainly sustain transmission.14 A aegypti are highly adapted to
Correspondence to: southeast and south Asia, Central and South America, and urban environments, and uncontrolled urbanisation is
Dr Daniel P Webster, Department the Caribbean.1 There are few data on the incidence of the rarely accompanied by investment in basic infrastructure
of Infectious Diseases and disease available from Africa. This growth is largely and public health measures,15 particularly in Asia and
Microbiology, John Radcliffe
explained by increases in human population, rapid Latin America. Containers used for water storage are
Hospital, Oxford OX3 9DU, UK
danwebster@doctors.org.uk urbanisation, the continued challenge of vector control, important aedes breeding sites alongside building
and international travel.2 There are about 50 million to projects, discarded plastics, and increasing numbers of
100 million cases of dengue infection each year, with an unused tyres.13 This increase in available breeding sites,
estimated 500 000 cases of life-threatening disease in the along with daytime feeding and resting in sheltered dark
form of severe dengue—including dengue haemorrhagic spaces in houses, makes aedes an efficient vector.13
fever and dengue shock syndrome.3,4 Children have Although urgently needed, a licensed vaccine for
historically borne the brunt of the disease burden and dengue is not yet available. Control of dengue by
severe dengue has been a leading cause of admission to widespread vaccination has been a priority of WHO for
hospital and death of children in several southeast Asian three decades.16 Vaccines are available for related viruses
countries.2 The age profile of dengue is changing in Asia such as yellow fever, Japanese encephalitis virus, and
and in Latin America, however, and adults are increasingly tick-borne encephalitis virus.17 In this Review, we aim to
being affected.5 Dengue is associated with major urban present an up-to-date understanding of dengue
epidemics that have substantial economic, political, and pathogenesis, how this understanding is influencing the
social effects,6 such as those seen in Delhi (1996),7 development of vaccines, and what progress has been
Cuba (1977–79 and 1997),8 Taiwan (2002),9 and Brazil (2008).5 made in developing a safe and effective dengue vaccine.
Dengue is now also a leading cause of morbidity in
American and European travellers and military Clinical features
personnel.10 Dengue viruses cause a range of well-described clinical
There are four serotypes of dengue virus (designated illnesses.18–20 Infection ranges from an asymptomatic
DENV1–4) belonging to the Flavivirus genus of the infection to a self-limiting febrile illness, dengue fever, to
Flaviviridae family. The dengue viruses fall into a distinct severe dengue, a clinical syndrome that typically presents
group among the flaviviruses, showing a phylogenetic with capillary permeability and can lead to dengue shock
association with the Japanese encephalitis virus group syndrome and dengue haemorrhagic fever. Less common
and more distantly with yellow fever, tick-borne features of severe dengue are encephalitis, hepatitis, and
encephalitis, and hepatitis C viruses.11 The dengue virion renal dysfunction. The WHO classification scheme for
is spherical and 40–50 nm in diameter. The nucleocapsid dengue was developed in the 1970s and is at present
is covered by a lipid bilayer envelope containing the under review because of concerns that it might lead to
envelope and membrane proteins. The genome is single- under-reporting of severe dengue.18,21 What defines the
stranded positive-sense RNA of about 11 kb in length that clinical outcome of dengue infection is not completely
encodes for three structural proteins (the capsid protein, understood, although numerous hypotheses exist.
the membrane protein precursor, and the envelope Epidemiological observations from studies in Thailand
protein) and seven non-structural proteins (NS1, NS2A, in the 1960s showed that greater than 85% of cases of
NS2B, NS3, NS4A, NS4B, and NS5).12 severe dengue happened in people with pre-existing
Dengue viruses are transmitted to human beings heterotypic dengue virus antibodies, and this has led to
through the bite of infected mosquitoes of the Aedes research into dengue pathogenesis focusing on the

678 www.thelancet.com/infection Vol 9 November 2009


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Review

immune response.22 Additional evidence from infant subneutralising levels of dengue antibody as a crucial
studies with maternal antibodies lends weight to the risk factor for severe dengue during infancy.
hypothesis of immune involvement in the development Antibody-enhanced dengue virus infection has been
of severe dengue.23 However, severe dengue can also linked to heightened production of vasoactive cytokines
happen in primary dengue infection, and only a small and chemokines that have roles in the vascular
proportion of secondary infections progress to severe dysfunction associated with severe dengue.44–47 Antibody-
disease, which suggests roles for other factors. There is enhanced infection has also been associated with a
evidence for links between disease severity and nutritional caspase-dependent apoptosis of target cells.48 However, it
status,24 ethnicity,25 virulence of infecting viral strain,8,26,27 is rare for a patient with dengue experiencing a secondary
and host genetic background.28,29 infection to progress to more severe forms of the disease,
which indicates that other factors must play a part in
Naturally acquired immunity pathogenesis.49 In addition, it might be that not all cases
The envelope glycoprotein plays the most important part of severe disease result from secondary infection.50 If so,
in protective immunity to dengue viruses, and contains it follows that antibody-dependent enhancement is
the main epitopes recognised by neutralising antibodies.30 neither sufficient nor entirely necessary for severe
Robust neutralising antibody responses develop after disease. Indeed, in some studies enhancing antibody
dengue infection and are believed to provide lifelong activity in pre-illness plasma has been shown not to
protection against reinfection with the same dengue predict subsequent disease severity or viraemia in
serotype and short-lived protection, of several months, secondary infection.51 Whatever the role of antibody-
against a heterologous dengue serotype.27,31 This naturally dependent enhancement, it seems that a vaccine inducing
acquired immunity provides optimism for the feasibility a long-lived neutralising antibody response against all
of a dengue vaccine. four serotypes simultaneously should not induce any risk
The role of dengue-specific cellular immunity in in this respect.52–54
protection against reinfection is less clearly defined.
T cells might limit the expansion of infection by killing Cell-mediated immunity
infected cells and secreting inflammatory cytokines, and The possible involvement of T cells in the pathogenesis
are likely to have a role in viral clearance.32,33 The important of dengue was suggested more than two decades ago and
protective features of the cellular response include a high developments in T-cell immunology have refined our
avidity, homologous response of a T-helper-1-type cell understanding of the key events and mechanisms
secreting interferon γ.34,35 However, cellular immunity involved.55–57 In this model, following the antibody-
cannot be isolated from the antibody response, and any dependent enhancement of viral replication in monocytes
model of protection against dengue or pathogenesis of and macrophages, viral antigens are presented, in
severe disease must take into consideration both arms of conjunction with human lymphocyte antigen molecules,
the immune response.36,37 to CD4 and CD8 memory T cells, sensitised during a
previous infection. This leads to cellular proliferation
Immunopathogenesis and release of proinflammatory cytokines such as tumour
Antibodies necrosis factor α (TNFα) that act directly upon vascular
Epidemiological studies showing an increased risk of endothelial cells and result in plasma leakage (figure).58
severe dengue in people who had been infected with The T cells generated during a primary dengue infection
the dengue virus in the past led to the hypothesis of respond to variant peptides during the secondary
antibody-dependent enhancement of infection. infection. In a detailed study in Thai children,59 these
Antibody-dependent enhancement is believed to T-cell responses showed a lower affinity for the infecting
happen because pre-existing non-neutralising virus and a higher affinity for another virus serotype,
concentrations of antibodies and the infecting dengue thus mimicking so-called original antigenic sin. This
virus form complexes that bind to Fc receptor-bearing leads to a selective expansion of lower avidity cross-
cells, leading to increased virus uptake and replication reactive memory T cells that might out-compete the naive
thereby increasing viraemia and consequent disease T cells with higher avidity for the infecting serotype.59,60
severity.38–40 Antibody-dependent enhancement is readily This response would be less efficient in eliminating
shown in vitro using dengue immune sera or newly encountered dengue virus serotypes and could
monoclonal antibodies and cells of monocytic and lead to increased virus replication and more severe
B-lymphocytic lineages bearing Fc receptors.38,41,42 disease.
Antibody-dependent enhancement has also been shown The extent of T-cell activation is related to disease
in monkeys infused with human dengue immune severity23,48 and cross-reactive T cells have been shown to
serum.43 Further support for the role of antibody- produce much higher levels of both type 1 and certain
dependent enhancement in complex disease is provided type 2 cytokines, many previously implicated in dengue
by results of infection-enhancement assays in pathogenesis.61–64 There is also evidence that previous
Vietnamese infants23 implicating maternally derived infection with other flaviviruses that commonly circulate

