Inmunidad Vacunas Dengue
Inmunidad Vacunas Dengue
Inmunidad Vacunas Dengue
Evidence consensus
Complete (absence)
Good
. ,,.
• ·:~,~ ~\ .
Moderate
•
Poor
Indeterminable
Poor
Moderate
Good
Complete (presence)
Fig. 19.1 National and subnational consensus on complete absence (green) through complete presence (red) of human dengue cases.
Blue dots are past reported cases or outbreaks. (From Brady OJ, Gething PW, Bhatt S, et al: Refining the global spatial limits of dengue virus
transmission by evidence-based consensus. PLoS Negl Trop Dis. 2012;6:e1760.)
DISEASE, VIRUS, EPIDEMIOLOGY, AND high concentrations and may activate toll-receptor 4 (TLR4),
directly damage endothelial cells, contribute to the damage of
PATHOGENESIS liver cells, activate complement, and stimulate the release of
Clinical Description cytokines.47–51 All of these are the observed features of DVPS.
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Dengue Vaccines 277
small protein that is hidden under the E protein but that proj- not detectable by immunoassays, may retain endothelial tox-
ects to the surface in its immature state. Each virus particle has icity, in vivo. Progress in understanding dengue pathogenesis 19
180 monomers of E that are organized into 90 tightly packed is hindered by the absence of an aggressive research agenda.
dimers that lie flat on the surface of the viral membrane. Specific antiviral medications are not available, but sup-
Each E protein monomer has three domains: DI, DII, and portive intensive care and careful fluid management is life-sav-
DIII.54–57 The hinge that connects DI to DII is used to flex DII ing.79 The single available preventive measure, comprehensive
in the low pH environment of the endosome, leading to the mosquito control, has not been effective. Increases in ambient
exposure of its fusion loop.56,57 DIII is thought to be involved temperatures increase vectoral capacity.80,81
in receptor binding. Individual subunits of E protein consist of
three β-barrel domains designated domains I (EDI), II (EDII),
and III (EDIII), with the native protein forming a head-to-tail Epidemiology
homodimer. The hydrophobic viral fusion peptide is located Infection of humans follows the bite of an infected Aedes
at the tip of domain II and is shielded by domain III of the mosquito (primarily Aedes aegypti, and to a lesser extent Aedes
adjacent subunit. Domain III appears to be responsible for albopictus or Aedes polynesiensis). Ae aegypti eggs are laid indi-
binding to cellular receptors as several mutations that affect vidually by the female mosquito on the walls of both artifi-
receptor binding are located in this domain.58 cial and natural water containers. Eggs resist desiccation for
weeks to months and hatch when submerged in water. Larvae
and pupae prefer clean water in many different types of artifi-
Pathogenesis as It Relates to Prevention cial containers: water storage containers (tanks, jars, cisterns,
Classical DHF/DSS accompanies first DENV infections in pots), ornamental containers (flower holders, ant traps, shrine
5–11-month-old infants born to dengue-immune mothers.59 objects), and discarded items (rubber tires, plastic contain-
Altogether 90+% of cases accompany second (occasionally ers, bottles). The species also occasionally uses natural larval
third) heterotypic DENV infections in individuals older than habitats, such a bromeliads and tree holes. Containers located
1 year.43,60,61 These two epidemiologic settings have in com- outside dwellings can be filled with rainwater and are produc-
mon a single immune factor—IgG1 DENV antibodies. Numer- tive during the rainy season. Indoor or sheltered containers
ous studies show that at subneutralizing concentrations DENV may produce pupae throughout the year. When extensive use
antibodies enhance DENV infections in Fc-receptor-bearing is made of outdoor larval habitats the prevalence of larvae
cells, a process referred to as antibody-dependent enhance- and adults are often subject to marked seasonal variation. The
ment (ADE).62,63 In humans, peak viremia titers measured duration of the larval stages is 7–9 days at 25°C and that of the
early in secondary DENV infections were positively correlated pupal stage is 2–3 days at the same temperature.
with the subsequent occurrence of DHF/DSS.64 DHF/DSS is The ecology of adult Ae aegypti in a domestic urban envi-
rare, seen in only 2–4% of second DENV infections.65 ronment is characterized by strong anthropophilia and diur-
The discovery that DVPS is a toxicosis, mediated by NS1 nal feeding usually with two peaks, one in mid-morning and
a protein with endotoxin-like properties, has fundamentally another in late afternoon. It seems likely that most females
changed the dengue pathogenesis landscape.47–51,66 For 60 can feed twice or even three times during a single gonotro-
years, competing camps promoted many alternative hypoth- phic cycle.82–84 The preferred resting sites of adults are sheltered
eses of the pathogenesis of severe dengue: intrinsic viral viru- dark spaces inside houses. The average life span for females is
lence, autoimmunity, direct DENV infection of endothelial 8–15 days and that for males about 3–6 days. Geographic dis-
cells, “original antigenic sin” and second-infection pathogenic persal of adults is usually limited, averaging about 30–50 m a
immune responses (“cytokine storm”).67–73 These hypotheses day for females, which means that a female rarely visits more
centered on explaining DHF/DSS due to second heterotypic than two or three houses during her lifetime. In some locales,
DENV infections but, ignored or made no attempt to explain such as Puerto Rico, female flight distance may be related to
DVPS during a first infection in infants born to dengue- the availability of oviposition sites (breeding sites) and so
immune mothers. NS1 toxicosis provides a unitary pathogene- might be much longer. Passive dispersal of eggs and larvae is
sis hypothesis. Antibodies, actively or passively acquired form common, including trains, boats, and aircraft. Because of the
immune-complexes with circulating virus, infect Fc-R-bearing weak spontaneous dispersal of the species and its easy passive
cells leading to increased production of DENV NS1. dispersal, the International Sanitary Regulations require that
Comprehensive pathology studies on 13 fatal DSS cases the area within 400 m of international ports and airports be
found splenic and lymph node macrophages were produc- kept free of Ae aegypti.
