Вы находитесь на странице: 1из 13

Series

Dengue 3
Dengue: knowledge gaps, unmet needs, and research priorities
Leah C Katzelnick, Josefina Coloma, Eva Harris

Dengue virus is a mosquito-borne pathogen that causes up to about 100 million cases of disease each year, placing a Lancet Infect Dis 2017;
major public health, social, and economic burden on numerous low-income and middle-income countries. Major 17: e88–100

advances by investigators, vaccine developers, and affected communities are revealing new insights and enabling novel Published Online
February 6, 2017
interventions and approaches to dengue prevention and control. Such research has highlighted further questions
http://dx.doi.org/10.1016/
about both the basic understanding of dengue and efforts to develop new tools. In this report, the third in a Series on S1473-3099(16)30473-X
dengue, we discuss existing approaches to dengue diagnostics, disease prognosis, surveillance, and vector control in See Series pages e70 and e79
low-income and middle-income countries, as well as potential consequences of vaccine introduction. We also This is the third in a Series of
summarise current knowledge and recent insights into dengue epidemiology, immunology, and pathogenesis, and three papers on dengue
their implications for understanding natural infection and current and future vaccines. Division of Infectious Diseases
and Vaccinology, School of
Introduction Aedes aegypti and Aedes albopictus. Dengue fever is Public Health, University of
California, Berkeley, CA, USA
Dengue is the most prevalent arboviral disease in human characterised by debilitating symptoms, including high (L C Katzelnick PhD,
beings, with 3·6 billion people living in areas with risk of fever, arthralgia, myalgia, anorexia, petechiae or rash, and J Coloma PhD, Prof E Harris PhD)
disease transmission, and with an estimated 390 million retro-orbital pain. Because symptoms are non-specific and Correspondence to:
dengue virus infections and 96 million dengue cases overlap with those of many other infections, such as Prof Eva Harris, Division of
annually.1 Dengue is endemic to the tropical belt of Asia, chikungunya virus and Zika virus, laboratory diagnosis is Infectious Diseases and
Vaccinology, School of Public
Latin America, and the Pacific, it circulates across Africa, required. Upon defervescence, during the critical phase
Health, University of California,
and it has recently caused local outbreaks in the USA and (days 4–6 of illness), a fraction of patients (about Berkeley, Berkeley,
parts of Europe.1–3 Dengue has expanded globally since 500 000 per year) develop dengue haemorrhagic fever/ CA 94720-3370, USA
the 1960s, driven by population growth, urbanisation, dengue shock syndrome, which is characterised by well eharris@berkeley.edu

increased travel, and insufficient vector control pro- described clinical and haematological features that can
grammes. Despite increased funding and advances in facilitate clinical diagnosis of dengue, even without
dengue research, dengue epidemics are intensifying in laboratory confirmation. These features include thrombo-
frequency, magnitude, and geographical reach.4 The cytopenia, haemorrhagic manifestations, liver damage,
global burden of dengue is estimated at 25·5 disability- leucopenia, pleural effusion and other signs of vascular
adjusted life-years per 100 000 individuals, with major leakage, hypovolaemic shock, and ultimately organ failure.7
economic, social, and political effects.5,6 Public health In 2009, a new classification of dengue disease was adopted
systems are strained by the relentless spread of dengue by WHO, with “severe dengue” encompassing additional
virus and other arboviruses, such as chikungunya virus manifestations of severe disease.8 Mortality due to dengue
and Zika virus, and they are discouraged by decades of can be greatly reduced by early diagnosis and timely and
failed vector control programmes and absence of new judicious fluid management; hence, laboratory diagnosis
interventions. together with clinical acumen is essential.8
However, never before has the level of resources and A combination of diagnostic methods that target
commitment from diverse researchers and stakeholders different periods after onset of symptoms maximises
been as great, or as focused on increasing basic diagnostic sensitivity (table 1); however, only early acute-
knowledge, potential treatments and vaccines, and new phase diagnostics can affect clinical management.
vector control strategies, with the ultimate goal of These include detection of whole virus, viral RNA, and
conquering dengue. In this paper, we address existing non-structural protein 1 (NS1). Variable quality of
tools and needs for dengue diagnostics and surveillance available assays and their high cost are still limiting
in low-income and middle-income countries, and factors,10 often placing them out of reach for national
review current knowledge and research gaps in coverage. The IgM ELISA is widely used for dengue
immunology, epidemiology, dengue pathogenesis, and diagnosis, but because samples for this assay are
vector control in the context of natural infection and collected more than 5 days after symptom onset, results
vaccines. Many of these insights call into question are obtained too late for clinical decision-making. There
existing paradigms in the dengue field, and raise many is a major unmet need for affordable, easy-to-perform,
new and exciting questions. acute-phase dengue diagnostics. This is even more
urgent given the recent, widespread introduction of
Case management Zika virus into the Americas, because conventional
Dengue is caused by four dengue virus serotypes dengue serological assays, and even NS1 diagnostic
(DENV1–4), transmitted by the daytime-biting mosquitoes assays, are cross-reactive with Zika virus infections,

www.thelancet.com/infection Vol 17 March 2017 e88


Series

and patients often do not receive timely feedback for


Application and future priorities
decision-making that can affect case management and
Case management tools: diagnostics using direct assays during the viraemic phase (<5 days of illness) disease outcome. New information and communication
Viral RNA detection such as RT-PCR, real-time RT-PCR, Promising lower-cost molecular biological technology tools are being developed that improve work
and virus isolation are highly sensitive but expensive techniques are being developed,9 but require
further validation to be commercialised and information flows around time-sensitive disease
There exist numerous commercial ELISA kits and rapid More affordable NS1 assays are needed, and new
diagnosis and response, as well as outbreak prevention. A
diagnostic tests for NS1 (non-structural protein 1), but commercial assays need to continue to be major priority is integration of these tools into national
they have a wide range of sensitivity depending on evaluated by a panel of reference laboratories10 data systems, and involvement of all stakeholders from
dengue virus serotype, primary or secondary infection,
the beginning to ensure bidirectional flow of clinical and
and commercial brand, and are too expensive for public
sector use diagnostic information that benefits both surveillance
NS1 is present early in infection, and anti-dengue virus Current assays are of variable quality and are systems and patients alike.
IgM antibodies are detectable later in illness, making a expensive; thus, they need to be further developed Prognostic tests to detect biomarkers associated with
combination test a promising tool for dengue diagnosis to be used affordably in low-income and severe dengue, to enable better triage and treatment of
middle-income countries
patients, are not available in clinical settings. Clinical
Case management tools: prognostic tests for triage and treatment of patients with severe dengue
manifestations are currently the only characteristics used
Clinical manifestations are currently the only tool for Promising biomarkers include: innate immunity
predicting severe dengue disease gene expression profiles, specific cytokines, T-cell
for predicting severity of dengue disease. Recent studies
and B-cell activation markers, and complement have revealed quantifiable molecules and immune
activation,11 but they require development into signatures during the course of dengue virus infection;
validated assays translating these potential biomarkers into practical
Surveillance and outbreak response tools: serological assays and crowd-sourcing clinical tests requires substantial research and evaluation,
IgM capture ELISAs are widely available and are used by IgM seroconversion in paired acute-phase and especially in the context of vaccine rollout. Advances in
most public health systems; however, a single positive convalescent-phase (15–21 days after onset)
dengue virus-specific IgM is only a probable diagnosis serum samples is required for confirmation of technology that allow for multiple biomarker analysis
dengue virus infection, but paired samples are with small sample volumes will enable implementation
often not available of these assays in a clinical context in low-income and
Commercial IgM ELISAs and rapid diagnostic tests are New commercial IgM assays should be evaluated middle-income countries, and could aid clinical trials of
available, but are of variable quality by a panel of reference laboratories
vaccine or drug interventions.
Changes in IgG titre and in the IgG:IgM ratio are often Some kits need to be better validated and
detected by IgG capture ELISA to differentiate primary standardised. For example, kits are commercially
from secondary dengue virus infections; many kits are available to distinguish primary and secondary Surveillance, outbreak response, and
commercially available infections using a single IgG test during the acute sero-epidemiogical studies
phase; these should be interpreted with caution Most dengue-endemic countries rely on syndromic
There has been some success using crowd-sourcing Crowd-sourcing technologies for illness reporting dengue surveillance, with laboratory-based confirmation
technologies to monitor and control dengue need to be validated by laboratory confirmation of
outbreaks12,13(Google Dengue Trends, DengueMap, disease cause; these technologies should be paired of a subset of cases. Laboratory-enhanced sentinel
DengueChat) with community entomology efforts and be used surveillance systems provide more precise information
to enhance national surveillance to public health authorities on time, location, serotype,
Sero-epidemiological tools: more specific serological assays in post-convalescent phase or vaccinees and disease severity, thus enabling an earlier trigger for
Dengue virus infection in a population can be These methods are not as specific as neutralising interventions to mitigate outbreaks. Functional
measured using IgG ELISA, inhibition ELISA, or assays, but are easier to do at large scale laboratory-based surveillance systems will be crucial for
haemagglutination inhibition
deployment of vaccine trials and for eventual vaccine
Neutralisation assays are considered most specific and In secondary dengue virus infections,
can differentiate certain flaviviruses, including dengue cross-reactivity among the four serotypes impairs rollout. Serological assays that detect increases in dengue
virus serotypes 1–4 and Zika virus, in primary infections interpretation of neutralisation titres in a single virus-specific antibodies are widely used by most public
and >6 months after infection sample, although longitudinal annual samples health systems for confirmation of disease, and for
have enabled reconstruction of dengue virus
immune history14,15
differentiating primary from secondary dengue virus
infections (table 1). Sero-epidemiological studies can
Table 1: Tools for case management, surveillance, outbreak response, and sero-epidemiological studies monitor the overall prevalence of dengue virus infection
in a population, and are usually based on IgG ELISA,
making the interpretation of results more complex in inhibition ELISA, or haemagglutination inhibition
endemic areas. Neutralisation tests can be performed to assays, although more specific methods, such as
detect virus-specific neutralising antibodies and to neutralisation tests, are available.
determine the cause of flavivirus infection, but only Crucially, once dengue virus vaccines are widely
after about 6 months post-infection. distributed, serological assays will no longer distinguish
For diagnostics to have a true effect on disease outcome, immune responses due to dengue virus infection from
systems need to close the loop between the clinic, the vaccine-induced immunity. Furthermore, cross-reactivity
laboratory, and the patient. Other than rapid diagnostic with other flaviviruses with widespread circulation, such
tests, which are rarely used in the public sector in low- as Zika virus and West Nile virus, and national vaccination
income and middle-income countries, most diagnostic programmes against flaviviral diseases, such as yellow
information obtained at centralised laboratories is used to fever in Latin America and Japanese encephalitis in Asia,
inform national surveillance systems, whereas clinicians can confound interpretation of serological assays.