www.thelancet.com/infection Vol 9 November 2009 679


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Review

disease severity in people with heterotypic immunity


necessitates a vaccine that will confer long-term
Monocyte Interferon γ
Apoptosis of protection against all four serotypes.70 All vaccines in
HLA- T-cell
naive T cells development are multivalent and aimed at producing
peptide receptor immunity against all four serotypes. However, whether a
vaccine needs all four serotypes remains an unanswered
Antigen Memory T-cell question, and will depend on the degree of longlasting
presentation Expansion of
low affinity
cross protection between serotypes. Vaccine-induced
cross-reactive enhancement of disease is not without precedent and
Fcγ receptor T cells
has been seen in respiratory syncytial virus infection of
Non-neutralising antibody Secretion of children previously immunised with a formalin-
vasoactive
Antibody-dependent cytokines: inactivated vaccine.71 Additionally, multiple serotypes
enhancement TNFα commonly circulate together in a particular geographical
Interleukin 2
Interferon γ region, with each serotype being capable of causing
Platelet
disease (although people very rarely have more than one
Dengue virus bout of severe illness, which suggests that there might be
more cross-serotypic protection than has been assumed),
Endothelium
and serotypes can change unpredictably from one season
Dengue virus and antibody complexing with to the next.72,73 A further challenge for vaccinologists has
platelets and activating clotting cascade Plasma leakage
been the lack of a suitable animal model with the same
Figure: Schematic representation of the immunopathogenesis of severe dengue disease
pathological features of human infection. Replication
and immunogenicity can be studied in rhesus macaques,
acknowledged as the best available primate model,
with dengue might affect the clinical course of subsequent although levels of viraemia are substantially lower than
infections with other members of the family. CD4 T-cell in human beings and the clinical syndrome is different,
clones from patients with dengue infection have been with little or no shock, making extrapolation of research
shown to have substantial epitope cross-reactivity among findings difficult.
flaviviruses.65 Recent advances in molecular biology and vaccinology
Regulatory T cells have been studied in acute infection have given rise to new hopes with the development of
because T-cell immunopathology is thought to play a live attenuated vaccines at present in clinical trials and a
major part in the pathogenesis of severe disease. pipeline of candidate inactivated, subunit protein, DNA,
Although regulatory T cells are fully functional, able to and viral-vector vaccines are in preclinical development.
suppress the production of vasoactive cytokines, and Progress is being made and phase 1 and 2 studies of
expand in acute dengue infection, this expansion is likely promising vaccine candidates have taken place. Extended
to be insufficient or happen too slowly to control the phase 2 and phase 3 trials will start in populations
immune response in patients with severe disease.66 exposed to dengue, and new guidelines for evaluating
these vaccines have been established by WHO.74 These
Cytokines guidelines emphasise the need for strong regulatory
Studies have suggested that the capillary permeability infrastructure as well as the importance of establishing
resulting from dengue infection is caused by the relevant study populations, defined trial clinical and
malfunction of vascular endothelial cells induced by laboratory endpoints, consistent classification of dengue
cytokines or chemical mediators rather than by syndromes, defined immune features that relate to
destruction of the small vessels.30,35,36 Following massive protection (including the relative protection afforded by
activation of memory T cells, a cytokine storm targeting neutralising antibody vs cellular immunity), and
vascular endothelial cells might lead to the crucial characterisation of inhibitory or enhancing effects of
pathological event of capillary permeability and hence earlier flavivirus infections.
plasma fluid and protein leakage. Patients with severe Correlates of immune-mediated protection to most
disease have higher concentrations of cytokines including vaccines are still unknown, and this slows the
interferon γ, TNFα, and interleukins 2, 6, 1, 8, and 10.30 development of new vaccines. However, recent studies
These findings have been supported by recent studies of clarifying the protective mechanisms of the successful
sera from severe dengue patients in Vietnam,67 India,68 yellow fever vaccine will be invaluable in vaccine
and Cuba.69 development. Early gene signatures that predict immune
responses in people vaccinated for yellow fever have been
Vaccines identified that might allow vaccine efficacy to be
There is an urgent need for a safe and effective vaccine established prospectively.75 Additionally, the use of
for dengue and the features of an ideal dengue vaccine functional genomics has identified a pattern of immune
are outlined in the panel. The association of increased responses to yellow fever vaccine that show that the