tively infected.74 Endothelial cells were not infected. There Two main factors regulate Ae aegypti populations: climate
was evidence of nonreplicative DENV infection of hepatocytes and the availability of breeding sites. Population changes may
and widespread staining of parenchymal cells by DENV NS or may not correlate with weather. Daily, seasonal, and inter-
1 and complement subunits.74,75 While there is a correlation annual variability in temperature, atmospheric moisture, and
between DVPS and peak NS1 blood levels measured soon after rainfall all influence mosquitoes in a variety of ways. From an
onset of fever, when measured during the period of severe vas- epidemiological perspective, the increased number of inter-
cular permeability NS1 blood concentrations may be rather rupted feeding per replete feeds that is the consequence of
low. During the late acute stage, anamnestic antibodies nearly 2° or 3° warmer temperatures is equivalent to a doubling of
obliterate viremias and RNAemias. Formal attempts to cor- the density of Ae aegypti. Under circumstances where a major-
relate NS1 blood levels with vascular permeability are prob- ity of breeding sites are indoors, warm temperature and high
ably only possible during primary DENV infections. In the moisture contribute to increased adult survival together with
literature almost all studies on NS1 blood levels and vascular the effect of warmer temperatures on shortening the extrin-
permeability have been performed during secondary DENV sic incubation period contribute to the occurrence of dengue
infections. The few studies on infants with severe primary den- outbreaks during the hot, rainy season of Southeast Asia. In
gue found relatively low viremias compared with peak values countries with seasonal variation, DENV transmission is high-
in secondary infections.76–78 Consecutive daily measurements est during warm or rainy seasons. In the true tropics, such as
were not made. Damage to endothelial cells may be a thresh- Singapore, DENV transmission is year-round, but, impacted
old phenomenon and related to duration of exposure to NS1. by seasonal variations in nearby countries whose populations
It is also possible that circulating NS1 antibody complexes, either work in or may visit Singapore.
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
278 SECTION II Licenced Vaccines and Vaccines in Development
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Dengue Vaccines 279
DENV-4 1036 were passaged 15–50 times in primary dog kid- find a balance between acceptable levels of reactogenicity and
ney (PDK) cells.128 Candidate vaccines were tested at Phase 1 obtaining high rates of tetravalent neutralizing antibodies in
in Thailand in flavivirus-susceptible adult Thai volunteers.129 humans. Manufacturing complexities and the desire to find a
DENV-1 PDK13, DENV-2 PDK53, and DENV-4 PDK48 dem- vaccine with a shorter time to protection led to the abandon-
onstrated acceptable reactogenicity and immunogenicity.130–132 ment of the project. An important outcome of these efforts
All 10 U.S. Army soldiers inoculated with DENV-2 PDK 53 was finding live DENVs that could be used as challenge viruses
developed neutralizing antibodies in the absence of dengue- to test protection of vaccinated subjects.150,151
like symptoms.133 DENV-3 16562 did not replicate in PDK
cells and was attenuated by 48 passages in primary African DNA Vaccines. Workers at the Naval Medical Research Center
green monkey kidney (AGMK) cells and 3 final passages in evaluated two eukaryotic plasmid expression vectors (pkC-
fetal rhesus lung cells (FRhL).129 The four monovalent candi- MVint-Polyli and pVR1012; Vical, Inc., San Diego, CA) express-
dates after a single dose of 103.7–104.4 plaque-forming units ing the PrM protein and 92% of the E protein for DENV-1
(pfu), tested separately, elicited neutralizing antibody sero- and DENV-2 virus (see Table 19.2). These constructs induced
conversions in 3/5, 5/5, 5/5, and 5/5 American volunteers, neutralizing antibody in mice,152 and they were subsequently
respectively.134 Bivalent and trivalent formulations using improved by adding immunostimulatory CpG motifs, and the
DENV-1, -2, and -4 vaccine candidates elicited balanced full-length E gene with PrM.152–155 In early studies, a DENV-1
seroconversions.135 The tetravalent vaccine was designed to DNA vaccine protected a portion of challenged monkeys from
induce primary-type immune responses to each of the four viremia for varying lengths of time.156 With the recognition
DENVs simultaneously as had been demonstrated in suscep- that dendritic cells are the percutaneous portal for dengue
tible rhesus monkeys.90,136 virus replication,157 efforts were taken to target DNA vaccines
In 1993, attenuated DENVs were shipped to Aventis to these cells.158
Pasteur for commercial production.129 Concerned about In a Phase 1 clinical trial, a DENV-1 DNA vaccine was
the safety of primary AGMK cells, the manufacturer chose administered intramuscularly to 22 flavivirus-negative volun-
FRhL to produce the DENV 3 GMK 30 component. This tet- teers, half of whom received high and low dosages, respec-
ravalent vaccine entered Phase 1 testing in 1995. Problems tively, in a three-dose series (at day 0, and at 1 and 5 months).
were recognized immediately. During Phase 1 and Phase 2, At the completion of this series, none of the low-dosage recip-
the vaccine was moderately reactogenic, failed to raise neu- ients and only 5 of 11 high-dosage recipients developed neu-
tralizing antibodies against DENV 1, 2, and 4 viruses, and tralizing antibodies. In addition, all volunteers immunized
exhibited a dominant DENV 3 viremia.134,137–141 It was con- with high-dose monovalent DENV-1 DNA vaccine developed
cluded that DENV-3 GMK 35 FRhL 3 had reverted toward T-cell responses as measured by interferon gamma ELISPOT
wild-type virus with attributes that suppressed the growth assay.159
of PDK-attenuated DENVs. An effort was made by Aventis To enhance neutralizing antibody responses, a tetrava-
to find a satisfactorily attenuated strain of DENV 3 GMK 30 lent dengue DNA vaccine was formulated in a lipid-based
FRhL 3 by plaque purification.142 A virus with small plaque adjuvant, Vaxfectin, and evaluated in a nonhuman primate
morphology, temperature sensitive, with reduced infectivity model. The vaccine was administered by intramuscular injec-
for vector mosquitoes and reduced viremia in monkeys was tions using the Biojector 2000 needle-free delivery device.