e89 www.thelancet.com/infection Vol 17 March 2017


Series

Serological methods to distinguish flavivirus infections dengue risk, possibly because of a false sense of
are urgently needed, particularly in secondary infections. protection and consequential absence of environmental
Crowd-sourcing of symptom data and community management activities.19
entomology efforts enable affected communities to gain Other approaches exist, such as biological control with
ownership of control strategies, and represent powerful organisms that prey upon the aquatic immature Aedes
sources of information for outbreak detection. Infor- spp—eg, small crustaceans and larvivorous fish21—and
mation and communication technologies and social biologically-derived insecticides, but more data are needed
networks can have an important role in disease, vector, to understand cost-effectiveness of large-scale imple-
and outbreak reporting, from the community to local and mentation. Trials of genetically modified mosquitoes as a
regional authorities. A handful of such technologies have population suppression strategy are ongoing, which
been used successfully in pilot implementations and involve engineering a lethal gene into mosquitoes to
within studies, including use of mobile phone tower ensure sterility after mating with wild mosquitoes.22 A
signals to track human movement for prediction of the new population replacement approach is underway in
most likely outbreak locations in Pakistan;12 online and several trial sites worldwide using mosquitoes infected
smartphone applications to report dengue mosquito with the wMel strain of the bacterium Wolbachia pipientis
breeding sites in Mexico,13 Nicaragua, and Colombia; and that shortens the mosquito lifespan and blocks virus
near real-time search-query data that estimate impending development, thus reducing dengue virus transmission.23
outbreaks globally (eg, Google Dengue Trends, The scale-up potential, the trade-off between the fitness
DengueMap). Although information gained by crowd- cost of these variants and their virus-blocking ability, and
sourcing needs to be validated by laboratory confirmation, the effect of introducing modified organisms into the
the power of these technologies, together with the ecosystem, need to be further evaluated.24 Finally,
strength of communities mobilised by knowledge, community-based approaches that include entomological
should be harnessed as integral aspects of national surveillance and evidence-based, active participation of
surveillance systems, and for documenting the effect of affected populations in source reduction, have been
vaccine introduction. shown to result in reduced household Aedes spp
entomological indices, dengue virus infection incidence,
Vector control and dengue disease.19,25 For vector control efforts to work,
Excellent vector control reviews have recently been they need to involve community participation and be
written;16 in this section, we discuss key points in the sustained year-to-year, during interepidemic periods, and
context of vaccination programmes. To date, preventing especially in high-risk locations.
or reducing dengue virus transmission has depended
entirely on controlling the mosquito vectors. Even after
Issues Research priorities
vaccine deployment, vector control will continue to be
part of control strategies to reduce disease risk and 1 Geographic dissemination of dengue virus is Develop models that accurately describe human
attributable to human movement, which movement and mosquito density to understand
burden. The concept of integrated dengue management functions by heterogeneous mixing27,28 different levels of disease risk within cities
is now widely accepted.17,18 2 Climate variables play a major part in Incorporate climate variables to improve prediction of
The recent chikungunya and Zika virus disease region-wide epidemics29,30 epidemics so that governments can prioritise dengue
epidemics have placed vector control at the forefront of control efforts
public health responses, but the same challenges that 3 The demographic transition affects basic Identify the role of population age structure on
reproduction number, leaving more individuals dengue virus transmission in additional settings
existed decades ago remain today. Low-income and dengue virus-naive until older ages31,32
middle-income countries mostly focus their efforts on
4 Homologous reinfection can occur, and infected Incorporate homologous reinfection and
source reduction (elimination of vector breeding individuals can have high viraemia, suggesting heterologous inapparent infections into
containers), environmental management, and larvicide they might contribute to transmission;33 epidemiological models to more accurately estimate
abatement of water storage receptacles, and they use heterologous asymptomatic infections dengue virus exposure and transmission
contribute to transmission, since these
chemical fogging as an outbreak response. Chemicals individuals can have high viraemia and have
in thermal fogs or ultra-low-volume aerosols target greater movement than individuals who are
adult mosquitoes and only work at a very local level. unwell34
These chemicals need to be repeatedly applied, which 5 Frequent dengue virus exposure in Take into account frequent dengue virus re-exposure
high-transmission settings might modify in maintaining long-term protection and attenuating
has led to the development of resistance as well as immunity and transmission in dengue transmission in the dengue virus-immune
environmental and health effects of unknown virus-immune individuals35,36
magnitude. The widely applied organophosphate 6 Worldwide, other flaviviruses circulate in Estimate changes in dengue virus transmission in
larvicides, such as temefos, are also of questionable dengue-endemic areas—eg, Japanese populations also infected by related flaviviruses
efficacy and pose potential health risks.19,20 Results from encephalitis in southeast Asia,37 yellow fever virus
and yellow fever vaccination in Africa and the
a recent large cluster randomised controlled trial Americas, and now Zika virus in the Americas and
showed that temefos had no effect in reducing Asia
household entomological indices or incidence of dengue
Table 2: Issues and research priorities for modelling dengue transmission
virus infection, and was associated with increased

www.thelancet.com/infection Vol 17 March 2017 e90


Series

New guidelines based on validated strategies and clade, or strain of the first infection), in relation to the
models, which expand the toolbox available to govern- second infecting strain, can also affect infection outcome
ments, are urgently needed. The effect that vector control (inapparent vs symptomatic) and disease severity at both
interventions have on health and disease burden needs be the individual and population level.39,47–50 Thus, a priority is
systematically investigated. Furthermore, consensus on estimating the effect of genetic variation on the breadth of
the timing and nature of a combination of vector control the immune response induced by natural infection and
approaches with dengue vaccines should be pursued. vaccination,51 and the resulting protection against diverse
Scaling up successful pilots and trials is the next frontier, dengue virus strains. Another recent discovery is that
and models informing this process are essential.26 Despite genetic differences alter the exposed epitopes on the virion
the challenges, vector control will need to be included in and thus affect neutralisation profiles.52,53 Some such
any integrated strategy for dengue prevention. aminoacid variants are seen in highly laboratory-adapted
strains that are parent strains for current vaccines;54
Modelling dengue transmission therefore, how these substitutions affect the immuno-
Modelling dengue transmission enables the evaluation genicity of vaccine strains compared with natural isolates
of the benefits and limitations of different dengue control needs to be investigated.
methods; this is increasingly important as interventions Clade replacement is a characteristic feature of
such as vaccine and new vector control strategies come dengue virus evolution.55 The relative contribution of
closer to implementation (table 2). Mathematical models intrinsic fitness differences,46,47,56 escape from
have been developed to identify determinants of the neutralisation,57–59 cross-serotype enhancement,53,60,61 or
oscillations of dengue virus serotypes in endemic stochastic effects (eg, climate, geography, genetic
settings, including the parts played by the mosquito bottlenecks)57,62 remain poorly understood. Estimating
vector and by immune enhancement from previous the contribution of these factors to dengue virus
exposure to heterologous dengue virus strains.38 Recent evolution could enable prediction of the strength or
research using statistical models has shown that dengue severity of epidemics. Tetravalent dengue vaccines
epidemic size and disease severity could be affected by induce imbalanced responses against dengue virus
human movement, changes in population age structure, serotypes 1–4 in some individuals,63,64 as well as different
and regional climate variables;27–32 further work is needed combinations of type-specific and cross-reactive
to better understand these effects and to build predictive neutralising antibodies, raising the question of whether
models for government preparedness on local and vaccination could create a level of population immunity
regional levels. Recent observations of homologous that facilitates evolution of certain lineages that better
dengue virus reinfection,33,39 viraemic heterologous escape host immunity.
asymptomatic infections,34 cross-reactivity with other
flaviviruses,37 and frequent dengue virus exposure35,36 Immune responses in the epidemiological context
should be further studied in an epidemiological Post-primary infections
modelling framework to understand their role in dengue The interval of time between first and second dengue
virus immunity and dengue virus transmission. In light virus infection modulates infection and disease
of the different degrees of vaccine efficacy in previously outcome (figure). The average period of cross-
dengue virus-naive and dengue virus-exposed protection is 1·6–2 years against symptomatic dengue
individuals,40–42 models are essential to estimate vaccine virus infection and 2·6 years against severe disease;15,65,66
efficacy stratified by immune status, and to explore these observations are consistent with long-term
effects of vaccination on dengue virus transmission.43 follow-up data from vaccine trials.40 Hypotheses to
Such models should continue to be developed in concert explain these observations include waning of cross-
with vaccination efforts, and should account for serotype neutralising antibodies, epidemic force,
heterogeneity in previous dengue virus exposure within increased risk of being exposed to a different serotype,
countries and across regions. or other immune mechanisms such as waning T-cell
protection.15,35,65–67 In the absence of dengue virus re-
The virus: serotypes, genotypes, and strains exposure, neutralising antibody titres decay rapidly
Dengue virus serotypes, genotypes, and clades can differ between 1–6 months, then decay gradually; however, in
in intrinsic virulence and epidemic capacity.14,35,44–46 A better endemic settings, after the initial decrease, the
understanding of the genetic basis for virulence and neutralisation titres remain relatively stable for years
enhanced replication in mosquitoes and people would after primary infection, with some individuals
improve our ability to predict epidemics that pose a greater exhibiting decay and others boosts.35,36,68,69 The responses
risk of disease. A further question is whether epidemic of anti-dengue virus neutralising antibodies are also
force (the symptomatic:inapparent infection ratio) affects thought to become more type-specific for months and
the threshold of neutralising antibodies or other immune possibly years after primary infection.70–72 However,
correlates that protect against subsequent infection.35 many individuals have broad neutralisation profiles 1
Previous immunity (determined by the serotype, genotype, year after infection in non-endemic settings,73,74 and