680 www.thelancet.com/infection Vol 9 November 2009


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Review

immune response to a successful vaccine is preceded by


Panel: Characteristics of the ideal dengue vaccine
a coordinated induction of transcription factors leading
to the development of a broad, polyfunctional, and • Good safety profile
persistent immune response that integrates all effector • Rapid immunisation regime requiring a single vaccine or
cells of the immune system.76 The candidate vaccines for two that fit in with established vaccine programmes
dengue that are in various stages of development are • Balance between reactogenicity and immunogenicity
discussed below and summarised in the table. • Suitable for use in target age groups
• Genetically stable
Chimeric vaccines • Stimulates neutralising antibodies and T-helper-1
The leading candidate vaccine in clinical trials at present cell-mediated immunity
is the ChimeriVax dengue vaccine. Using a new • Provide longlasting immunity
technology, the premembrane and envelope genes of • Generates neutralising immunity to all four serotypes
yellow fever 17D virus are replaced with those of each • Does not contribute to immunopathogenesis
wild-type dengue virus serotype. ChimeriVax dengue (vaccine-induced enhancement)
vaccine viruses are then prepared by electroporation of • Easily stored and transported
Vero cells with RNA transcripts prepared from viral • Affordable and cost effective
cDNA.77 Three independent, phase 1 clinical trials have
been done in Australia and USA in animals unexposed
or immune to flaviviruses. As in previous studies in non- and Marchette82 who used primary dog kidney cells. Several
human primates (in which they afford protection against monovalent vaccines passaged through primary dog
virulent virus challenge), the vaccines were shown to be kidney cells were developed and combined into several
safe in both groups and induced interferon γ responses tetravalent formulations that have been tested in phase 1
for both CD4 and CD8 T cells. This vaccine is in phase 2 and 2 trials in American adult volunteers or Thai adults
and soon to enter phase 3 trials.54,77,78 and children where they were shown to be safe and
immunogenic.52,53,80,81,83,102–104 However, the leading live
Live attenuated vaccines attenuated vaccine candidates have encountered problems
The cost effectiveness, safety, long-term immunity, and and plans for phase 3 studies have been suspended.
efficacy associated with the live attenuated vaccines Reverse genetics allows the recovery of well-defined,
against the flaviviruses yellow fever virus and Japanese genetically homogeneous virus populations from cloned
encephalitis virus serve as a model for the feasibility of a cDNA in tissue-culture cells.100 Using recombinant DNA
live attenuated dengue vaccine. technology, panels of mutant DENV4 viruses with engine-
Live attenuated vaccines have been among the leading ered deletions of varying lengths in the 3´ untranslated
vaccine candidates for dengue and could provide an region were generated.105 One such deletion mutant,
economically viable approach to inducing durable lacking nucleotides 10 478–10 507 from the 3´untranslated
humoral and cellular immunity.88,98 Neutralising region (now referred to as rDEN4Δ30), does not produce
antibodies are known to mediate protection from detectable viraemia after inoculation of rhesus monkeys
reinfection with a homologous serotype, and long-term but does induce high levels of neutralising antibodies.105,106
immunity to dengue viruses has been described.99 Importantly however, it replicates to very high titres in
Different strategies have been adopted to develop Vero cells that are suitable for vaccine production.100 The
monovalent live attenuated vaccines against each of the vaccine was found to be safe and highly immunogenic
four dengue serotypes that balance attenuation and even at low doses. However, asymptomatic rash, neutro-
immunogenicity. The virus must be able to replicate penia, and raised serum aminotransferase concentrations
sufficiently well in vivo to provoke an immune response, were noted in some vaccinees receiving the highest dose
but must be sufficiently restricted in systemic replication tested.84 A further mutation has been introduced to
to avoid inducing any of the dengue-associated symptoms. generate the vaccine rDEN4Δ30-200,201 that shows
The vaccines need to induce a broad neutralising antibody retained immunogenicity with an improved safety
response against each serotype, because if the four vaccine profile.88
viruses interfere with virus replication they could result in Since the Δ30 mutation in DENV4 is in the non-
preferential antibody responses to the better replicating structural gene regions of the virus, this provides a
components, leading to disease susceptibility.100,101 genetic DENV4 backbone for the creation of chimeric
For over 20 years, monovalent live attenuated vaccine viruses containing the structural genes encoding the
candidates, propagated and attenuated in primary and capsid, premembrane, and envelope glycoproteins of the
diploid cell cultures have been evaluated in human other dengue serotypes.85,86 These have been shown to be
beings.79–81 Initially, many candidates were either under- safe and immunogenic although studies are needed to
attenuated with unacceptable side-effect profiles or over- evaluate tetravalent combinations.87 Low doses are needed
attenuated and lacking desirable immunogenicity. An to induce immunity that could make these vaccines
acceptable balance was ultimately achieved by Halstead economical to manufacture.87

www.thelancet.com/infection Vol 9 November 2009 681


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Review

Details Phase of Comment


clinical trial
Chimeric
ChimeriVax (Acambis/ Recombinant infectious cDNA clone of yellow fever 17D vaccine 2b Leading candidate; safe and immunogenic in
Sanofi Pasteur)54,77,78 strain as a backbone, substituting membrane precursor protein human trials
and envelope protein genes with those of dengue viruses
Live attenuated
Mahidol University Passage in primary cell culture 2* Monovalent vaccines show good immune
(Sanofi Pasteur)52,53,79–83 responses; difficulties with tetravalent
formulations
WRAIR (GSK)52,53,79–83 Passage in primary cell culture 2* Monovalent vaccines show good immune
responses; difficulties with tetravalent
formulations
Infectious clone
rDEN4Δ30 (NIAID)84–87 30 nucleotide deletion from DENV4 3’ untranslated region as 1 Monovalent vaccines show promise; tetravalent
genetic backbone for vaccines with structural genes from other formulations to be evaluated
serotypes
rDEN4Δ30-200,201 Further mutation in rDEN4Δ30 construct 1 Retained immunogenicity of rDEN4Δ30, but
(NIAID)88 with improved safety profile
Inactivated
WRAIR89 Whole purified inactivated virus Preclinical Safe and immunogenic in rhesus macaques with
evidence of efficacy
Replication-incompetent
RepliVax (Novartis)90 Capsid gene-deleted WNV with membrane precursor protein and Preclinical Immunogenicity and efficacy shown in mice
envelope protein genes substituted for dengue genes
Protein
r80E (Hawaii Amino-terminal 80% of the DENV2 envelope with adjuvants Preclinical Immunogenic with non-sterilising immunity in
Biotechnology)89 mice
cEDIII (IPK/CIGB)91 Consensus dengue virus envelope protein domain III of all four Preclinical Immunogenic in mice
serotypes
DNA
US Navy92–95 Several encoding membrane precursor protein and envelope Preclinical Immunogenic with very short-lived protection
protein genes
Virus vector
Adenovirus Tetravalent formulation combining two bivalent adenovirus Preclinical Neutralising antibody responses and short-term
(GenPhar Inc)96 constructs and long-term protection against challenge
from each serotype in rhesus macaques
Measles virus (CNRS)97 Expression of a DENV1 antigen by a vector derived from live- Preclinical Long-term production of dengue neutralising
attenuated Schwarz measles vaccine antibody in mice

*Development suspended.

Table: Candidate dengue vaccines in development

Inactivated vaccines protection against viraemia (by bioassay) in a virulent


Inactivated vaccines have attracted less attention than dengue challenge, although the vaccine did not confer
live attenuated vaccines because they are likely to induce sterile immunity.89 To date, there have been no clinical
a less robust immune response (failing to induce trials of inactivated dengue vaccines in people.
T-helper-1 cellular immunity) that is less durable, are
likely to need multiple inoculations, and are more Replication-incompetent vaccines
expensive to produce. However, unlike vaccine The new approach of creating replication-incompetent
formulations consisting of four different attenuated (encapsidation-defective) viruses has been taken to resolve
viruses, they are likely to induce a more reliably consistent the problem of increasing the immunogenicity of
immune response (reducing fears of vaccine-induced inactivated vaccines while maintaining their safety profile.90
enhancement of disease) and would be safe in people These are capsid gene-deleted viruses, named RepliVAX,
that are immunocompromised. The safety, immuno- that undergo only a single cycle of infection in vaccinated
genicity, and protective efficacy of a whole purified hosts. A DENV2 vaccine has been generated by replacing
inactivated virus formulated with one of five different the premembrane and envelope genes of RepliVAX WN
adjuvants has been evaluated in rhesus macaques. High (a West Nile virus vaccine construct). After passage the
concentrations of neutralising antibodies were observed vaccine developed further mutations enabling efficient
after booster immunisation with nearly complete growth in suitable cell lines (named RepliVAX DEN2.2)