grown in Vero cells and given at a low dose (10−2 pfu) to 15 The results showed a significant increase in neutralizing anti-
Hong Kong medical students. This produced nonhospital- body responses to DENV-1, DENV-3, and DENV-4 in animals
ized dengue fever in all, graded as “severe” in 13. This ended immunized with the formulated tetravalent vaccine compared
the effort by Aventis for commercial production of the PDK/ to animals given the unformulated dengue DNA vaccine. The
GMK-passaged dengue vaccine. use of Vaxfectin also resulted in significantly greater protec-
The WRAIR developed and tested a large number of tion against live DENV-2 challenge.160 A subsequent Phase 1
monovalent and tetravalent live-attenuated DENVs. In col- clinical trial of the formulated tetravalent dengue DNA vac-
laboration with GlaxoSmithKline Vaccines (GSK), DENV-1–4 cine delivered by needle injection showed that the vaccine
serially passaged in PDK cells were prepared as vaccines in was safe but generated minimal tetravalent antidengue anti-
FRhL. Some of the resultant candidate vaccines were evaluated body responses.161 Robust IFNgamma T-cell responses were
in Phase 2 tests in adults and children, both susceptibles and observed, as was seen in the Phase 1 trial of the monovalent
partial dengue immunes.143–149 The group found it difficult to DENV-1 DNA vaccine.
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
280 SECTION II Licenced Vaccines and Vaccines in Development
To enhance dengue DNA vaccine immune responses, Wil- vaccinated rhesus monkeys had breakthrough viremias or
liams et al. explored different methods of delivery.162 Investi- RNAemias. Nearly all had anamnestic antibody responses and
gators compared electroporation and needle-free jet injection elevated levels of AST and distinctive cytokine response pat-
given by either the intramuscular or intradermal route. Utiliz- terns IL-10, IL-18, IFN-gamma, IL-12, and liver enzymes.174
ing high (5 mg) and low (1 mg) doses of tetravalent dengue This is the first demonstration of dengue vaccine challenge
DNA vaccine, the highest neutralizing antibody and IFN- virus enhanced abnormal host physiological responses in an
gamma T-cells responses were achieved with the high dose experimental animal model. Detection of subclinical ADE in a
administered by intradermal electroporation. When chal- vaccinated monkey model resulted in termination of the PIV
lenged 1 year postimmunization with live DENV-1, animals development program.
in this group also exhibited significantly fewer mean days of
viremia (1.5 days) postchallenge compared to control unvac-
cinated animals (6.5 days) as determined by RT-PCR. The only Vaccines Completing Phase 3 Clinical Evaluation
other group showing significant protection was the low dose
intradermal electroporation group. Further evaluation of tet- TAK-003. Produced by Takeda Pharmaceutical Company, TAK
ravalent dengue DNA vaccines by electroporation in a Phase 1 003 is a two-dose vaccine given 3 months apart consisting of
clinical trial is under consideration. the prM and E protein genes of DENV-1, -3, and -4 inserted
Incorporating dengue DNA vaccines into a heterologous into a DENV-2 genetic backbone.175,176 This is the same DENV
prime-boost approach was explored for inducing protec- 2 PDK-attenuated DENV 2 strain used in the Mahidol/Aventis
tive antidengue immune responses. Using a rhesus monkey Pasteur vaccine. In January 2018, Takeda announced the com-
model, Simmons et al. showed that, compared to priming pletion of its Phase 3 trial conducted in eight dengue endemic
with DENV-2 DNA vaccine and boosting with two doses of countries in Southeast Asia and Latin America among 20,071
DENV-2 protein, the highest neutralizing antibodies were children 4–16 years of age randomized 2:1 to vaccine or pla-
achieved with three doses of a DENV-2 DNA combined with cebo.177 At 12 months after the second dose, the efficacy of
either a recombinant DENV-2 E protein or purified inacti- preventing dengue among 12,696 vaccinated children, sero-
vated DENV-2 virus.163 However, the best protection against negatives, and seropositives, was 80.2% (95% CI: 73.3–85.3).
live DENV-2 challenge was observed in animals receiving three There were 61 cases (0.5%) of virologically confirmed dengue
doses of purified inactivated virus.164 A more recent study com- in the vaccine group and 149 (2.4%) in the placebo group.
pared priming with either a tetravalent dengue DNA vaccine In the seronegative population of 3531 vaccinated children,
or tetravalent purified inactivated dengue vaccine (PIV) and 20 (0.6%) had virologically confirmed dengue contrasted with
boosting with a tetravalent live-attenuated vaccine (LAV) in 39 among 1726 seronegative controls (2.3%), an efficacy of
the same rhesus monkey model.158 74.9 % (CI: 57.0–85.4). Of 12,696 vaccinated children only
In an effort to simplify the tetravalent dengue DNA vac- five were hospitalized versus 53 among 6314 in the placebo
cine consisting of four separate DNA vaccine plasmids, DNA group. Vaccine efficacy against hospitalization, regardless of
shuffling technology was utilized to generate a single plasmid immune status, was 95.4%. Among seronegatives, two vac-
containing a chimeric tetravalent prME gene that expresses cinated children were hospitalized with severe dengue or
proteins from all four dengue serotypes. The chimeric plas- DHF compared with five among controls. Vaccine efficacy was
mid was created by shuffling the envelope genes from the 97.7% against DENV-2 disease, 73.7 % against DENV-1, and
four dengue viruses. Selected shuffled DNA was transfected 62.6% against DENV-3. There were too few DENV-4 cases to
into human cells, subjected to flow cytometry, and reacted enable efficacy calculations. Notably, DENV-4 antibody titers
with type-specific dengue antibodies. Antibody markers per- and seropositivity persistence were lowest out to 48 months
mitted rapid screening of libraries and identification of novel and 36 months using an older formulation (before a DENV-2
expressed chimeric antigens. A panel of chimeric clones reduction) in Phase 2 studies in dengue-endemic areas driven
expressing C-terminal truncated antigens that combined enve- by lower initial immune responses to DEN-4 in baseline
lope and prM epitopes from all four DENV types when inoc- seronegatives.178,179
ulated in mice and monkeys successfully raised neutralizing Over 27 months of observation the cumulative efficacy for
antibodies. Monkeys resisted challenge with DENV-1 but not all dengue cases, baseline seronegatives and seropostives, was
DENV-2.165,166 Further clinical testing is pending.167 72.7% (95% CI: 67.1–77.3).180 Because TAK-003 efficacy varies
by serotype, the observed changes in serotype dominance in
Inactivated Whole Virus Vaccines. Comprehensive efforts vaccine study sites may contribute to year-to-year efficacy dif-