e91 www.thelancet.com/infection Vol 17 March 2017


Series

after many years in endemic settings.35,36,75 Comparing The study of symptomatic homologous infection will
longitudinal patterns of neutralisation titres in non- help us understand why type-specific protection fails,
endemic versus endemic settings is important for which might provide insight into cases of low vaccine
establishing the decay rate and cross-reactivity of efficacy.
neutralising antibodies over time.
Boosting
Post-secondary infections Dengue virus re-exposures might boost neutralising
After a secondary infection with a different dengue virus antibody titres in dengue virus-immune individuals,
serotype, the neutralising antibody response becomes providing them with longer protection against a sub-
broadly neutralising and is thought to reduce incidence sequent symptomatic infection. Vaccine trials show
of subsequent severe disease. Symptomatic third and boosted neutralising antibody responses on repeat
fourth dengue virus infections do occur, but are rarely vaccination (although not necessarily improvements in
severe.48,50,76 Understanding the properties of post- efficacy);80 experimentally inoculated non-human
secondary immunity, for instance, the cross-reactive primates challenged with the homologous strain a year
envelope dimer epitope (EDE),77 will improve under- later display boosted antibody responses;81 and
standing of what constitutes a protective multivalent neutralisation titres in children in endemic areas, on
response. average, increase in magnitude between first and
second infection.35,36 It is essential to determine whether
Homologous symptomatic reinfection natural boosting in endemic areas modifies an
Type-specific neutralising responses were thought to individual’s subsequent infection or disease risk, and to
provide sterilising immunity against homologous re- evaluate the implications for vaccine deployment—
infection;78,79 however, evidence now exists that homo- does natural boosting improve long-term vaccine
logous reinfection can lead to symptomatic disease.33,39 efficacy?

Primary infection Secondary infection

DENV

MBC

DENV1 DENV2
MBC SLPC
SLPC

Homotypic LLPC
B cell T cell reinfection?

Determinants of infection MBC stimulation LLPC repopulation?


outcome (S:I):
• B cells/antibodies?
• T cells? Potent cross-reactive neutralising
• Innate immunity? antibodies:
• Epidemic force? • How long-lasting?
• Lineage origin?
• Epitopes?

Neutralising antibodies:
• Decaying in magnitude?
Immune response

• Increasing in type-specificity? Risk of third and


• Critical protective epitopes? fourth symptomatic
infections?
Determinant of disease severity:
Determinants
CD8-posit T cells?
• CD8-positive
anti
• Other antibody mechanisms
Time between infections and disease outcome: (complem
(complement)?
• Waning cross-protective antibodies, T–cell response, dete
• Host determinants?
other immune mechanism(s)? gene
• Viral genetics?
• Boosting: homotypic or heterotypic exposure? • NS1?
• Epidemic force (S:I ratio) or periodicity?

Time

Figure: Key questions about the adaptive immune response following primary and secondary dengue virus infection
The black line represents the level of individual host immunity (y-axis) over time (x-axis). DENV=dengue virus. EDE=envelope dimer epitope. LLPC=long-lived plasma
cell. MBC=memory B cell. NS1=non-structural protein 1. S:I ratio=symptomatic to inapparent infection ratio. SLPC=short-lived plasma cell.

www.thelancet.com/infection Vol 17 March 2017 e92


Series

Immune correlates of protection and risk correlate of protection against dengue virus, only recently
Neutralising antibodies have results of studies shown a significant association
A crucial metric of protection is the potency of between the quantity of cross-reactive pre-infection
neutralising antibodies against future infecting strains; neutralising antibody titres and reduced risk of
thus, neutralisation assays are a key tool for decision- symptomatic secondary infection.35,89,90
making by clinicians, epidemiologists, vaccine Aside from the quantity of neutralising antibodies,
developers, and policy makers. However, current assays more attention has recently been focused on their
do not adequately capture the full complexity of the quality: type-specificity versus serotype cross-reactivity
neutralising antibody response, and results are highly and epitope repertoire. To date, most antibodies
variable across laboratories. Moreover, the results of generated after primary dengue virus infection have
recent clinical trials of Sanofi’s tetravalent dengue been cross-reactive and weakly neutralising.91 Many
vaccine showed that quality as well as quantity of bind around the fusion loop region on envelope protein
neutralisation titres need to be taken into account.63 domain II (EDII); nonetheless, some antibodies that
Neutralisation titres vary greatly across laboratories bind this region are potently neutralising.92,93 After both
and between methods, with assay parameters such as primary and secondary infection, the most potently
virus preparation (maturation state, cell source), virus neutralising antibodies are virion-specific and often
strain, complement, cell type, DC-SIGN (CD209 antigen) bind across dimers, contacting up to three monomers
expression, use of plasma versus serum samples to simultaneously, thus throwing a wrench into the dengue
measure antibody neutralisation, presence of EDTA virus E protein machinery that successfully prevents
(edetic acid) in plasma, percent plaque or immunofocus fusion.77,94–97 These antibodies bind multiple sites on the
reduction, and method of measurement of infected cells, E protein, including regions of EDIII, the fusion loop,
influencing outcome.82,83 The maturation state of the and the hinge of EDI/II.95 New tools are becoming
virus stock can alter how well the virus is neutralised— available to dissect the repertoire of polyclonal sera,
for instance, antibodies that recognise the fusion loop or using depletion methods and epitope-transplanted
prM protein only bind to immature virions, where these recombinant viruses;52,98,99 applying these to better
epitopes are exposed.83,84 Dengue virus virions are now understand the antibody response and potential
thought to be mosaic, with some regions of the virus immune correlates in dengue virus natural infections
smooth and mature, and others spiky and immature.85 and vaccines is an area of active research.
Even in the mature form, dengue virus particles undergo
so-called virus breathing—whereby viruses explore Antibody-dependent enhancement
multiple structural conformations—with envelope A major concern for dengue vaccines is that they might
protein monomers sampling a range of structures induce enhancing dengue virus antibodies that could
between the mature and pre-fusion state.54 Higher increase risk of severe disease in vaccinated individuals.
(eg, febrile) temperatures and longer incubation periods Antibody-dependent enhancement is posited to occur
enable antibodies access to otherwise concealed cryptic when antibodies to a previous dengue virus infection
epitopes as the virions breathe, affecting neutralisation recognise but do not neutralise a subsequent infection
titres.54,86 Finally, the virus strain chosen can also impart with a different serotype, and the resulting immune
variability, because some strains are better or worse complexes facilitate virus entry into target Fcγ receptor-
neutralised than others.75,87 bearing cells, leading to higher viral load and activation of
The cell type used as a substrate for measuring T cells that secrete tumour necrosis factor (TNF)α and
neutralisation remains a critical concern. The validated other vasoactive cytokines. Passively transferred
cell line for vaccine developers is Vero, derived from heterotypic antibodies increase viremia levels in rhesus
green monkey kidneys.88 Neutralisation assays done on monkeys,100,101 and induce lethal vascular leak syndrome
Vero cells appear to detect only type-specific neutralising with an otherwise sublethal dose of dengue virus in
responses, whereas other cell lines, including those interferon receptor-knockout mice.102,103
expressing DC-SIGN, can capture type-specific and In human beings, studies of the incidence of severe
cross-reactive neutralising antibody responses.83 Develop- dengue in infants show a strong correlation between
ment of assays with biologically relevant substrates is peak incidence of dengue haemorrhagic fever/dengue
paramount to better understand immune correlates and shock syndrome, age of the infants, decay of maternal
neutralisation titres. neutralising antibodies, persistence of anti-dengue virus
The variability in neutralisation titres, and thus maternal IgG antibodies, and fold-enhancement by neat
generalisability of what titre corresponds with protection antisera on Fcγ receptor-bearing cells.104–109 Fold-
under all assay conditions, has been a point of contention enhancement titres measured on primary human
and has complicated studies of immune correlates in monocytes can also distinguish between asymptomatic
natural infections and vaccines. An optimal neutralisation and severe dengue virus serotype 2 infections in
assay remains the holy grail for the dengue field. children.110 Heterotypic immunity is a risk factor for
Although neutralisation titres have long been treated as a severe dengue, consistent with the hypothesis that