682 www.thelancet.com/infection Vol 9 November 2009


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Review

and has been shown to have good immunogenicity and responses in people113,114 although they are effective at
efficacy in an established immunodeficient mouse model priming cellular immune responses, including those of
of dengue.90 Similar vaccines have been produced for T-helper-1-type CD4 T cells and CD8 cytotoxic T cells, for
West Nile virus and Japanese encephalitis virus, which are boosting by recombinant viruses.115,116 DNA vaccines are
potent and efficacious in animals, showing potential use of expensive to produce and are likely to need complicated
this technology for the production of third-generation immunisation regimens; therefore they are unlikely to
flavivirus vaccines that should be potent, economical to be of real use for prophylactic vaccination in resource-
produce, and safe in immunocompromised people.107 poor settings.
Several different virus vectors have been investigated
Subunit vaccines as flaviviral vaccines. Poxvirus vectors, such as modified
The use of recombinant DNA technology as a means of vaccinia virus Ankara and fowlpox are attractive
generating subunit protein vaccines in various expression candidates because they are able to tolerate the insertion
systems represents a substantial advance. However, of large amounts of foreign genetic material and are safe
many of these non-infectious, subunit vaccines are poorly and immunogenic (particularly in heterologous prime-
immunogenic and need potent immunostimulatory boost combinations) in the study of other major
adjuvants that activate the innate immune system and worldwide infectious diseases such as malaria,117,118
thereby elicit antigen-specific immune responses.108–111 tuberculosis,119 and HIV.120 However, an attenuated
Aluminium-containing adjuvants are an important vaccinia virus (NYVAC) and a canarypox vector (ALVAC)
component of many vaccines, but few new adjuvants when used to generate Japanese encephalitis virus
have received approval because of concerns of adverse vaccines were insufficiently immunogenic.121
effects.111 Monophosphoryl lipid A is a detoxified form of Subsequently, research efforts have targeted replication-
the endotoxin lipopolysaccharide, and is among the first deficient adenoviruses that are safe and easy to produce
of a new generation of toll-like receptor agonists likely to and store.96 A tetravalent vaccine formulated by mixing
be used as vaccine adjuvants on a mass scale in human two bivalent adenovirus constructs (named CAdVax-
populations.109 Den12 and CAdVax-Den34) elicited a neutralising
A recombinant subunit protein containing the amino- antibody response to all four serotypes and provided both
(N)-terminal 80% of the DENV2 envelope protein (named short-term and long-term protection against challenges
r80E) has been studied in formulations with one of five from each of the four serotypes in rhesus macaques.96 To
different adjuvants based on aluminium, fraction QS-21 bypass pre-existing immunity to human adenoviruses
of saponin, and monophosphoryl lipid A. In mice, all and to improve vaccine efficacy, vaccine vectors based on
formulations were immunogenic with evidence of non- simian adenoviruses have been generated and found to
sterilising protective efficacy.89 be immunogenic for antibodies and T cells in mice.122
A new tetravalent protein vaccine candidate has been However, a major setback for adenovirus vaccines came
developed that comprises a consensus dengue virus with the results of the STEP HIV adenovirus vaccine trial
envelope protein domain III (cED III) obtained by in which the vaccine appeared to increase the rate of HIV
alignment of aminoacid sequences from different isolates infection in people that had already acquired immunity
of all four serotypes. Formulation of recombinant cED III to the adenovirus vector.123 Whether the failure of this
with aluminium phosphate induces cross-neutralising trial is attributable to something specific about the Merck
antibodies and memory immunity after challenge with vector or whether all adenovirus vectors will face similar
dengue virus in Balb/c mice.91 problems remains to be seen.
DNA vaccines use plasmids of double-stranded DNA The measles vaccine is another potential vaccine vector.
in which the genetic sequence for a protein or series of Over the past 30 years, it has been given to hundreds of
epitopes has been inserted under the control of a millions of children and has proved to be both effective
mammalian promoter. They are usually given and safe. This attenuated live virus induces lifelong
intramuscularly and are taken up by muscle and dendritic immunity after only one or two injections and can be
cells, where the genetic information for the encoded produced on a large scale and at a low cost.124 A new
protein is translated.108 Several dengue DNA vaccines vaccine has been developed based on the expression of a
(encoding premembrane and envelope genes) have been minimal dengue antigen (of DENV1) by a vector derived
studied in mice and monkeys.92–95 In aotus monkeys, a from paediatric live-attenuated Schwarz measles vaccine.
DENV3 DNA vaccine elicited a neutralising antibody Immunisation of mice resulted in a long-term production
response and partly protected against live virus of DENV1 serotype-specific neutralising antibodies.97 The
challenge;95 a DENV1 DNA vaccine was capable of protective efficacy of this vaccine should be further
inducing neutralising antibodies in rhesus macaques evaluated in non-human primates. A combined measles–
after multiple inoculations but with very short-lived dengue vaccine might provide an opportunity to
protection.94 Although DNA vaccines showed early immunise children against both diseases where they
promise in other disease models in animals,112 DNA coexist, although immunological interference would
vaccination alone generally elicits only weak immune need to be evaluated in people.97

www.thelancet.com/infection Vol 9 November 2009 683


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Review

update the vaccine as dengue viruses evolve and if this


Search strategy and selection criteria will drive evolution, whether the vaccine can be produced
Data for this Review were identified by searching PubMed for at a cost acceptable to endemic countries, and the cost-
relevant articles. Search terms included “dengue”, effectiveness of dengue prevention through vaccination.
“epidemiology”, “vaccine”, “pathogenesis”, Comparisons need to be made of different vaccination
“immunopathogenesis”, “antibody”, “T-cell”, and “cell- strategies and vector control programmes as well as the
mediated”. Articles were selected on the basis of their relevance estimated cost-effectiveness of dengue prevention
to dengue vaccine development. No date restrictions were compared with potential treatment with new antiviral
used. Although all included publications were in English, the drugs.
search was not restricted to English language articles.
Conclusion
Progress with the development of dengue vaccines has
Unanswered questions accelerated but, alongside this, the scale of the problem
Substantial progress has been made with dengue vaccine has rapidly grown and a safe effective vaccine remains
development and yet there remain a number of questions an urgent worldwide priority. Several candidate vaccines
that need to be answered before the widespread use of have been developed that have shown promise in
any candidate vaccine. Preclinical and clinical studies are phase 2 clinical studies. The leading candidate must
needed to address these issues and well-designed vaccine now be taken forward into larger studies in appropriate
studies could also contribute to our understanding. populations in endemic areas. A coordinated approach
is needed to ensure consistency, with defined endpoints
Preclinical that can be extrapolated between different studies and
Questions that need answering through preclinical that will also contribute to our understanding of dengue
studies relate to the immune correlates of protection and immunity and pathogenesis. Ideally, a defined structure
severe disease, and if virus-to-virus interference in linking organisations involved in vaccine development
tetravalent formulations inhibits the antibody response and coordinated by WHO would allow the sharing of
to one or more serotype in the vaccine when used in information from vaccine studies particularly in relation
endemic areas. With respect to the immune correlates, to adverse events, safety, and knowledge gained on the
appropriate and improved laboratory tests need to be potential for immune enhancement during trials.
developed to find what constitutes protective immune Convincing arguments need to be in place for
responses and which responses might predispose to introducing dengue vaccine initiatives. The
severe disease. socioeconomic effect of dengue is substantially larger
than that attributable to severe dengue alone and this
Clinical effect needs to be considered in the case for vaccine
Clinical studies are needed to answer many further licensure and use alongside the cost-effectiveness of
questions, such as the duration of protection afforded by other approaches.74
candidate vaccines. Mass immunisation with a vaccine The opportunity now exists to bring effective and safe
that only affords short-lived protection (even if this is dengue vaccines to populations at greatest risk, and there
only to one serotype) could simply make whole is unprecedented momentum to bring about a
populations susceptible to the more severe forms of coordinated worldwide effort.
disease. Further clinical questions relate to the effect of Contributors
pre-existing immunity to other flaviviruses (through The content and structure of this Review were agreed by all authors.
natural exposure or immunisation), the safety of DPW searched the published work and prepared the first draft.
Contributions were received from JF and SR-J. All authors edited the
candidate vaccines to infants (so far there have been no final draft.
studies in infants in whom safety profiles and immune
Conflicts of interest
response kinetics might differ from adults), the We declare that we have no conflicts of interest.
appropriate age to immunise, and if these vaccines are
Acknowledgments
safe for people infected with HIV. DPW is an employee of the Oxford Radcliffe Hospitals NHS Trust, UK.
JF is funded by the Wellcome Trust. SR-J is funded by the Medical
Implementation Research Council, UK.
Beyond questions for preclinical and clinical studies are References
questions about the logistics of a vaccine implementation 1 WHO. Strengthening implementation of the global strategy for
dengue fever/dengue haemorrhagic fever prevention and control—
strategy. Answers are needed on how long follow-up data report of the Informal Consultation. Geneva, Switzerland: World
will be needed to confirm long-term safety, the age of the Health Organization, 1999. http://www.whoindia.org/LinkFiles/
targeted vaccinees, the proportion of vaccine coverage Dengue_GlobalStrategy.pdf (accessed Sept 22, 2009).
2 Kyle JL, Harris E. Global spread and persistence of dengue.
needed for herd immunity, and, crucially, what the Annu Rev Microbiol 2008; 62: 71–92.
vaccine will do—will it stop all disease or just severe 3 Gubler DJ. The global emergence/resurgence of arboviral diseases
disease? Other questions relate to the need to regularly as public health problems. Arch Med Res 2002; 33: 330–42.