were undertaken to develop and test candidate purified for- ferences. During the second year, dengue efficacy declined to
malin inactivated dengue vaccines (PIV). The results illustrate 56.2% (95% CI: 42.3–66.8) with the largest decline to 24.5%
the risks of the partial protection phenomenon. At WRAIR, (−34.2 to 57.5) in 4–5-year-old children, with efficacies of
monovalent or tetravalent formalin-inactivated DENV vac- 60.6% (43.8–72.4) in 6–11-year-olds and 71.2% (41.0–85.9)
cines were prepared with or without adjuvants, inoculated in the 12–16-year age groups. Prevention of hospitalization by
in rhesus monkeys and challenged with wild-type DENVs at vaccine remained high at 89.2% (96% CI: 82.4–93.3). Dur-
varying intervals. All PIVs raised significant levels of neutral- ing 27 months, among children vaccinated as seronegatives,
izing antibodies, but usually low levels of challenge viremias prevention of dengue was 67.0% (95% CI: 53.6–76.5). There
or RNAemias were detected.168–170 Because challenge viremia were five vaccinated children, ages 4–5 years, hospitalized with
levels were thought to be below the level that would cause breakthrough dengue infections in year 2 compared with just
disease in humans and antibody responses were fairly robust, one in year 1. While not significant, a trend of increasing inci-
these vaccines were advanced to Phase 1 in susceptibles and dence of breakthrough dengue disease in vaccinated children
partially dengue-immunes.171–173 WRAIR awarded an exclusive may be a warning.
license to GSK and the Oswaldo Cruz Foundation (FioCruz)
for producing and testing a commercial candidate tetravalent
PIV formulation (DPIV). Adjuvanted with AlOH or a GSK
proprietary Adjuvant System (AS), a short-interval challenge
Vaccines in Phase 3 Clinical Trials
concluded that immunized rhesus monkeys were protected.168 Live-Attenuated Tetravalent Dengue Vaccine (LATV-
When a larger group of animals were immunized and chal- 003/005). For nearly 20 years, workers at the National Insti-
lenged at 8 months with wild-type DENV-1–4, a number of tute of Allergy and Infectious Diseases (NIAID) and the Johns
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Dengue Vaccines 281
Hopkins Bloomberg School of Public Health have continuously DENV-4, 1228 that have been tested in extensive preclinical
designed and tested dengue vaccine candidates. DENV-1 and -4 and Phases 1 and 2 trials in flavivirus susceptible and immune 19
were attenuated by removing 30 nucleotides (Δ30) from the individuals. Vero cells serve as the substrate for vaccine virus
nontranslated region (NTR) of the DENV genome. A DENV-2 production. These earlier data are described in the sixth and
vaccine was constructed as a chimera with DENV-4 Δ30 and the seventh editions of this book.
DENV-3 Δ30/31vaccine strain and by deleting additional NTR A three-dose chimeric yellow fever tetravalent dengue vac-
nucleotides.113 A crucial component of this development pro- cine was evaluated in multipart Phases 2b and 3 efficacy trials
gram was that all monovalent vaccine candidates were tested involving 35,000 children in 10 dengue-endemic countries.
for immunogenicity and attenuation in seronegative human Among trials started in 2011, efficacy results were mixed.141,209
volunteers. Innovatively, a second vaccine dose was used as a Pooled efficacy rates for symptomatic dengue 25 months after
test of protection.181–184 A single dose of LATV has solidly pro- complete immunization were 60.3% for all participants, 65.6%
tected seronegative human volunteers for short and long peri- for those 9–16 years of age and 44.6% for those younger than
ods by challenge with nonparental wild-type-like DENV-2 Δ30 9 years of age. Vaccine protection against hospitalization was
(Tonga 74) or DENV-3 Δ30 (Sleman 78). (Durbin, A., personal better at 72.6% for children of all ages, 80.75% for children 9
communication, December 28, 2020). After a single dose of years and older and 55.8% for those under 9 years. Eighteen
vaccine was given to 20 susceptible volunteers, challenge at 6 children were hospitalized for severe dengue among the 22,177
months with DENV-2 Δ30 (Tonga 74) resulted in no viremia, (0.08%) who were vaccinated compared with six among 11,089
dengue rash, or anamnestic antibody response.185 That LATV (0.05%) controls. A high rate of hospitalization among vacci-
protection is likely to be of long duration is supported by solid nated 2–5-year-olds was observed. Among the 2029 children
immunity against a booster dose of live-attenuated vaccine 5 years or younger who received vaccine, 20 were hospitalized
given 12 months after initial dose.186 for dengue, a rate significantly higher (0.99%) than controls
These results are complemented by data showing that a 2/1005 (0.2%), a relative risk of 4.95, P = 0.03. This was attrib-
single dose of LATV raises monospecific DENV-1–4 neutraliz- uted to novel, unstudied pathogenic mechanisms such as young
ing antibodies that are conformationally similar to those after age, a temporal “clustering” of vaccine-related cases occurring
human infections with wild-type DENVs and found to corre- in young children due to the large numbers given vaccine over
late with protection.187–189 Moreover, CD4+ and CD8+ T cell a short period, or to the immunological immaturity of recipi-
responses to LATV closely resemble those raised after infections ents.209,210 These data led the manufacturer and WHO advisory
with wild-type DENVs.190–192 There is growing evidence that groups to recommend vaccine be given to children 9 years of
human T cell responses directed at epitopes on nonstructural age and older and directed to populations in settings, national
proteins contribute importantly to homotypic and heterotypic and regional, with a dengue seroprevalence of 70% or grea
DENV protective immunity.193–195 Finally, LATV contains genes ter.209,211,212 To provide guidance for the deployment of vaccine,
for three of the four DENV NS1 proteins. There is evidence WHO described sampling and statistical methods for measur-
from experimental animal models that DENV NS1 used as a ing population-based DENV seroprevalence.213,214
vaccine antigen protected against lethal DENV challenge.196 Experts not affiliated with clinical trials suggested post-
LATV has been licensed independently to Butantan, a vac- vaccination clinical responses exhibited hallmarks of vaccine
cine manufacturer in Sao Paulo, Brazil, Panacea Biotec Ltd, New ADE and recommended that efficacy should be separately cal-
Delhi, India and to Merck. Vaccine manufactured by Butantan culated for individuals vaccinated when seronegative or sero-
(Butantan-DV) is in the fourth year of Phase 3 clinical testing. positive.215 The manufacturer implemented this suggestion.