e93 www.thelancet.com/infection Vol 17 March 2017


Series

enhancing antibodies could contribute to severe disease.44 non-structural proteins,125 vaccines that do not include
The initial observation of higher rates of hospitalised dengue virus non-structural proteins might miss an
dengue cases in young recipients of vaccines compared important immune mechanism for preventing severe
with controls in year 3 follow-up data of clinical trials, disease.126 By contrast, tetravalent vaccination that
has raised concerns about vaccine-induced antibody- includes non-structural proteins from multiple dengue
dependent enhancement of infection.40 However, some virus types induces a multifunctional CD8-positive T-cell
other studies did not observe an association between response that is directed toward epitopes conserved
enhancement titre and disease severity.107,111,112 Further- among serotypes, potentially providing CD8-positive, T cell-
more, there is still controversy about which cell substrate mediated control of infection.120
to use for the in-vitro enhancement assay.112,113 One CD4-positive T cells indirectly control dengue virus
research priority is to establish whether disease severity infection by facilitating B and CD8-positive T-cell activation
is mediated by the degree of antibody-dependent and memory, and by secreting inflammatory cytokines.67
enhancement. Enhancing antibodies, in the absence of Certain CD4-positive T-cell responses are thought to be a
sufficient neutralising antibodies, are possible con- signature for dengue haemorrhagic fever pathogenesis;127
tributing factors, but symptomatic infections only however, this relation has not been tested to predict disease
progress to severe disease in the context of other host- outcome. In mouse models, memory CD4-positive T cells
related and virus-related determinants. mediate protection in secondary cases.128 Anti-dengue virus
CD4-positive T cells in people might also directly control
B cells dengue virus infection by killing infected cells.129 The CD4-
The B-cell repertoire after dengue virus infection is positive T-cell epitopes identified to date focus on structural
composed of long-lived plasma cells and memory B cells. and non-structural proteins, with additional charac-
After primary infection, individuals are thought to develop terisation ongoing in human populations.67 An important
a type-specific neutralising response over time in non- area of research is the role of memory CD4-positive T cells
endemic regions.70–72 One hypothesis is that this occurs in that are induced by natural infection and vaccination in the
the absence of reinfection, whereby high-affinity memory stimulation of protective antibody-mediated immunity and
B cells naturally replenish the long-lived plasma cell robust CD8-positive T-cell responses during secondary
population.114 An alternative hypothesis is that long-lived infection in human beings.
plasma cells are long-lived, but not life-long, and that the
long-lived plasma cell-derived serum neutralisation titre Innate immunity
decays over time in a pathogen-specific way.115–117 An Interferons constitute a powerful antiviral response, and
essential priority is to determine whether maintenance of dengue virus has evolved numerous mechanisms to
long-lived type-specific neutralising antibodies requires evade the human innate immune response, targeting
re-exposure to dengue virus. After secondary infection, multiple stages in the interferon signalling pathway.130
potent cross-reactive neutralising antibodies have been Gene expression studies of dengue cases of varying
identified,77,118 but the origin of these B cells remains poorly severity highlighted differences according to day of
understood. A major priority is to identify these antibodies illness, and revealed shock signatures—dengue shock
in post-secondary infection peripheral blood mononuclear syndrome cases had reduced interferon-stimulated genes
cells, and then to trace them back to identify their lineage and increased mitochondrial function.131,132 Another
in samples collected after previous infection(s). Another notable observation was that dengue virus induces CD14-
outstanding question is whether dengue virus in human positive, CD16-positive monocytes, which can promote
beings resembles West Nile virus in mice, in which long- B cell differentiation into plasmablasts and antibody
lived plasma cells expressed the highest-affinity receptors secretion.133 Current efforts are focused on generating
and memory B cells had lower-affinity receptors, but network models of dengue virus infection, and on
recognised a wider range of epitopes.119 correlating innate immune signatures with adaptive
immune responses.
T cells
The importance of T cells in protection against, and Other antibody effector functions
pathogenesis of, severe dengue remains an active area of Few studies have examined other antibody attributes in
research. Cross-reactive T cells have long been postulated human populations. Regarding antibody isotypes, IgG1
to have a role in dengue immunopathogenesis, but have and IgG3 differed between cases of dengue fever and
also recently been shown to be protective as well.120,121 dengue haemorrhagic fever, whereas IgG4 and IgA were
Generally, CD8-positive T cells contribute to the antiviral more common in cases of dengue shock syndrome.134
response by directly killing infected cells and secreting Antibody-dependent cell-mediated cytotoxicity in pre-
interferon-γ and TNFα. Recent research indicates that infection samples was associated with reduced viraemia
CD8-positive T cells can protect against secondary during secondary infection,135 but others found antibody-
dengue virus infection in mice and human beings.122–124 dependent cell-mediated cytotoxicity activity in acute
Because CD8-positive T cells primarily target serum samples from dengue haemorrhagic fever and

www.thelancet.com/infection Vol 17 March 2017 e94


Series

dengue shock syndrome cases, but not dengue fever Pathogenesis


cases.136 More research is needed to explore these Host genetics, dengue virus immune history, infection
potential additional immune correlates. sequence with particular dengue virus serotypes and
strains, and viral genetics modulate the immune
Issues Research priorities response and affect disease outcome. A dominant theory
has been that immunopathogenic mechanisms result in
1 Some immune biomarkers might be predictive of Develop prognostic tests for research and clinical trials
disease severity11 a so-called cytokine storm that leads to vascular leak and
2 Dengue vaccine rollout will make distinguishing Develop serological methods to distinguish natural
thus contributes to severe dengue disease in secondary
previous dengue virus infection from and vaccine-induced immunity; develop new infections.121 However, severe disease does develop in
vaccine-induced immunity difficult; other serological assays to differentiate acute infections with some instances after primary dengue virus infection, and
flaviviruses, such as Zika virus, confound dengue virus from Zika virus and other flaviviruses many patients with high viraemia recover from dengue
serological diagnostic methods
fever without developing plasma leakage.45
3 Crowd-sourcing technologies used by Validate and incorporate crowd-sourcing for
empowered communities have great potential surveillance systems and documenting the effect of
Complement activation, which coincides with the
for outbreak detection vaccine introduction timing of plasma leakage, can be triggered by dengue
4 Vaccines can have different efficacy in previously Stratify vaccine efficacy by immune status,43 and virus-antibody complexes.137 A hypothesis based on
dengue virus-naive and dengue virus-immune update and develop models with additional vaccine molecular mimicry posits that some dengue virus-
individuals40–42 trial and follow-up data induced antibodies can cross-react with host proteins
5 Dengue vaccines might protect against disease Use models to compare effects of full vs partial such as plasminogen, thrombin, and platelets.138–142 More
but not transmission protection against transmission following vaccination research is needed to understand the potential role of
6 Vaccine-induced neutralising antibody responses Study how immune responses induced by vaccination these pathways in dengue pathogenesis.
are often higher to vaccine parent strains than protect against diverse strains
genetically distinct isolates51
Dengue virus NS1 contributes to vascular leak via both
cytokine-dependent and cytokine-independent mecha-
7 Aminoacid substitutions in laboratory-adapted Estimate how substitutions in laboratory-adapted
or vaccine parent strains can alter the exposed strains and vaccine parent strains affect nisms. In one study, purified NS1 directly activated mouse
epitopes54 immunogenicity, compared with immunogenicity of macrophages and human peripheral blood mononuclear
natural isolates cells via Toll-like receptor 4 (TLR4), leading to induction
8 Tetravalent dengue vaccines can induce Evaluate whether vaccination could increase the risk of and release of proinflammatory and vasoactive cytokines
unbalanced responses or different combinations dengue virus lineages evolving to escape host
and chemokines. Both NS1-mediated activation of
of type-specific and cross-reactive antibodies immunity
against dengue virus serotypes 1–463,64 peripheral blood mononuclear cells and NS1-induced
9 Homologous reinfection can lead to Understand why type-specific protection fails, to
permeability in vitro were inhibited by a TLR4 antagonist
symptomatic disease.33,39 provide potential insight into low vaccine efficacy and by anti-TLR4 antibodies.143 Results of a parallel study
10 Frequent dengue virus re-exposures may boost Evaluate whether natural boosting improves showed that recombinant NS1 directly induced vascular
levels of neutralising antibody titres immunity and long-term vaccine efficacy leak and a lethal, NS1-mediated disease in a mouse model;144
11 Neutralisation titres are highly variable under Define the most crucial variables that modulate these pathogenic effects were blocked by NS1-immune
distinct assay conditions82,83 neutralisation assays and identify neutralisation tests polyclonal mouse serum and anti-NS1 monoclonal
that correlate with vaccine-induced protection
antibodies, and mice immunised with NS1 from each of
12 New tools are available to dissect the repertoire Apply these new tools to assess the quality of the the four dengue virus serotypes were protected against
of polyclonal sera52,98,99 neutralising antibody response after natural infection
and vaccination
lethal DENV2 challenge.144 NS1 also induced hyper-
permeability in human endothelial cell monolayers,
13 CD8-positive T cells primarily target dengue virus Determine the role of non-structural proteins in
non-structural proteins120,125 inducing protective CD8-positive T cells in vaccine independently of cytokines, via disruption of the glycocalyx
recipients layer that lines the endothelium.145 These findings identify
14 CD4-positive T cells facilitate B cell and Determine the role of memory CD4-positive T cells in new potential targets for dengue therapeutics and support
CD8-positive T-cell activation and memory120 inducing protective natural and vaccine-induced inclusion of NS1 in dengue vaccines.
immunity
15 Mice immunised with non-structural Investigate whether anti-NS1 immunity after dengue Dengue vaccines
protein 1 (NS1) are protected against lethal vaccination contributes to protection against
dengue virus challenge, and anti-NS1 antibodies endothelial hyperpermeability and severe disease Of the dengue vaccines in development, live-attenuated
prevent vascular leak and endothelial vaccines are the most advanced, with three candidates in
hyperpermeability144 phase 2/3 (NIH TV003/TV005 and Takeda TDV vaccines)
16 Following CYD (yellow fever/dengue chimeric Determine whether the risk of hospitalisation for or phase 4 (Sanofi Dengvaxia) trials. Other approaches in
vaccine) vaccination, young vaccine recipients dengue in young vaccinated individuals is mediated by preclinical or early clinical development include non-
had a higher risk of hospitalisation for dengue age or dengue virus-naive status,149 and whether risk is
than controls in follow-up data of clinical trials40 mediated in part by antibody-dependent replicating or single-replication vectored vaccines
enhancement packaged in dengue virus structural proteins, purified,
17 Neutralising antibody titres might not be the Study diverse measures of the immune response in inactivated virus vaccines, subunit-based vaccines
only correlate of protection150 relation to disease outcome for each vaccine (eg, EDIII or NS1 protein), and DNA-based vaccines.146
18 Improved correlate of protection measures will Establish biorepositories of vaccine samples for future Novel approaches being explored include scaffolding
be developed over time research complex epitopes, such as the cross-neutralising EDE
Table 3: Issues and research priorities in the context of dengue vaccination rollout and evaluation epitope, bivalent vaccines that present two potent
neutralising type-specific epitopes simultaneously, and