684 www.thelancet.com/infection Vol 9 November 2009


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Review

4 Gubler DJ. Epidemic dengue/dengue hemorrhagic fever as a 28 LaFleur C, Granados J, Vargas-Alarcon G, et al. HLA-DR antigen
public health, social and economic problem in the 21st century. frequencies in Mexican patients with dengue virus infection:
Trends Microbiol 2002; 10: 100–03. HLA-DR4 as a possible genetic resistance factor for dengue
5 Teixeira MG, Costa MC, Coelho G, Barreto ML. Recent shift in age hemorrhagic fever. Hum Immunol 2002; 63: 1039–44.
pattern of dengue hemorrhagic fever, Brazil. Emerg Infect Dis 29 Lan NT, Kikuchi M, Huong VT, et al. Protective and enhancing HLA
2008; 14: 1663. alleles, HLA-DRB1*0901 and HLA-A*24, for severe forms of dengue
6 Gubler DJ, Meltzer M. Impact of dengue/dengue hemorrhagic virus infection, dengue hemorrhagic fever and dengue shock
fever on the developing world. Adv Virus Res 1999; 53: 35–70. syndrome. PLoS Negl Trop Dis 2008; 2: e304.
7 Anuradha S, Singh NP, Rizvi SN, Agarwal SK, Gur R, 30 Kurane I. Dengue hemorrhagic fever with special emphasis on
Mathur MD. The 1996 outbreak of dengue hemorrhagic fever in immunopathogenesis. Comp Immunol Microbiol Infect Dis 2007;
Delhi, India. Southeast Asian J Trop Med Public Health 1998; 30: 329–40.
29: 503–06. 31 Sabin AB. Research on dengue during World War II.
8 Vaughn DW. Invited commentary: Dengue lessons from Cuba. Am J Trop Med Hyg 1952; 1: 30–50.
Am J Epidemiol 2000; 152: 800–03. 32 Bukowski JF, Kurane I, Lai CJ, Bray M, Falgout B, Ennis FA. Dengue
9 Chen HL, Lin SR, Liu HF, King CC, Hsieh SC, Wang WK. virus-specific cross-reactive CD8+ human cytotoxic T lymphocytes.
Evolution of dengue virus type 2 during two consecutive J Virol 1989; 63: 5086–91.
outbreaks with an increase in severity in southern Taiwan in 33 Kurane I, Meager A, Ennis FA. Dengue virus-specific human T cell
2001–2002. Am J Trop Med Hyg 2008; 79: 495–505. clones: serotype crossreactive proliferation, interferon gamma
10 Freedman DO, Weld LH, Kozarsky PE, et al. Spectrum of disease production, and cytotoxic activity. J Exp Med 1989; 170: 763–75.
and relation to place of exposure among ill returned travelers. 34 Rothman AL. Dengue: defining protective versus pathologic
N Engl J Med 2006; 354: 119–30. immunity. J Clin Invest 2004; 113: 946–51.
11 Kuno G, Chang GJ, Tsuchiya KR, Karabatsos N, Cropp CB. 35 Green S, Rothman A. Immunopathological mechanisms in dengue
Phylogeny of the genus Flavivirus. J Virol 1998; 72: 73–83. and dengue hemorrhagic fever. Curr Opin Infect Dis 2006;
12 Chang G. Molecular biology of dengue viruses. In: Gubler D, 19: 429–36.
Kuno G, eds. Dengue and dengue hemorrhagic fever. New York, 36 Rothman AL, Ennis FA. Immunopathogenesis of dengue
NY: CAB International, 1997: 175–98. hemorrhagic fever. Virology 1999; 257: 1–6.
13 Rodhain F, Rosen L. Mosquito vectors and dengue virus–vector 37 Guy B, Almond JW. Towards a dengue vaccine: progress to date and
relationships. In: Gubler D, Kuno G, eds. Dengue and dengue remaining challenges. Comp Immunol Microbiol Infect Dis 2008;
hemorrhagic fever. New York, NY: CAB International, 1997: 31: 239–52.
45–60. 38 Goncalvez AP, Engle RE, St Claire M, Purcell RH, Lai CJ.
14 Effler PV, Pang L, Kitsutani P, et al. Dengue fever, Hawaii, Monoclonal antibody-mediated enhancement of dengue virus
2001–2002. Emerg Infect Dis 2005; 11: 742–49. infection in vitro and in vivo and strategies for prevention.
15 Knudsen AB, Slooff R. Vector-borne disease problems in rapid Proc Natl Acad Sci USA 2007; 104: 9422–27.
urbanization: new approaches to vector control. 39 Guzman MG, Alvarez M, Rodriguez-Roche R, et al. Neutralizing
Bull World Health Organ 1992; 70: 1–6. antibodies after infection with dengue 1 virus. Emerg Infect Dis 2007;
16 Brandt WE. From the World Health Organization: development of 13: 282–86.
dengue and Japanese encephalitis vaccines. J Infect Dis 1990; 40 Vaughn DW, Green S, Kalayanarooj S, et al. Dengue viremia titer,
162: 577–83. antibody response pattern, and virus serotype correlate with disease
17 Stephenson JR. Developing vaccines against flavivirus diseases: severity. J Infect Dis 2000; 181: 2–9.
past success, present hopes and future challenges. 41 Littaua R, Kurane I, Ennis FA. Human IgG Fc receptor II mediates
Novartis Found Symp 2006; 277: 193–201. antibody-dependent enhancement of dengue virus infection.
18 WHO. Dengue haemorrhagic fever: diagnosis, treatment, J Immunol 1990; 144: 3183–86.
prevention and control, 2nd edn. Geneva: World Health 42 Morens DM, Venkateshan CN, Halstead SB. Dengue 4 virus
Organization, 1997. http://www.who.int/csr/resources/ monoclonal antibodies identify epitopes that mediate immune
publications/dengue/Denguepublication/en/ infection enhancement of dengue 2 viruses. J Gen Virol 1987;
(accessed Sept 22, 2009). 68: 91–98.
19 Kuo CH, Tai DI, Chang-Chien CS, Lan CK, Chiou SS, Liaw YF. 43 Halstead SB. In vivo enhancement of dengue virus infection in
Liver biochemical tests and dengue fever. Am J Trop Med Hyg rhesus monkeys by passively transferred antibody. J Infect Dis 1979;
1992; 47: 265–70. 140: 527–33.
20 Kalayanarooj S, Vaughn DW, Nimmannitya S, et al. Early clinical 44 Anderson R, Wang S, Osiowy C, Issekutz AC. Activation of
and laboratory indicators of acute dengue illness. J Infect Dis 1997; endothelial cells via antibody-enhanced dengue virus
176: 313–21. infection of peripheral blood monocytes. J Virol 1997; 71: 4226–32.
21 Deen JL, Harris E, Wills B, et al. The WHO dengue classification 45 King CA, Marshall JS, Alshurafa H, Anderson R. Release of
and case definitions: time for a reassessment. Lancet 2006; vasoactive cytokines by antibody-enhanced dengue virus infection
368: 170–73. of a human mast cell/basophil line. J Virol 2000; 74: 7146–50.
22 Halstead SB, Nimmannitya S, Cohen SN. Observations related to 46 King CA, Anderson R, Marshall JS. Dengue virus selectively
pathogenesis of dengue hemorrhagic fever: IV—Relation of induces human mast cell chemokine production. J Virol 2002;
disease severity to antibody response and virus recovered. 76: 8408–19.
Yale J Biol Med 1970; 42: 311–28. 47 Chareonsirisuthigul T, Kalayanarooj S, Ubol S. Dengue virus
23 Chau TN, Quyen NT, Thuy TT, et al. Dengue in Vietnamese (DENV) antibody-dependent enhancement of infection
infants—results of infection-enhancement assays correlate with upregulates the production of anti-inflammatory cytokines, but
age-related disease epidemiology, and cellular immune responses suppresses anti-DENV free radical and pro-inflammatory
correlate with disease severity. J Infect Dis 2008; 198: 516–24. cytokine production, in THP-1 cells. J Gen Virol 2007; 88: 365–75.
24 Thisyakorn U, Nimmannitya S. Nutritional status of children with 48 Brown MG, Huang YY, Marshall JS, King CA, Hoskin DW,
dengue hemorrhagic fever. Clin Infect Dis 1993; 16: 295–97. Anderson R. Dramatic caspase-dependent apoptosis in antibody-
25 Restrepo BN, Ramirez RE, Arboleda M, Alvarez G, Ospina M, enhanced dengue virus infection of human mast cells.
Diaz FJ. Serum levels of cytokines in two ethnic groups with J Leukoc Biol 2009; 85: 71–80.
dengue virus infection. Am J Trop Med Hyg 2008; 79: 673–77. 49 Guzman MG, Kouri G. Dengue: an update. Lancet Infect Dis 2002;
26 Rosen L. The emperor’s new clothes revisited, or reflections on 2: 33–42.
the pathogenesis of dengue hemorrhagic fever. 50 Wichmann O, Hongsiriwon S, Bowonwatanuwong C,
Am J Trop Med Hyg 1977; 26: 337–43. Chotivanich K, Sukthana Y, Pukrittayakamee S. Risk factors and
27 Guzman MG, Kouri GP, Bravo J, Soler M, Vazquez S, Morier L. clinical features associated with severe dengue infection in adults
Dengue hemorrhagic fever in Cuba, 1981: a retrospective and children during the 2001 epidemic in Chonburi, Thailand.
seroepidemiologic study. Am J Trop Med Hyg 1990; 42: 179–84. Trop Med Int Health 2004; 9: 1022–29.