An interim analysis found no significant early adverse events and Because fewer than 12% of Phase 3 children had blood col-
high rates of tetravalent DENV neutralizing antibody seroconver- lected prior to vaccination, efficacy and safety calculations were
sions in seronegatives.197 The accrual of efficacy data has been based on inferred serostatus from blood taken 12 months after
slowed by an inadvertent event, the Zika pandemic of 2016-16 the initial dose.216 These data resulted in a revision of estimates
in South America which was associated with an 80% reduction of vaccine efficacy and safety. There was a decline in vaccine
in reported dengue cases.198 Release of Phase 3 results from Brazil efficacy against symptomatic dengue to 34%, compared with
are expected in 2022. Panacea has completed a phase 2 safety 60.3% at 2 years, with a relative risk of 0.66. The cumulative
and immunogenicity analysis for its version of LATV, Tetravalent 5-year incidence of hospitalization was 1.37% among vaccin-
Dengue Vaccine (TDV) with satisfactory results.198a Phase 3 test- ees and 2.0% among controls. Among hospitalized children
ing plans have not been released. Plans for the development and of all ages, 76 receiving vaccine had severe dengue (0.37%)
release of vaccine by Merck have not been announced. compared with 49 among controls (0.45%), a vaccine efficacy
of 25% and relative risk of 1.33.
Vaccine-enhanced hospitalizations were documented for
2–8-year-old seronegative children. Among 1820 vaccinated
Licensed Vaccine children, 131 were hospitalized compared with 33 among 1010
Dengvaxia: Yellow Fever Dengue Chimeras. Insertion of controls (chi-square with Yates = 15.86, P = 0.000068, rela-
flavivirus preM and E genes into the cDNA backbone of yel- tive risk = 2.2). Of these, 25 had severe dengue adverse events
low fever (YF) 17D was pioneered at the St. Louis University compared with four in the placebo group (chi-square with
Health Sciences Center.199,200 Using this technology, DENV Yates = 5.0944, P = 0.024, relative risk = 2.8). In 9–16-year-
chimeras were developed by Acambis, Inc., and licensed for olds, among 3300 vaccinated seronegatives and 1710 controls,
manufacture to Sanofi Pasteur.201,134 It was expected that these 12 vaccinated children were hospitalized with severe dengue
vaccines would benefit from the high fidelity of YF 17D poly- versus 1 in the controls (chi-square = 2.9443, P = 0.086182,
merase.202 Studies to date suggest that chimeric structure of N.S.). Absence of statistical power can be attributed to small
DENV and West Nile viruses provides attenuation.199,201,203–205 numbers. Using Phase 3 data, in the Philippines, where
In contrast, when wild-type prM- E genes of JE, tick-borne, 880,464 9-year-old children were vaccinated in 2016, among
and St Louis encephalitis viruses were inserted into YF 17D an estimated 132,070 seronegatives, during the next 4 years
they retained encephalitic properties requiring alterations in there would be 2241 hospitalizations and 480 cases of severe
prM-E genes to achieve attenuation.199,203,206–208 Sanofi Pasteur dengue expected.217 Serious consequences would be expected
produced chimeras of yellow fever virus and PrM-E genes from as the case fatality rate among Philippine children hospital-
DENV-1, PUO-359; DENV-2, PUO-218; DENV-3, PaH881; and ized for dengue is approximately 5% (Tables 19.3 and 19.4).
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
282 SECTION II Licenced Vaccines and Vaccines in Development
TABLE 19.3 Hospitalizations, Severe Dengue Cases and Relative Risk in 2–8 and 9–16-Year-Olds Who Were Given Vaccine or Placebo 5 Years
Earlier216
Hospitalizations Severe
Cases/10% Sample Cases/10% Sample
Categories 2–8 Years 9–16 Years 2–8 Years 9–16 Years
Vacc 137.4/192.8 64.2/375.1 30.1/192.8 14.8/375.1
Sero Neg Placebo 37.2/100.6 25.3/207.2 5/100.6 3.6/207.2
RR 1.95 1.41 3.31 2.44
Vacc 96.3/313/2 58.8/1502.9 23.9/313.2 11.2/1502.9
Sero Pos Placebo 89.8/156.4 137.7/729.8 20/156.4 33.4/729.8
RR 0.50 0.21 0.58 0.16
Numerator data, derived by multiple imputation method, are total symptomatic patients hospitalized or severe dengue. Denominator data are children
in the 10% serological sample.