e95 www.thelancet.com/infection Vol 17 March 2017


Series

Panel: Major public health and research priorities


• Develop affordable, easy-to-perform, acute-phase • Understand why type-specific protection can fail and result
diagnostic and prognostic tests to enable timely in homologous reinfection, and further study whether
evidence-based case management, including rapid feedback frequent dengue virus exposure maintains long-term
from central national laboratories to clinics to directly protective immunity.
improve patient treatment. • Establish why post-secondary responses induce multivalent
• Promote laboratory-enhanced sentinel surveillance to protective immunity, including identifying B cells that
enable early introduction of interventions to mitigate produce potently neutralising antibodies, and characterising
dengue outbreaks, which can be further supplemented with their lineage.
information from crowd-sourcing technologies that enable • Determine to what degree disease severity is mediated by
communities to be directly involved in control efforts. antibody-dependent enhancement.
• Generate new guidelines for vector control, based on • Identify the role of memory CD4-positive T cells in stimulating
validated strategies proven to reduce dengue virus infection protective antibody-mediated immunity and robust
and disease, including community-based approaches for CD8-positive T-cell responses during secondary infection.
source reduction, that governments can use to improve • Investigate protective effector functions of anti-dengue
control programmes. virus antibodies.
• Incorporate recent insights into the immune responses to • Further study the role of the non-structural protein 1 (NS1)
natural dengue virus infection and vaccination, including the in dengue pathogenesis to develop new targets for
differences observed between dengue virus-naive and dengue therapeutics and protective dengue vaccines.
virus-exposed individuals in vaccine efficacy, to develop • Clearly convey to public health practitioners in
increasingly sophisticated models of dengue immunity. dengue-affected areas that naive dengue status is likely to
• Understand the genetic basis for virulence, enhanced be a major risk factor for the increase in severe disease
replication in mosquitoes and human beings, observed in young children in the Sanofi phase 3 clinical
immunogenicity, and neutralising properties of genetically trial, so that public health practitioners can decide whether
diverse dengue virus strains. the vaccine should be used in low-endemic settings and in
• Estimate decay rates of cross-reactive neutralising areas with only one circulating dengue virus serotype.
antibodies after primary infection in both endemic and
non-endemic settings.

recombinant viruses on which epitopes recognised by To date, no published vaccine correlate of protection
enhancing antibodies are masked.99,147,148 against dengue exists. Results of recent studies of natural
Remaining knowledge gaps and research priorities in dengue virus infections show a significant association
relation to dengue vaccines are summarised in table 3. between the quantity of cross-reactive pre-infection
Additionally, some questions were raised by the first neutralising antibody titres and reduced risk of sympto-
phase 3 efficacy trials and long-term follow-up studies. matic secondary infection.35,89,90 However, neutralising
The year 3 follow-up data from the Sanofi phase 3 antibody titres could be a correlate of protection, but not a
clinical trial showed an increased risk of hospitalisation mechanistic correlate of protection.150 Establishing both
in young vaccinated people versus control individuals.40 mechanistic and non-mechanistic correlates of protection
Furthermore, data for one of the phase 3 clinical trials will require examining a suite of measures of the immune
showed no statistically significant protective effect of response in relation to disease outcome for each vaccine.
vaccination in naive individuals younger than 9 years.40 Collecting and maintaining sample banks or bio-
In discussing these results, the vaccine developers posit, repositories for continued monitoring of vaccines and
among other hypotheses, that primary vaccination in
young individuals places them at risk of a severe
infection sooner than the placebo group, which will Search strategy and selection criteria
eventually catch up over time.149 We identified articles published in English and Spanish by searching PubMed for the section
Whether young age or naive status is the greater risk headings: “case management”, “surveillance”, “outbreak response”, “epidemiology”, “vector
factor is a crucial remaining question, as is the effect of control”, “modeling”, “transmission”, “genotypes”, “evolution”, “neutralisation”, “antibody-
potential vaccine distribution in highly-endemic versus dependent enhancement”, “B cell”, “CD4+ T cell”, “CD8+ T cell”, “innate immunity”,
low-endemic settings, or in areas with only one “pathogenesis”, and “dengue vaccines”. These articles were reviewed for relevant references.
circulating dengue virus serotype. Another question is Historical articles describing major discoveries, recent studies (published in the past
the quality of the immune response after primary 10 years), and studies covering distinct topics (eg, infant and paediatric cohort studies for
vaccination in naive individuals, compared with a understanding correlates of protection) were selected for review. The search was conducted
primary natural dengue virus infection, and the risk of between January 11, 2016, and March 1, 2016.
subsequent severe infection.

www.thelancet.com/infection Vol 17 March 2017 e96


Series

research is of crucial importance for identifying dengue 10 Hunsperger EA, Yoksan S, Buchy P, et al. Evaluation of commercially
virus correlates of protection. available diagnostic tests for the detection of dengue virus NS1
antigen and anti-dengue virus IgM antibody. PLoS Negl Trop Dis 2014;
8: e3171.
Conclusion 11 John DV, Lin Y-S, Perng GC. Biomarkers of severe dengue
We have reviewed progress in diagnostics, clinical disease—a review. J Biomed Sci 2015; 22: 83.
12 Wesolowski A, Qureshi T, Boni MF, et al. Impact of human mobility
research, epidemiology, entomology, virology, immuno- on the emergence of dengue epidemics in Pakistan.
logy, pathogenesis, and vaccine development, as well as Proc Natl Acad Sci USA 2015; 112: 11887–92.
the results of multiple high-quality clinical studies and 13 Lozano-Fuentes S, Wedyan F, Hernandez-Garcia E, et al.
Cell phone-based system (Chaak) for surveillance of immatures
field sites, in the context of understanding natural of dengue virus mosquito vectors. J Med Entomol 2013;
dengue virus infections and current and future 50: 879–89.
vaccination efforts. In addition, we have highlighted 14 Morrison AC, Minnick SL, Rocha C, et al. Epidemiology of dengue
knowledge gaps (panel) and the urgent need for virus in Iquitos, Peru 1999 to 2005: interepidemic and epidemic
patterns of transmission. PLoS Negl Trop Dis 2010; 4: e670.
translational research. With the recent licensure of the 15 Montoya M, Gresh L, Mercado JC, et al. Symptomatic versus
first dengue vaccine and several others entering phase inapparent outcome in repeat dengue virus infections is
3 trials, as well as the current Zika virus pandemic, influenced by the time interval between infections and study year.
PLoS Negl Trop Dis 2013; 7: e2357.
researchers, funders, vaccine developers and public 16 Achee NL, Gould F, Perkins TA, et al. A critical assessment of
health professionals have the responsibility to join vector control for dengue prevention. PLoS Negl Trop Dis 2015;
efforts to focus research on key questions that will 9: e0003655.
17 PAHO. State of the art in the prevention and control of dengue
ensure that populations living in flavivirus-affected in the Americas. Washington, DC: Pan American Health
areas have access to cost-effective and accurate Organization, 2014.
diagnostics, responsive surveillance systems, and the 18 Gubler DJ. The partnership for dengue control—a new global
alliance for the prevention and control of dengue. Vaccine 2015;
safest and most effective treatments and vaccines 33: 1233.
possible. 19 Andersson N, Nava-Aguilera E, Arosteguí J, et al. Evidence based
Contributors community mobilization for dengue prevention in Nicaragua and
All authors contributed to the report design, literature review, writing, Mexico (Camino Verde, the Green Way): cluster randomized
controlled trial. BMJ 2015; 351: h3267.
revision, figures, and tables.
20 George L, Lenhart A, Toledo J, et al. Community-effectiveness of
Acknowledgments temephos for dengue vector control: a systematic literature review.
The authors thank Henry Puerta Guardo and Paulina Andrade for PLoS Negl Trop Dis 2015; 9: e0004006.
contributions to the figure. Funding support was from the National 21 Hurst TP, Kay BH, Brown MD, Ryan PA. Melanotaenia duboulayi
Institutes of Health (NIH), National Institute for Allergy and influence oviposition site selection by Culex annulirostris (Diptera:
Infectious Diseases grant P01 AI106695 (EH). Culicidae) and Aedes notoscriptus (Diptera: Culicidae) but not
Culex quinquefasciatus (Diptera: Culicidae). Environ Entomol 2010;
Declaration of interests 39: 545–51.
LCK, JC, and EH report grants from the NIH. JC and EH report grants 22 Winskill P, Carvalho DO, Capurro ML, Alphey L, Donnelly CA,
from the Bill & Melinda Gates Foundation, the Carlos Slim Health McKemey AR. Dispersal of engineered male Aedes aegypti
Institute, and the UBS Optimus Foundation, during the study. EH mosquitoes. PLoS Negl Trop Dis 2015; 9: 1–18.
reports personal fees from the Sanofi Pasteur Scientific Advisory Board, 23 Hoffmann AA, Montgomery BL, Popovici J, et al.
and a grant from Takeda for laboratory analysis of vaccine samples, Successful establishment of wolbachia in aedes populations to
outside the submitted work. suppress dengue transmission. Nature 2011; 476: 454–57.
References 24 Nguyen TH, Nguyen H Le, Nguyen TY, et al. Field evaluation of
1 Bhatt S, Gething PW, Brady OJ, et al. The global distribution and the establishment potential of wMelPop wolbachia in Australia
burden of dengue. Nature 2013; 496: 504–07. and Vietnam for dengue control. Parasit Vectors 2015; 8: 563.
2 Radke EG, Gregory CJ, Kintziger KW, et al. Dengue outbreak in 25 Arunachalam N, Tyagi BK, Samuel M, et al. Community-based
Key West, Florida, USA, 2009. Emerg Infect Dis 2012; 18: 135–37. control of Aedes aegypti by adoption of eco-health methods in
Chennai City, India. Pathog Glob Health 2012; 106: 488–96.
3 Schaffner F, Mathis A. Dengue and dengue vectors in the WHO
European region: past, present, and scenarios for the future. 26 Ferguson NM, Kien DTH, Clapham H, et al. Modeling the
Lancet Infect Dis 2014; 14: 1271–80. impact on virus transmission of wolbachia-mediated blocking of
dengue virus infection of Aedes aegypti. Sci Transl Med 2015;
4 WHO. Global strategy for dengue prevention and control. Geneva: 7: 279ra37.
World Health Organization, 2012.
27 Reiner RC, Stoddard ST, Scott TW. Socially structured human
5 Shepard DS, Undurraga EA, Betancourt-Cravioto M, et al. movement shapes dengue transmission despite the diffusive effect
Approaches to refining estimates of global burden and economics of mosquito dispersal. Epidemics 2014; 6: 30–36.
of dengue. PLoS Negl Trop Dis 2014; 8: e3306.
28 Perkins TA, Garcia AJ, Paz-Soldan VA, et al. Theory and data for
6 GBD 2015 DALYs and HALE Collaborators. Global, regional, and simulating fine-scale human movement in an urban environment.
national disability-adjusted life-years (DALYs) for 315 diseases and J R Soc Interface 2014; 11: 20140642.
injuries and healthy life expectancy (HALE), 1990–2015: a systematic
analysis for the Global Burden of Disease Study 2015. Lancet 2016; 29 Rabaa MA, Simmons CP, Fox A, et al. Dengue virus in sub-tropical
388: 1603–58. northern and central Viet Nam: population immunity and climate
shape patterns of viral invasion and maintenance.
7 WHO. Dengue haemorrhagic fever: diagnosis, treatment, prevention PLoS Negl Trop Dis 2013; 7: e2581.
and control, 2nd edn. Geneva: World Health Organization, 1997.
30 van Panhuis WG, Choisy M, Xiong X, et al. Region-wide
8 WHO and TDR. Dengue: guidelines for diagnosis, treatment, synchrony and traveling waves of dengue across eight countries in
prevention and control. Geneva: World Health Organization, Southeast Asia. Proc Natl Acad Sci 2015; 112: 13069–74.
2009.
31 Cummings DAT, Iamsirithaworn S, Lessler JT, et al. The impact of
9 Teoh BT, Sam SS, Tan KK, et al. Early detection of dengue virus by the demographic transition on dengue in Thailand: insights from
use of reverse transcription-recombinase polymerase amplification. a statistical analysis and mathematical modeling. PLoS Med 2009;
J Clin Microbiol 2015; 53: 830–37. 6: e1000139.