www.thelancet.com/infection Vol 9 November 2009 685


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Review

51 Laoprasopwattana K, Libraty DH, Endy TP, et al. Dengue Virus 72 Nisalak A, Endy TP, Nimmannitya S, et al. Serotype-specific dengue
(DV) enhancing antibody activity in preillness plasma does not virus circulation and dengue disease in Bangkok, Thailand from
predict subsequent disease severity or viremia in secondary 1973 to 1999. Am J Trop Med Hyg 2003; 68: 191–202.
DV infection. J Infect Dis 2005; 192: 510–19. 73 Recker M, Blyuss KB, Simmons CP, et al. Immunological serotype
52 Sabchareon A, Lang J, Chanthavanich P, et al. Safety and interactions and their effect on the epidemiological pattern of
immunogenicity of a three dose regimen of two tetravalent live- dengue. Proc Biol Sci 2009; 276: 2541–48.
attenuated dengue vaccines in five- to twelve-year-old Thai 74 Edelman R, Hombach J. “Guidelines for the clinical evaluation of
children. Pediatr Infect Dis J 2004; 23: 99–109. dengue vaccines in endemic areas”: summary of a World Health
53 Sabchareon A, Lang J, Chanthavanich P, et al. Safety and Organization technical consultation. Vaccine 2008; 26: 4113–19.
immunogenicity of tetravalent live-attenuated dengue vaccines in 75 Querec TD, Akondy RS, Lee EK, et al. Systems biology approach
Thai adult volunteers: role of serotype concentration, ratio, and predicts immunogenicity of the yellow fever vaccine in humans.
multiple doses. Am J Trop Med Hyg 2002; 66: 264–72. Nat Immunol 2009; 10: 116–25.
54 Guirakhoo F, Kitchener S, Morrison D, et al. Live attenuated 76 Gaucher D, Therrien R, Kettaf N, et al. Yellow fever vaccine induces
chimeric yellow fever dengue type 2 (ChimeriVax-DEN2) vaccine: integrated multilineage and polyfunctional immune responses.
phase I clinical trial for safety and immunogenicity: effect of J Exp Med 2008; 205: 3119–31.
yellow fever pre-immunity in induction of cross neutralizing 77 Guirakhoo F, Pugachev K, Zhang Z, et al. Safety and efficacy of
antibody responses to all 4 dengue serotypes. Hum Vaccin 2006; chimeric yellow fever-dengue virus tetravalent vaccine formulations
2: 60–67. in nonhuman primates. J Virol 2004; 78: 4761–75.
55 Halstead SB. The Alexander D Langmuir lecture: the 78 Guy B, Nougarede N, Begue S, et al. Cell-mediated immunity
pathogenesis of dengue—molecular epidemiology in infectious induced by chimeric tetravalent dengue vaccine in naive or
disease. Am J Epidemiol 1981; 114: 632–48. flavivirus-primed subjects. Vaccine 2008; 26: 5712–21.
56 Pang T, Cardosa MJ, Guzman MG. Of cascades and perfect 79 Innis BL, Eckels KH. Progress in development of a live-attenuated,
storms: the immunopathogenesis of dengue haemorrhagic fever- tetravalent dengue virus vaccine by the United States Army Medical
dengue shock syndrome (DHF/DSS). Immunol Cell Biol 2007; Research and Materiel Command. Am J Trop Med Hyg 2003;
85: 43–45. 69 (suppl 6): 1–4.
57 Pang T. Delayed-type hypersensitivity: probable role in the 80 Edelman R, Wasserman SS, Bodison SA, et al. Phase I trial of
pathogenesis of dengue hemorrhagic fever/dengue shock 16 formulations of a tetravalent live-attenuated dengue vaccine.
syndrome. Rev Infect Dis 1983; 5: 346–52. Am J Trop Med Hyg 2003; 69 (suppl 6): 48–60.
58 Mangada MM, Endy TP, Nisalak A, et al. Dengue-specific T cell 81 Sun W, Edelman R, Kanesa-Thasan N, et al. Vaccination of human
responses in peripheral blood mononuclear cells obtained prior to volunteers with monovalent and tetravalent live-attenuated dengue
secondary dengue virus infections in Thai schoolchildren. vaccine candidates. Am J Trop Med Hyg 2003; 69 (suppl 6): 24–31.
J Infect Dis 2002; 185: 1697–703.
82 Halstead SB, Marchette NJ. Biologic properties of dengue viruses
59 Mongkolsapaya J, Dejnirattisai W, Xu XN, et al. Original antigenic following serial passage in primary dog kidney cells: studies at the
sin and apoptosis in the pathogenesis of dengue hemorrhagic University of Hawaii. Am J Trop Med Hyg 2003; 69 (suppl 6): 5–11.
fever. Nat Med 2003; 9: 921–27.
83 Bhamarapravati N, Sutee Y. Live attenuated tetravalent dengue
60 Mathew A, Rothman AL. Understanding the contribution of vaccine. Vaccine 2000; 18 (suppl 2): 44–47.