TABLE 19.4 Hospitalizations and Relative Risk in Three Age Groups of Children, Ages 4–16 Years, Given TAK 003 or Placebo 2 Years Earlier
Hospitalizations Severe
Cases/Total Cases/Total
Categories 4–5 Years 6–11 Years 12–16 Years 4–5 Years 6–11 Years 12–16 Years
Vacc 3/662 4/2200 0/669 1/662 1/2200 1/669
Sero Neg Placebo 3/337 18/1065 3/324 1/337 0 0
RR 0.51 0.11 0.16
Vacc 3/957 3/4808 3/3402 0 1/4808 0
Sero Pos Placebo 4/464 30/2423 17/1700 1/464 5/2423 4/1700
RR 0.36 0.05 0.088
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Dengue Vaccines 283
neutralizing antibodies. Mouse mAbs that bind to the A strand of modelers.226–233 The regulatory and ethical issues associated
epitope cross react with more than one serotype of DENV and with the known occurrence of severe hospitalized dengue in 19
are designated dengue subcomplex neutralizing mAbs. It is also vaccinated seronegatives experiencing their first DENV infec-
becoming clear that the hypothetical structure of the mature tion have not been formally addressed. Without consensus
flavivirus particle generated by cryoelectron microscopy and on the safe use of Dengvaxia or approved serological screen-
molecular fitting does not always predict epitope exposure. ing test(s) these models do not contribute usefully to vaccine
Binding of some E-reactive antibodies depends on the dynamic deployment.
movement of protein molecules (“breathing”) in the virion
particle leading to transient exposure of hidden epitopes.218,219
Recently, studies on DENV antibodies have shifted to those EXCRETION OF VACCINE VIRUS, IF ANY
raised by DENV infection of humans. Technological devel-
opments now enable the rapid generation of human mAbs. Risk of Spread to Contacts
Polyclonal human monotypic dengue-immune sera contain A widely accepted criterion for live DENV vaccines is demon-
mostly weakly neutralizing cross-reactive and enhancing anti- strated low ability to be transmitted by Ae aegypti.
bodies.89,220 DIII antibodies, although highly neutralizing, are
only present in human sera at low levels. Only a small fraction
of antibodies is serotype specific and highly neutralizing and DURATION OF IMMUNITY AND PROTECTION,
these recognize a quaternary structure on the intact virion.89,221 INCLUDING DESCRIPTION OF REINFECTION IF ANY
A similar DENV-1 mAb has been studied by cryoelectron
microscopy.222 Presumably, live-attenuated tetravalent DENV Dengvaxia induced neutralizing antibodies waned to low lev-
vaccines should raise neutralizing antibodies directed at each els in seronegative adults 5 years after receiving a full course
of the four type-specific quaternary structures. This has been of vaccine.
confirmed to be the case for the attenuated DENV-1 developed
by the National Institutes of Health (NIH).187
DENGUE HUMAN INFECTION MODEL (DHIM)
Cellular Responses It is essential that DENV vaccines achieve solid protective
immunity against each of the four viruses following the
Cellular responses to monovalent or tetravalent vaccines in administration of combination vaccines. Recently, vaccine-
seronegative subjects have usually been measured by stimulat- associated ADE was observed in a macaque DENV challenge
ing CD 8+ or CD 4+ T cells with viral antigens. model.174 The vaccine tested had been formalin-inactivated.
It is not known if this model can be extended to test for sen-
Correlates of Protection sitization to ADE by live-attenuated vaccines. In the same
study, viremia following a freeze–thaw cycle for serum con-
As discussed below, the Sanofi Pasteur Phase 2b and Phase 3 taining neutralizing antibody was lower than for challenge
clinical trials have demonstrated that vaccine-induced neutral- of naïve animals though RNAemia was not affected; this
izing antibodies measured in epithelial cell monolayers are not must be considered for challenge models. The absence of
correlates of protection against DENV infection and disease. validated immune protection correlates in humans together
However, when the infection experience of humans has been with an imprecise understanding of protective immunity
a single wild-type DENV, homotypic neutralizing antibodies have crippled dengue vaccine development. Early tests of
are a protection correlate as demonstrated in challenge experi- candidate vaccine protective immunity in humans would be
ments conducted by Sabin.19,223 Following an initial wild-type useful. To that end, the NIH group has used monovalent-
DENV infection in humans, the development of heterospecific attenuated DENV-2 and -3 as challenge viruses (described
neutralizing antibodies correlates with protection against devel- above). Historically, there has been a huge experience of
oping severe disease during a heterotypic DENV infection.224 experimental infections of humans with wild-type DENVs
While these predictive correlates may extend to live-attenuated beginning in the early 1900s with no adverse outcomes repo
viruses, as discussed below, full protection may require infec- rted.6,19,107,108,234–237,239–241
tion by a complete dengue virus, not a chimera, as protective A single dose of LATV solidly protected seronegative
T-cell immunity may depend on presentation of DENV non- human volunteers for short and long periods to challenge
structural antigens.99,195,225 Of interest, administration of the with nonparental wild-type-like DENV-2 Δ30 (Tonga 74) or
monovalent or tetravalent NIH vaccine elicits type-specific and DENV-3 Δ30 (Sleman 78). After a single dose of vaccine was
more broadly reactive human T-cell responses.190,192 given to 20 susceptible volunteers, challenge at 6 months with
DENV-2 Δ30 (Tonga 74) resulted in no viremia, dengue rash,
In Special Groups (Premature Infants, or anamnestic antibody response.185 A similar outcome was
Immunosuppressed) achieved among 20 seronegative adults given a single dose of
LATV and at 6 months challenged with DENV-3. (Durbin, A.,
Dengvaxia is not recommended for infants or immunosup- personal communication, December 28, 2020). In view of this
pressed individuals. experience, a group of investigators at the Walter Reed Army
Institute of Research initiated trials to reestablish a DHIM
EFFICACY AND EFFECTIVENESS using partially attenuated DENV to test vaccine-induced pro-
tective immunity.150,151 Human safety studies on DENV can-
For the Individual Vaccinee didates are in progress at SUNY Upstate School of Medicine,
Efficacy data from Phase 3 have been described above while Syracuse, New York as well as at the University of Maryland
vaccine safety issues are discussed below. with a goal of optimizing the dose that induces a mild den-
gue like illness. At SUNY Upstate Medical University to date,
21 volunteers have been infected with DENV-1 LVHC strain
For the Community 45AZ5 and 6 volunteers with DENV-3 (Endy, T. P., personal
Estimating effectiveness of giving Dengvaxia to communities communication), demonstrating reproducible viremia with
based on epidemiological variables has attracted the attention viral loads equivalent to wild-type infection and a clinical
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
284 SECTION II Licenced Vaccines and Vaccines in Development
illness consistent with mild dengue fever.242 Studies are ongo- vaccinating partially immune humans is good news, but it is
ing at the University of Maryland where this strain of DENV-1 troubling that CYD vaccines do not present DENV NS1 anti-
is being used to assess the performance of a new dengue vac- gens to the immune system. As described above, new evidence
cine in clinical development. No serious adverse events were strongly suggests that NS1 is a viral toxin and the direct cause
noted. The DHIM using these attenuated viruses is safe, result- of the DVPS.47,50
ing in clinical illness consistent with wild-type dengue infec-
tions. In addition to simplifying, accelerating, and reducing
costs of dengue vaccine development, the human challenge
INDICATIONS FOR VACCINE—WHO AND WHY
model should help achieve a better understanding of clinical, Licensed for individuals age 9 years and older with evidence of
pathophysiological and immunological correlates of disease a prior dengue infection.