e97 www.thelancet.com/infection Vol 17 March 2017


Series

32 Rodríguez-Barraquer I, Buathong R, Iamsirithaworn S, et al. 53 Wang C, Katzelnick LC, Montoya M, Hue KDT, Simmons CP,
Revisiting Rayong: shifting seroprofiles of dengue in Thailand and Harris E. Evolutionarily successful asian 1 dengue virus 2 lineages
their implications for transmission and control. Am J Epidemiol contain one substitution in envelope that increases sensitivity to
2014; 179: 353–60. polyclonal antibody neutralization. J Infect Dis 2016; 213: 975–84.
33 Waggoner JJ, Balmaseda A, Gresh L, et al. Homotypic dengue virus 54 Dowd KA, DeMaso CR, Pierson TC. Genotypic differences in
reinfections in Nicaraguan children. J Infect Dis 2016; 214: 968–93. dengue virus neutralization are explained by a single amino acid
34 Duong V, Lambrechts L, Paul RE, et al. Asymptomatic humans mutation that modulates virus breathing. MBio 2015; 6: e01559–15.
transmit dengue virus to mosquitoes. Proc Natl Acad Sci 2015; 55 Zhang C, Mammen MP, Chinnawirotpisan P, et al.
112: 14688–93. Clade replacements in dengue virus serotypes 1 and 3 are associated
35 Katzelnick LC, Montoya M, Gresh L, Balmaseda A, Harris E. with changing serotype prevalence. J Virol 2005; 79: 15123–30.
Neutralizing antibody titers against dengue virus correlate with 56 Quiner CA, Parameswaran P, Ciota AT, et al. Increased replicative
protection from symptomatic infection in a longitudinal cohort. fitness of a dengue virus 2 clade in native mosquitoes: potential
Proc Natl Acad Sci USA 2016; 113: 728–33. contribution to a clade replacement event in Nicaragua. J Virol 2014;
36 Clapham HE, Rodriguez-Barraquer I, Azman AS, et al. 88: 13125–34.
Dengue virus (DENV) neutralizing antibody kinetics in children 57 Adams B, Holmes EC, Zhang C, et al. Cross-protective immunity
after symptomatic primary and postprimary DENV infection. can account for the alternating epidemic pattern of dengue virus
J Infect Dis 2016; 213: 1428–35. serotypes circulating in Bangkok. Proc Natl Acad Sci USA 2006;
37 Fox A, Whitehead S, Anders KL, et al. Investigation of dengue and 103: 14234–39.
Japanese encephalitis virus transmission in Hanam, Vietnam. 58 Bennett SN, Holmes EC, Chirivella M, et al. Molecular evolution of
Am J Trop Med Hyg 2014; 90: 892–96. dengue 2 virus in Puerto Rico: positive selection in the viral
38 Perkins TA, Reiner RC, Rodriguez-Barraquer I, Smith DL, envelope accompanies clade reintroduction. J Gen Virol 2006;
Scott TW, Cummings DAT. A review of transmission models of 87: 885–93.
dengue: a quantitative and qualitative analysis of model features. 59 Twiddy SS, Woelk CH, Holmes EC. Phylogenetic evidence for
In: Gubler DJ, Ooi EE, Vasudevan SG, Farrar J, eds. Dengue and adaptive evolution of dengue viruses in nature. J Gen Virol 2002;
dengue hemorrhagic fever, 2nd edn. Wallingford: CAB 83: 1679–89.
International, 2014: 99–114. 60 Cummings DAT, Schwartz IB, Billings L, Shaw LB, Burke DS.
39 Forshey BM, Reiner RC, Olkowski S, et al. Incomplete protection Dynamic effects of antibody-dependent enhancement on the fitness
against dengue virus type 2 re-infection in Peru. PLoS Negl Trop Dis of viruses. Proc Natl Acad Sci USA 2005; 102: 15259–64.
2016; 10: e0004398. 61 Recker M, Blyuss KB, Simmons CP, et al. Immunological serotype
40 Hadinegoro SR, Arredondo-García JL, Capeding MR, et al. interactions and their effect on the epidemiological pattern of
Efficacy and long-term safety of a dengue vaccine in regions of dengue. Proc Biol Sci 2009; 276: 2541–48.
endemic disease. N Engl J Med 2015; 373: 1195–206. 62 Wearing HJ, Rohani P. Ecological and immunological determinants
41 Capeding MR, Tran NH, Hadinegoro SR, et al. Clinical efficacy and of dengue epidemics. Proc Natl Acad Sci USA 2006; 103: 11802–07.
safety of a novel tetravalent dengue vaccine in healthy children in 63 Sabchareon A, Wallace D, Sirivichayakul C, et al. Protective efficacy
Asia: a phase 3, randomised, observer-masked, placebo-controlled of the recombinant, live-attenuated, CYD tetravalent dengue vaccine
trial. Lancet 2014; 384: 1358–65. in Thai schoolchildren: a randomised, controlled phase 2b trial.
42 Villar L, Dayan GH, Arredondo-García JL, et al. Efficacy of a Lancet 2012; 380: 1559–67.
tetravalent dengue vaccine in children in Latin America. 64 Durbin AP, Kirkpatrick BD, Pierce KK, et al. A single dose of any of
N Engl J Med 2015; 372: 113–23. four different live attenuated tetravalent dengue vaccines is safe and
43 Coudeville L, Baurin N, Vergu E. Estimation of parameters related immunogenic in flavivirus-naive adults: a randomized, double-blind
to vaccine efficacy and dengue transmission from two large clinical trial. J Infect Dis 2013; 207: 957–65.
phase III studies. Vaccine 2015; 34: 6417–25. 65 Anderson KB, Gibbons R V, Cummings DAT, et al. A shorter time
44 Nisalak A, Endy TP, Nimmannitya S, et al. Serotype-specific dengue interval between first and second dengue infections is associated
virus circulation and dengue disease in Bangkok, Thailand from with protection from clinical illness in a school-based cohort in
1973 to 1999. Am J Trop Med Hyg 2003; 68: 191–202. Thailand. J Infect Dis 2014; 209: 360–68.
45 Balmaseda A, Hammond SN, Pérez L, et al. Serotype-specific 66 Reich NG, Shrestha S, King AA, et al. Interactions between
differences in clinical manifestations of dengue. Am J Trop Med Hyg serotypes of dengue highlight epidemiological impact of
2006; 74: 449–56. cross-immunity. J R Soc Interface 2013; 10: 20130414.
46 OhAinle M, Harris E. Dengue pathogenesis: viral factors. 67 Weiskopf D, Sette A. T-cell immunity to infection with dengue virus
In: Gubler DJ, Ooi EE, Vasudevan SG, Farrar J, eds. Dengue and in humans. Front Immunol 2014; 5: 93.
dengue hemorrhagic fever, 2nd edn. Wallingford: CAB 68 Corbett KS, Katzelnick L, Tissera H, Amerasinghe A, de Silva AD,
International, 2014: 231–50. de Silva AM. Preexisting neutralizing antibody responses distinguish
47 OhAinle M, Balmaseda A, Macalalad AR, et al. Dynamics of dengue clinically inapparent and apparent dengue virus infections in a
disease severity determined by the interplay between viral genetics Sri Lankan pediatric cohort. J Infect Dis 2015; 211: 590–99.
and serotype-specific immunity. Sci Transl Med 2011; 3: 114ra128. 69 Puschnik A, Lau L, Cromwell EA, Balmaseda A, Zompi S, Harris E.
48 Bhoomiboonchoo P, Nisalak A, Chansatiporn N, et al. Correlation between dengue-specific neutralizing antibodies and
Sequential dengue virus infections detected in active and passive serum avidity in primary and secondary dengue virus 3 natural
surveillance programs in Thailand, 1994–2010. BMC Public Health infections in humans. PLoS Negl Trop Dis 2013; 7: e2274.
2015; 15: 1–10. 70 Halstead SB, Papaevangelou G. Transmission of dengue 1 and 2
49 Kochel TJ, Watts DM, Halstead SB, et al. Effect of dengue-1 viruses in Greece in 1928. Am J Trop Med Hyg 1980; 29: 635–37.
antibodies on American dengue-2 viral infection and dengue 71 Guzman MG, Alvarez M, Rodriguez-Roche R, et al. Neutralizing
haemorrhagic fever. Lancet 2002; 360: 310–12. antibodies after infection with dengue 1 virus. Emerg Infect Dis 2007;
50 Olkowski S, Forshey BM, Morrison AC, et al. Reduced risk of 13: 282–86.
disease during postsecondary dengue virus infections. J Infect Dis 72 Halstead SB, Grosz CR. Subclinical Japanese encephalitis. I.
2013; 208: 1026–33. Infection of Americans with limited residence in Korea.
51 Guirakhoo F, Pugachev K, Arroyo J, et al. Viremia and Am J Hyg 1962; 75: 190–201.
immunogenicity in nonhuman primates of a tetravalent yellow 73 Hickey AC, Koster JA, Thalmann CM, et al. Serotype-specific host
fever-dengue chimeric vaccine: genetic reconstructions, dose responses in rhesus macaques after primary dengue challenge.
adjustment, and antibody responses against wild-type dengue virus Am J Trop Med Hyg 2013; 89: 1043–57.
isolates. Virology 2002; 298: 146–59. 74 Koraka P, Benton S, van Amerongen G, Stittelaar KJ,
52 VanBlargan LA, Mukherjee S, Dowd KA, Durbin AP, Whitehead SS, Osterhaus AD. Characterization of humoral and cellular immune
Pierson TC. The type-specific neutralizing antibody response responses in cynomolgus macaques upon primary and subsequent
elicited by a dengue vaccine candidate is focused on two amino heterologous infections with dengue viruses. Microbes Infect 2007;
acids of the envelope protein. PLoS Pathog 2013; 9: e1003761. 9: 940–46.