cellular immunity to dengue disease pathogenesis. Immunol Rev
84 Durbin AP, Whitehead SS, McArthur J, et al. rDEN4Δ30, a live
2008; 225: 300–13.
attenuated dengue virus type 4 vaccine candidate, is safe,
61 Screaton G, Mongkolsapaya J. T cell responses and dengue immunogenic, and highly infectious in healthy adult volunteers.
haemorrhagic fever. Novartis Found Symp 2006; 277: 164–71. J Infect Dis 2005; 191: 710–18.
62 Mongkolsapaya J, Duangchinda T, Dejnirattisai W, et al. T cell 85 Blaney JE Jr, Sathe NS, Goddard L, et al. Dengue virus type 3
responses in dengue hemorrhagic fever: are cross-reactive T cells vaccine candidates generated by introduction of deletions in the
suboptimal? J Immunol 2006; 176: 3821–29. 3Ľ untranslated region (3Ľ-UTR) or by exchange of the DENV-3
63 Bashyam HS, Green S, Rothman AL. Dengue virus-reactive CD8+ 3Ľ-UTR with that of DENV-4. Vaccine 2008; 26: 817–28.
T cells display quantitative and qualitative differences in their 86 Blaney JE Jr, Sathe NS, Hanson CT, Firestone CY, Murphy BR,
response to variant epitopes of heterologous viral serotypes. Whitehead SS. Vaccine candidates for dengue virus type 1 (DEN1)
J Immunol 2006; 176: 2817–24. generated by replacement of the structural genes of rDEN4 and
64 Dong T, Moran E, Vinh Chau N, et al. High pro-inflammatory rDEN4Delta30 with those of DEN1. Virol J 2007; 4: 23.
cytokine secretion and loss of high avidity cross-reactive cytotoxic 87 Durbin AP, McArthur JH, Marron JA, et al. rDEN2/4Delta30(ME),
T-cells during the course of secondary dengue virus infection. a live attenuated chimeric dengue serotype 2 vaccine is safe and
PLoS One 2007; 2: e1192. highly immunogenic in healthy dengue-naive adults. Hum Vaccin
65 Moran E, Simmons C, Vinh Chau N, et al. Preservation of a 2006; 2: 255–60.
critical epitope core region is associated with the high degree of 88 McArthur JH, Durbin AP, Marron JA, et al. Phase I clinical
flaviviral cross-reactivity exhibited by a dengue-specific CD4+ evaluation of rDEN4Delta30-200,201: a live attenuated dengue 4
T cell clone. Eur J Immunol 2008; 38: 1050–57. vaccine candidate designed for decreased hepatotoxicity.
66 Luhn K, Simmons CP, Moran E, et al. Increased frequencies of Am J Trop Med Hyg 2008; 79: 678–84.
CD4+ CD25(high) regulatory T cells in acute dengue infection. 89 Robert Putnak J, Coller BA, Voss G, et al. An evaluation of dengue
J Exp Med 2007; 204: 979–85. type-2 inactivated, recombinant subunit, and live-attenuated vaccine
67 Nguyen TH, Nguyen TL, Lei HY, et al. Association between sex, candidates in the rhesus macaque model. Vaccine 2005; 23: 4442–52.
nutritional status, severity of dengue hemorrhagic fever, and 90 Suzuki R, Winkelmann ER, Mason PW. Construction and
immune status in infants with dengue hemorrhagic fever. characterization of a single-cycle chimeric flavivirus vaccine
Am J Trop Med Hyg 2005; 72: 370–74. candidate that protects mice against lethal challenge with dengue
68 Chakravarti A, Kumaria R. Circulating levels of tumour necrosis virus type 2. J Virol 2009; 83: 1870–80.
factor-α & interferon-γ in patients with dengue & dengue 91 Leng CH, Liu SJ, Tsai JP, et al. A novel dengue vaccine candidate
haemorrhagic fever during an outbreak. Indian J Med Res 2006; that induces cross-neutralizing antibodies and memory immunity.
123: 25–30. Microbes Infect 2009; 11: 288–95.
69 Perez AB, Garcia G, Sierra B, et al. IL-10 levels in Dengue 92 De Paula SO, Lima DM, de Oliveira Franca RF, Gomes-Ruiz AC,
patients: some findings from the exceptional epidemiological da Fonseca BA. A DNA vaccine candidate expressing dengue-3 virus
conditions in Cuba. J Med Virol 2004; 73: 230–34. prM and E proteins elicits neutralizing antibodies and protects mice
70 Halstead SB. Pathogenesis of dengue: challenges to molecular against lethal challenge. Arch Virol 2008; 153: 2215–23.
biology. Science 1988; 239: 476–81. 93 Konishi E, Yamaoka M, Kurane I, Mason PW. A DNA vaccine
71 Castilow EM, Olson MR, Varga SM. Understanding respiratory expressing dengue type 2 virus premembrane and envelope genes
syncytial virus (RSV) vaccine-enhanced disease. Immunol Res induces neutralizing antibody and memory B cells in mice. Vaccine
2007; 39: 225–39. 2000; 18: 1133–39.