responses, evaluate anti-DENV drugs and study vector-human
interactions. A crucial benefit of DHIM challenge tests of
candidate vaccines is the identification of reactogenic and/or
CONTRAINDICATIONS AND PRECAUTIONS
nonprotective vaccines early in the development process.243,244 Dengvaxia should not be given to pregnant women or to sero-
negatives of any age.
SAFETY (ADVERSE EVENTS)
Two safety issues have been identified for live-attenuated
PUBLIC HEALTH CONSIDERATIONS
dengue vaccines: (a) preexisting dengue or nondengue fla- There are no meaningful published data on the public health
vivirus antibodies in the vaccinated person may increase outcomes of large-scale administration of Dengvaxia. The
the reactogenicity of the attenuated vaccine virus, and (b) potential adverse outcomes and possible deaths in seronega-
in the event of primary or secondary vaccine failure or wan- tive 9-year-old Philippine children have received comment,
ing neutralizing antibody, enhanced infection and disease but organized Phase 4 studies of postvaccination outcomes
may accompany breakthrough wild-type DENV infections. have not been undertaken at sufficient scale.217,254
With respect to the first issue, no dengue-like symptoms
accompanied administration of vaccine to seropositive chil-
dren.245,246 However, months after vaccination an increased
FUTURE VACCINES (BRIEF)
relative risk of hospitalizations of vaccinated children, ages In addition to the candidate vaccines described here, many
2–5 years, was observed. A WHO advisory committee labeled other recombinant DENV vaccines have been produced, some
this a “safety signal,” while soon, others found vaccine to be reaching Phase 1.
a causal association.217,247–249
CYD dengue vaccine was designed to raise type specific
antibodies to structural antigens for each of the four dengue-
Subunit Vaccines
like viruses in seronegatives and group-reactive antibodies Much of this approach entailed mapping DENV T- and B-cell
in seropositives. In addition, it was expected that successive epitopes on linear structural and nonstructural proteins.192,253,255
doses regardless of initial immune status would raise dengue It was assumed that a combination of epitopes could provide
group reactive cross protective neutralizing antibodies. CYD a safe effective vaccine at moderate cost.256 Structural and non-
rarely raised type-specific neutralizing antibodies as defined structural DENV proteins can be produced in large quantities
by the de Silva group to any of the four DENV either in sus- in many expression systems including Escherichia coli,257,258
ceptibles or partial immunes (immune to a single wild-type baculovirus in Spodoptera frugiperda insect cells,259–261 yeast,262
DENV).89,250,251 Whatever the mix of immune responses that vaccinia virus,263–265 and Drosophila cells.266,267 A Phase 1 clini-
follows administration of one or more doses of CYD, these cal trial demonstrated that both the 10 μg and 50 μg formula-
have failed to prevent symptomatic primary DENV infections. tions of DEN1-80E with 1.25 mg of elemental aluminum were
There is an urgent need to solve this problem at a fundamental immunogenic when administered in a three-injection series
level.215 (0, 1, 2 months) to healthy, flavivirus-naïve adults. The vaccine
In addition, based on data from children 5 years of age formulations induced DENV-1 neutralizing antibodies in the
or younger, one or more doses of CYD vaccine produced an majority of subjects. Titers in most subjects were modest and
immune response that followed by a wild-type DENV infection waned over time.268 A tetravalent vaccine consisting of 80% E
resulted in hospitalization due to increased vascular permea- proteins of each of the four dengue viruses was tested for pro-
bility. In retrospect, this outcome could have been anticipated tection in rhesus monkeys. Formulations at low (3, 3, 3, 6 μg
in preclinical testing. All four monotypic CYD viruses should of DEN1-80E, DEN2-80E, DEN3-80E, and DEN4-80E, respec-
have been inoculated into susceptible monkeys who were then tively), medium (10, 10, 10, 20 μg), and high (50, 50, 50, 100
to be challenged at an interval no earlier than 6 months with μg) doses were comparably immunogenic, inducing high titer,
homotypic wild-type virus. This challenge regime should have balanced neutralizing antibodies against all four DENV. Upon
been repeated in susceptible humans who were to be chal- challenge with the four wild-type DENV, all animals in the
lenged with partially attenuated DENVs (see “Dengue Human low and medium dose groups were protected against viremia
Infection Model” later).252 In the Sanofi vaccine clinical trials, while two animals in the high-dose group exhibited break-
vaccination of seropositives provided moderate to good pro- through viremia.269
tection against severe DENV disease. Presumably, this reflects Cuban investigators have produced candidate dengue vac-
a booster immunological effect. cines using recombinant proteins. DENV-3 DIII protein fused
There are several unestablished explanations for the poor with a P64k Neisseria meningitidis carrier protein, adminis-
vaccine protection in seronegatives compared with seroposi- tered with Freund’s complete adjuvant, protected nonhuman
tives. First, CYD vaccines do not present DENV nonstruc- primates after challenge.270 However, only partial protection
tural proteins to the immune system. This is likely to result was obtained using aluminum hydroxide as the adjuvant,
in suboptimal protective CD8+ T cell responses. In humans, and this required use of the Neisseria serogroup A polysac-
nonstructural DENV antigens may contribute importantly to charide.271 The authors have also expressed a domain III
T-cell protection against DENV-2 infections and disease.192,253 capsid chimeric protein of the four DENV in Escherichia coli
The observed protection against severe disease obtained by and are testing this construct as a vaccine.272 Tetra DIIIC, a
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Ooogue Vaccines 285
DNA vaccines consist of a plasmid (or plasmids) contain- synthetic peptides h a n been detected in sera from patients
ing DENY genes reproduced to high copy number in bacteria convalescing from dengue infections. 236• •279•2so Peptides 2 37
such as E. coli. The plasmid contains a eukaryotic promoter are unlikely to raise antibodies that will completely protect
and termination sequence to drive transcription in the vac- against DENY infection as they may provide fewer of the con-
cine recipient. The transcribed RNA is translated to produce formational epitopes required.