www.thelancet.com/infection Vol 17 March 2017 e98


Series

75 Katzelnick LC, Fonville JM, Gromowski GD, et al. Dengue viruses 97 Teoh EP, Kukkaro P, Teo EW, et al. The structural basis for
cluster antigenically but not as discrete serotypes. Science 2015; serotype-specific neutralization of dengue virus by a human
349: 1338–43. antibody. Sci Transl Med 2012; 4: 139ra83.
76 Gibbons RV, Kalanarooj S, Jarman RG, et al. Analysis of repeat 98 de Alwis R, Smith SA, Olivarez NP, et al. Identification of human
hospital admissions for dengue to estimate the frequency of third neutralizing antibodies that bind to complex epitopes on dengue
or fourth dengue infections resulting in admissions and dengue virions. Proc Natl Acad Sci USA 2012; 109: 7439–44.
hemorrhagic fever, and serotype sequences. Am J Trop Med Hyg 99 Messer WB, Yount BL, Royal SR, et al. Functional transplant of
2007; 77: 910–13. a dengue virus serotype 3 (DENV3)-specific human monoclonal
77 Rouvinski A, Guardado-Calvo P, Barba-Spaeth G, et al. antibody epitope into DENV1. J Virol 2016; 90: 5090–97.
Recognition determinants of broadly neutralizing human 100 Halstead SB. In vivo enhancement of dengue virus infection in
antibodies against dengue viruses. Nature 2015; 520: 109–13. rhesus monkeys by passively transferred antibody. J Infect Dis
78 Sabin AB. Research on dengue during World War II. 1979; 140: 527–33.
Am J Trop Med Hyg 1952; 1: 30–50. 101 Goncalvez AP, Engle RE, St Claire M, Purcell RH, Lai C-J.
79 Halstead S, Casals J. Studies on the immunization of monkeys Monoclonal antibody-mediated enhancement of dengue virus
against dengue I. Protection derived from single and sequential infection in vitro and in vivo and strategies for prevention.
virus infections. Am J Trop Med Hyg 1973; 22: 365–74. Proc Natl Acad Sci USA 2007; 104: 9422–27.
80 Halstead SB. Identifying protective dengue vaccines: guide to 102 Zellweger RM, Prestwood TR, Shresta S. Enhanced infection of
mastering an empirical process. Vaccine 2013; 31: 4501–07. liver sinusoidal endothelial cells in a mouse model of
81 Bernardo L, Izquierdo A, Prado I, et al. Primary and secondary antibody-induced severe dengue disease. Cell Host Microbe 2010;
infections of Macaca fascicularis monkeys with Asian and American 7: 128–39.
genotypes of dengue virus 2. Clin Vaccine Immunol 2008; 15: 439–46. 103 Williams KL, Sukupolvi-Petty S, Beltramello M, et al.
82 Thomas SJ, Nisalak A, Anderson KB, et al. Dengue plaque reduction Therapeutic efficacy of antibodies lacking Fcγ receptor binding
neutralization test (PRNT) in primary and secondary dengue virus against lethal dengue virus infection is due to neutralizing
infections: how alterations in assay conditions impact performance. potency and blocking of enhancing antibodies [corrected].
Am J Trop Med Hyg 2009; 81: 825–33. PLoS Pathog 2013; 9: e1003157.
83 Mukherjee S, Dowd KA, Manhart CJ, et al. Mechanism and 104 Simmons CP, Chau TNB, Thuy TT, et al. Maternal antibody and
significance of cell type-dependent neutralization of flaviviruses. viral factors in the pathogenesis of dengue virus in infants.
J Virol 2014; 88: 7210–20. J Infect Dis 2007; 196: 416–24.
84 Pierson TC, Xu Q, Nelson S, et al. The stoichiometry of 105 Chau TNB, Quyen NTH, Thuy TT, et al. Dengue in Vietnamese
antibody-mediated neutralization and enhancement of West Nile infants—results of infection-enhancement assays correlate with
virus infection. Cell Host Microbe 2007; 1: 135–45. age-related disease epidemiology, and cellular immune
85 Plevka P, Battisti AJ, Sheng J, Rossmann MG. Mechanism for responses correlate with disease severity. J Infect Dis 2008;
maturation-related reorganization of flavivirus glycoproteins. 198: 516–24.
J Struct Biol 2014; 185: 27–31. 106 Chau TNB, Hieu NT, Anders KL, et al. Dengue virus infections
86 Austin SK, Dowd K, Shrestha B, et al. Structural basis of differential and maternal antibody decay in a prospective birth cohort study of
neutralization of DENV-1 genotypes by an antibody that recognizes Vietnamese infants. J Infect Dis 2009; 200: 1893–900.
a cryptic epitope. PLoS Pathog 2012; 8: e1002930. 107 Kliks SC, Nimmanitya S, Nisalak A, Burke DS. Evidence that
87 Brien JD, Austin SK, Sukupolvi-Petty S, et al. Genotype-specific maternal dengue antibodies are important in the development of
neutralization and protection by antibodies against dengue virus dengue hemorrhagic fever in infants. Am J Trop Med Hyg 1988;
type 3. J Virol 2010; 84: 10630–43. 38: 411–19.
88 Timiryasova TM, Bonaparte MI, Luo P, Zedar R, Hu BT, 108 Clapham H, Cummings DAT, Nisalak A, et al. Epidemiology of
Hildreth SW. Optimization and validation of a plaque reduction infant dengue cases illuminates serotype-specificity in the
neutralization test for the detection of neutralizing antibodies to interaction between immunity and disease, and changes in
four serotypes of dengue virus used in support of dengue vaccine transmission dynamics. PLoS Negl Trop Dis 2015; 9: e0004262.
development. Am J Trop Med Hyg 2013; 88: 962–70. 109 Hammond SN, Balmaseda A, Pérez L, et al. Differences in
89 Endy TP, Nisalak A, Chunsuttitwat S, et al. Relationship of dengue severity in infants, children, and adults in a 3-year
preexisting dengue virus (DV) neutralizing antibody levels to hospital-based study in Nicaragua. Am J Trop Med Hyg 2005;
viremia and severity of disease in a prospective cohort study of DV 73: 1063–70.
infection in Thailand. J Infect Dis 2004; 189: 990–1000. 110 Kliks SC, Nisalak A, Brandt WE, Wahl L, Burke DS.
90 Buddhari D, Aldstadt J, Endy TP, et al. Dengue virus Antibody-dependent enhancement of dengue virus growth in
neutralizing antibody levels associated with protection from human monocytes as a risk factor for dengue hemorrhagic fever.
infection in Thai cluster studies. PLoS Negl Trop Dis 2014; 8: e3230. Am J Trop Med Hyg 1989; 40: 444–51.
91 de Alwis R, Beltramello M, Messer WB, et al. In-depth analysis of 111 Libraty DH, Acosta LP, Tallo V, et al. A prospective nested
the antibody response of individuals exposed to primary dengue case-control study of dengue in infants: rethinking and refining
virus infection. PLoS Negl Trop Dis 2011; 5: e1188. the antibody-dependent enhancement dengue hemorrhagic fever
model. PLoS Med 2009; 6: e1000171.
92 Smith SA, de Alwis AR, Kose N, et al. The potent and broadly
neutralizing human dengue virus-specific monoclonal antibody 112 Laoprasopwattana K, Libraty DH, Endy TP, et al. Dengue virus
1C19 reveals a unique cross-reactive epitope on the bc loop of (DV) enhancing antibody activity in preillness plasma does not
domain II of the envelope protein. MBio 2013; 4: e00873–13. predict subsequent disease severity or viremia in secondary DV
infection. J Infect Dis 2005; 192: 510–19.
93 Tsai W-Y, Durbin A, Tsai J-J, Hsieh S-C, Whitehead S, Wang W-K.
Complexity of neutralizing antibodies against multiple dengue 113 Halstead SB. Dengue antibody-dependent enhancement: knowns
virus serotypes after heterotypic immunization and secondary and unknowns. Microbiol Spectr 2014; 2: 249–71.
infection revealed by in-depth analysis of cross-reactive antibodies. 114 Bernasconi NL, Traggiai E, Lanzavecchia A. Maintenance of
J Virol 2015; 89: 7348–62. serological memory by polyclonal activation of human memory
94 Fibriansah G, Tan JL, Smith SA, et al. A highly potent human B cells. Science 2002; 298: 2199–202.
antibody neutralizes dengue virus serotype 3 by binding across 115 Amanna IJ, Slifka MK. Mechanisms that determine plasma cell
three surface proteins. Nat Commun 2015; 6: 6341. lifespan and the duration of humoral immunity. Immunol Rev
95 Fibriansah G, Tan JL, Smith SA, et al. A potent anti-dengue human 2010; 236: 125–38.
antibody preferentially recognizes the conformation of E protein 116 Amanna IJ, Carlson NE, Slifka MK. Duration of humoral immunity
monomers assembled on the virus surface. EMBO Mol Med 2014; to common viral and vaccine antigens. N Engl J Med 2007;
6: 358–71. 357: 1903–15.
96 Fibriansah G, Ibarra KD, Ng T, et al. Cryo-EM structure of an 117 Slifka MK. Vaccine-mediated immunity against dengue and the
antibody that neutralizes dengue virus type 2 by locking E protein potential for long-term protection against disease. Front Immunol
dimers. Science 2015; 349: 88–91. 2014; 5: 195.