686 www.thelancet.com/infection Vol 9 November 2009


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Review

94 Putnak R, Fuller J, VanderZanden L, Innis BL, Vaughn DW. 110 Ishii KJ, Akira S. Toll or toll-free adjuvant path toward the optimal
Vaccination of rhesus macaques against dengue-2 virus with vaccine development. J Clin Immunol 2007; 27: 363–71.
a plasmid DNA vaccine encoding the viral pre-membrane and 111 Hem SL, Hogenesch H. Relationship between physical and
envelope genes. Am J Trop Med Hyg 2003; 68: 469–76. chemical properties of aluminum-containing adjuvants and
95 Blair PJ, Kochel TJ, Raviprakash K, et al. Evaluation of immunity immunopotentiation. Expert Rev Vaccines 2007; 6: 685–98.
and protective efficacy of a dengue-3 pre-membrane and envelope 112 Ulmer JB, Donnelly JJ, Parker SE, et al. Heterologous protection
DNA vaccine in Aotus nancymae monkeys. Vaccine 2006; against influenza by injection of DNA encoding a viral protein.
24: 1427–32. Science 1993; 259: 1745–49.
96 Raviprakash K, Wang D, Ewing D, et al. A tetravalent dengue 113 Wang R, Doolan DL, Le TP, et al. Induction of antigen-specific
vaccine based on a complex adenovirus vector provides significant cytotoxic T lymphocytes in humans by a malaria DNA vaccine.
protection in rhesus monkeys against all four serotypes of dengue Science 1998; 282: 476–80.
virus. J Virol 2008; 82: 6927–34. 114 Wang R, Epstein J, Baraceros FM, et al. Induction of CD4(+)
97 Brandler S, Lucas-Hourani M, Moris A, et al. Pediatric measles T cell-dependent CD8(+) type 1 responses in humans by a malaria
vaccine expressing a dengue antigen induces durable serotype- DNA vaccine. Proc Natl Acad Sci USA 2001; 98: 10817–22.
specific neutralizing antibodies to dengue virus. PLoS Negl Trop Dis 115 Schneider J, Gilbert SC, Hannan CM, et al. Induction of CD8+
2007; 1: e96. T cells using heterologous prime-boost immunisation strategies.
98 Edelman R. Dengue vaccines approach the finish line. Immunol Rev 1999; 170: 29–38.
Clin Infect Dis 2007; 45 (suppl 1): S56–60. 116 McConkey SJ, Reece WH, Moorthy VS, et al. Enhanced T-cell
99 Imrie A, Meeks J, Gurary A, et al. Antibody to dengue 1 detected immunogenicity of plasmid DNA vaccines boosted by recombinant
more than 60 years after infection. Viral Immunol 2007; modified vaccinia virus Ankara in humans. Nat Med 2003;
20: 672–75. 9: 729–35.
100 Blaney JE Jr, Durbin AP, Murphy BR, Whitehead SS. 117 Webster DP, Dunachie S, McConkey S, et al. Safety of recombinant
Development of a live attenuated dengue virus vaccine using fowlpox strain FP9 and modified vaccinia virus Ankara vaccines
reverse genetics. Viral Immunol 2006; 19: 10–32. against liver-stage P falciparum malaria in non-immune volunteers.
101 Blaney JE Jr, Matro JM, Murphy BR, Whitehead SS. Recombinant, Vaccine 2006; 24: 3026–34.
live-attenuated tetravalent dengue virus vaccine formulations 118 Webster DP, Dunachie S, Vuola JM, et al. Enhanced T cell-mediated
induce a balanced, broad, and protective neutralizing antibody protection against malaria in human challenges by using the
response against each of the four serotypes in rhesus monkeys. recombinant poxviruses FP9 and modified vaccinia virus Ankara.
J Virol 2005; 79: 5516–28. Proc Natl Acad Sci USA 2005; 102: 4836–41.
102 Kanesa-thasan N, Sun W, Kim-Ahn G, et al. Safety and 119 Hawkridge T, Scriba TJ, Gelderbloem S, et al. Safety and
immunogenicity of attenuated dengue virus vaccines (Aventis immunogenicity of a new tuberculosis vaccine, MVA85A, in healthy
Pasteur) in human volunteers. Vaccine 2001; 19: 3179–88. adults in South Africa. J Infect Dis 2008; 198: 544–52.
103 Sun W, Cunningham D, Wasserman SS, et al. Phase 2 clinical 120 Cebere I, Dorrell L, McShane H, et al. Phase I clinical trial safety of
trial of three formulations of tetravalent live-attenuated dengue DNA- and modified virus Ankara-vectored human
vaccine in flavivirus-naïve adults. Hum Vaccin 2009; 5: 33–40. immunodeficiency virus type 1 (HIV-1) vaccines administered alone
104 Simasathien S, Thomas SJ, Watanaveeradej V, et al. Safety and and in a prime-boost regime to healthy HIV-1-uninfected
immunogenicity of a tetravalent live-attenuated dengue vaccine in volunteers. Vaccine 2006; 24: 417–25.
flavivirus naive children. Am J Trop Med Hyg 2008; 78: 426–33. 121 Kanesa-thasan N, Smucny JJ, Hoke CH, et al. Safety and
105 Men R, Bray M, Clark D, Chanock RM, Lai CJ. Dengue type 4 immunogenicity of NYVAC-JEV and ALVAC-JEV attenuated
virus mutants containing deletions in the 3’ noncoding region of recombinant Japanese encephalitis virus—poxvirus vaccines in
the RNA genome: analysis of growth restriction in cell culture vaccinia-nonimmune and vaccinia-immune humans. Vaccine 2000;
and altered viremia pattern and immunogenicity in rhesus 19: 483–91.
monkeys. J Virol 1996; 70: 3930–37. 122 Gao G, Wang Q, Dai Z, et al. Adenovirus-based vaccines generate
106 Hanley KA, Manlucu LR, Manipon GG, et al. Introduction of cytotoxic T lymphocytes to epitopes of NS1 from dengue virus that
mutations into the non-structural genes or 3’ untranslated region are present in all major serotypes. Hum Gene Ther 2008; 19: 927–36.
of an attenuated dengue virus type 4 vaccine candidate further 123 Sekaly RP. The failed HIV Merck vaccine study: a step back or a
decreases replication in rhesus monkeys while retaining launching point for future vaccine development? J Exp Med 2008;
protective immunity. Vaccine 2004; 22: 3440–48. 205: 7–12.
107 Widman DG, Frolov I, Mason PW. Third-generation flavivirus 124 Brandler S, Tangy F. Recombinant vector derived from live
vaccines based on single-cycle, encapsidation-defective viruses. attenuated measles virus: potential for flavivirus vaccines.
Adv Virus Res 2008; 72: 77–126. Comp Immunol Microbiol Infect Dis 2008; 31: 271–91.
108 Webster D, Hill AV. Progress with new malaria vaccines.
Bull World Health Organ 2003; 81: 902–09.
109 Casella CR, Mitchell TC. Putting endotoxin to work for us:
monophosphoryl lipid A as a safe and effective vaccine adjuvant.
Cell Mol Life Sci 2008; 65: 3231–40.

www.thelancet.com/infection Vol 9 November 2009 687


Descargado para Anonymous User (n/a) en Simon Bolivar University de ClinicalKey.es por Elsevier en febrero 12, 2022. Para uso
personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.

You might also like