e99 www.thelancet.com/infection Vol 17 March 2017


Series

118 Dejnirattisai W, Wongwiwat W, Supasa S, et al. A new class of 135 Laoprasopwattana K, Libraty DH, Endy TP, et al.
highly potent, broadly neutralizing antibodies isolated from Antibody-dependent cellular cytotoxicity mediated by plasma
viremic patients infected with dengue virus. Nat Immunol 2015; obtained before secondary dengue virus infections: potential
16: 785. involvement in early control of viral replication. J Infect Dis 2007;
119 Purtha WE, Tedder TF, Johnson S, Bhattacharya D, Diamond MS. 195: 1108–16.
Memory B cells, but not long-lived plasma cells, possess antigen 136 García G, Arango M, Pérez AB, et al. Antibodies from patients with
specificities for viral escape mutants. J Exp Med 2011; dengue viral infection mediate cellular cytotoxicity. J Clin Virol 2006;
208: 2599–606. 37: 53–57.
120 Weiskopf D, Angelo MA, Bangs DJ, et al. The human CD8+ T cell 137 Avirutnan P, Punyadee N, Noisakran S, et al. Vascular leakage in
responses induced by a live attenuated tetravalent dengue vaccine are severe dengue virus infections: a potential role for the nonstructural
directed against highly conserved epitopes. J Virol 2015; 89: 120–28. viral protein NS1 and complement. J Infect Dis 2006; 193: 1078–88.
121 Rothman AL. Immunity to dengue virus: a tale of original antigenic 138 Markoff LJ, Innis BL, Houghten R, Henchal LS. Development of
sin and tropical cytokine storms. Nat Rev Immunol 2011; 11: 532–43. cross-reactive antibodies to plasminogen during the immune
122 Yauch LE, Zellweger RM, Kotturi MF, et al. A protective role for response to dengue virus infection. J Infect Dis 1991; 164: 294–301.
dengue virus-specific CD8+ T cells. J Immunol 2009; 182: 4865–73. 139 Falconar AK. The dengue virus nonstructural-1 protein (NS1)
123 Hatch S, Endy TP, Thomas S, et al. Intracellular cytokine production generates antibodies to common epitopes on human blood clotting,
by dengue virus-specific T cells correlates with subclinical secondary integrin/adhesin proteins and binds to human endothelial cells:
infection. J Infect Dis 2011; 203: 1282–91. potential implications in haemorrhagic fever pathogenesis.
124 Weiskopf D, Angelo MA, de Azeredo EL, et al. Comprehensive Arch Virol 1997; 142: 897–916.
analysis of dengue virus-specific responses supports an HLA-linked 140 Lin Y-S, Yeh T-M, Lin C-F, et al. Molecular mimicry between virus
protective role for CD8+ T cells. Proc Natl Acad Sci USA 2013; and host and its implications for dengue disease pathogenesis.
110: E2046–53. Exp Biol Med (Maywood) 2011; 236: 515–23.
125 Weiskopf D, Cerpas C, Angelo MA, et al. Human CD8+ T cell 141 Chuang Y-C, Lin Y-S, Liu H-S, Yeh T-M. Molecular mimicry between
responses against the 4 dengue virus serotypes are associated with dengue virus and coagulation factors induces antibodies to inhibit
distinct patterns of protein targets. J Infect Dis 2015; 212: 1743–51. thrombin activity and enhance fibrinolysis. J Virol 2014; 88: 13759–68.
126 Guy B, Nougarede N, Begue S, et al. Cell-mediated immunity 142 Chuang Y-C, Lin Y-S, Liu H-S, Wang J-R, Yeh T-M.
induced by chimeric tetravalent dengue vaccine in naive or Antibodies against thrombin in dengue patients contain both
flavivirus-primed subjects. Vaccine 2008; 26: 5712–21. anti-thrombotic and pro-fibrinolytic activities. Thromb Haemost
127 Mangada MM, Rothman AL. Altered cytokine responses of 2013; 110: 358–65.
dengue-specific CD4+ T cells to heterologous serotypes. J Immunol 143 Modhiran N, Watterson D, Muller DA, et al. Dengue virus NS1
2005; 175: 2676–83. protein activates cells via Toll-like receptor 4 and disrupts
128 Yauch LE, Prestwood TR, May MM, et al. CD4+ T cells are not endothelial cell monolayer integrity. Sci Transl Med 2015;
required for the induction of dengue virus-specific CD8+ T cell or 7: 304ra142.
antibody responses but contribute to protection after vaccination. 144 Beatty PR, Puerta-Guardo H, Killingbeck SS, Glasner DR,
J Immunol 2010; 185: 5405–16. Hopkins K, Harris E. Dengue virus NS1 triggers endothelial
129 Weiskopf D, Bangs DJ, Sidney J, et al. Dengue virus infection elicits permeability and vascular leak that is prevented by NS1 vaccination.
highly polarized CX3CR1+ cytotoxic CD4+ T cells associated with Sci Transl Med 2015; 7: 304ra141.
protective immunity. Proc Natl Acad Sci USA 2015; 112: E4256–63. 145 Puerta-Guardo H, Glasner DR, Harris E. Dengue Virus NS1
130 Pagni S, Fernandez-Sesma A. Evasion of the human innate immune disrupts the endothelial glycocalyx, leading to hyperpermeability.
system by dengue virus. Immunol Res 2012; 54: 152–9. PLoS Pathog 2016; 12: e1005738.
131 Popper SJ, Gordon A, Liu M, Balmaseda A, Harris E, Relman DA. 146 Vannice KS, Roehrig JT, Hombach J. Next generation dengue
Temporal dynamics of the transcriptional response to dengue virus vaccines: a review of the preclinical development pipeline.
infection in Nicaraguan children. PLoS Negl Trop Dis 2012; 6: e1966. Vaccine 2015; 33: 7091–99.
132 Simmons CP, Popper S, Dolocek C, et al. Patterns of host genome- 147 Sampath S, Carrico C, Janes J, et al. Glycan masking of Plasmodium
wide gene transcript abundance in the peripheral blood of patients vivax Duffy binding protein for probing protein binding function
with acute dengue hemorrhagic fever. J Infect Dis 2007; and vaccine development. PLoS Pathog 2013; 9: e1003420.
195: 1097–107. 148 McLellan JS, Correia BE, Chen M, et al. Design and characterization
133 Kwissa M, Nakaya HI, Onlamoon N, et al. Dengue virus infection of epitope-scaffold immunogens that present the motavizumab
induces expansion of a CD14+CD16+ monocyte population that epitope from respiratory syncytial virus. J Mol Biol 2011; 409: 853–66.
stimulates plasmablast differentiation. Cell Host Microbe 2014; 149 Guy B, Jackson N. Dengue vaccine: hypotheses to understand
16: 115–27. CYD-TDV-induced protection. Nat Rev Microbiol 2015; 14: 45–54.
134 Koraka P, Suharti C, Setiati TE, et al. Kinetics of dengue 150 Plotkin SA, Gilbert PB. Nomenclature for immune correlates of
virus-specific serum immunoglobulin classes and subclasses protection after vaccination. Clin Infect Dis 2012; 54: 1615–17.
correlate with clinical outcome of infection. J Clin Microbiol 2001;
39: 4332–38.

www.thelancet.com/infection Vol 17 March 2017 e100

Вам также может понравиться