proteins to be processed and presented to the immune system
in the context o f MHC molecules. Additional genes such as KEY REFERENCES
intracellular trafficking and immunostimulatory sequences
can be added to the plasmid. Expressed antigens lead to B- 15. Powell JR, Gloria-Soria A, Kotsakiozi P. Recent history of Aed.is
aegypti: vector genomics and epidemiology records. Bioscience.
and T-cell responses. DNA vaccines afford numerous theo- 2018;68:854-860.
retical advantages over conventional vaccines, including ease 41. World Health Organization. Dengue Haemorrhagic Fever: Diag-
of production, stability and ability to be transported at room nosis, 'Treatment, Prevention and Control. 2nd ed. Geneva: World
temperature, ability to add new genes to the vaccine, abil- Health Organiz.alion, 1997.
ity to immunize with or without adjuvant systems against 52. WHO. Denglli: Guuulines fOT Diagnosis, Jroatment Prevention and
multiple pathogens with a single construct, and reduced Conl:Tol. Geneva: WHO; 2009.
reactogenicity. 274 61. Nimmannitya S, Halstead SB, Cohen S, et al. Dengue and chi-
kungunya virus infeclion in man in Thailand, 1962-1964. I.
Observations on hospitalized patients with hemorrhagic fever.
Vectored Vaccines Am J 'Trop Med Hyg. 1969;18:954-971.
Recombinant po:xviruses, adenoviruses, and measles viruses 66. Halstead SB, Russell PK, Brandt WE. NSl, dengue's dagger.
J Infect Dis. 2020;221 :857-860.
expressing foreign proteins have been demonstrated to induce 216. Sridhar S, Luedtke A, Langevin E, et al. Effect of dengue serosla-
strong humoral and cellular responses in humans against tus on dengue vacdne safety and efficacy. N Engl J Med.
various pathogens. These viruses can infect cells and express 2018;379:327-340.
their proteins de novo in the cell. The antigens are then nat- 21 7. Halstead SB, Russell PK, Katzelnick LC, et al. Ethics of a partially
urally processed, glycosylated, and associated with the cell effective dengue vaccine: lessons from the Philippines. Vaccine.
membrane. MHC class I-dependent immune responses are 2020;38:5572-5576.
induced as a result of intracellular translation and processing
of the gene products. Visit eBooks.Health.Elsevier.com for a complete reference Ii.st @
Descargado para Alonso Hmnberto Bolbaran Castillo (alonso.boll,[email protected]) en Universiry o f Chili, de Clinica!Ki,y.es por Elsevier en mayo 06,
2024. Para uso personal ex.clusivameole. No se permiten otros usos sin aulorizaci6n. Copyrigbl ©2024. Elsevier fuc. Todos los de:redws resenrados.
Dengue Vaccines 285.e1
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
285.e2 SECTION II Licenced Vaccines and Vaccines in Development
54. Kanai R, Kar K, Anthony K, et al. Crystal structure of West Nile 80. Gubler DJ. Epidemic dengue/dengue hemorrhagic fever as a
virus envelope glycoprotein reveals viral surface epitopes. J. Virol. public health, social and economic problem in the 21st century.
2006;80:11000–11008. Trends Microbiol. 2002;10:100–103.
55. Modis Y, Ogata SA, Clements D, et al. A ligand-binding pocket in 81. Murray KA, Escobar LE, Lowe R, et al. Tracking infectious diseases
the dengue virus envelope glycoprotein. Proc Natl Acad Sci USA. in a warming world. BMJ. 2020;371:m3086.
2003;100:6986–6991. 82. Hervy JP. Experience de marquage-lacher-recapture portant sur
56. Rey FA, Heinz FX, Mandl C, et al. The envelope glycoprotein from Aedes aegypti Linne, en zone de savane soudanienne ouest-afric-
tick-borne encephalitis virus at 2 angstrom resolution. Nature. aine. 1. Le cycle gonotrophique. Cahiers l’ORSTOM Ser Enntomoog
1995;375:291–298. Med Parasitol. 1977;22:135–143.
57. Zhang Y, Zhang W, Ogata S, et al. Conformational changes of the 83. Pant CP, Yasuno M. Field studies on the gonotrophic cycle of Aedes
flavivirus E glycoprotein. Structure. 2004;12:1607–1618. aegypti in Bangkok, Thailand. J Med Entomol. 1973;10:219–223.
58. Roehrig JT. Antigenic structure of flavivirus proteins. Adv Virus 84. Trpis M, Haussermann W. Dispersal and other population
Res. 2003;59:141–175. parameters on Aedes aegypti in an African village and their pos-
59. Halstead SB, Nimmannitya S, Cohen SN. Observations related to sible significance in epidemiology of vector-borne diseases. Am J
pathogenesis of dengue hemorrhagic fever. IV. Relation of disease Trop Med Hyg. 1986;35:1263–1279.
severity to antibody response and virus recovered. Yale J Biol Med. 85. Anderson KB, Gibbons RV, Cummings DA, et al. A shorter time
1970;42:311–328. interval between first and second dengue infections is associated
60. Halstead SB. Immunological parameters of Togavirus disease with protection from clinical illness in a school-based cohort in
syndromes. In: Schlesinger RW, ed. The Togaviruses, Biology, Struc- Thailand. J Infect Dis. 2013;209:360–368.
ture, Replication. New York: Academic Press; 1980:107–173.
86. Guzman MG, Alvarez M, Halstead SB. Secondary infection as
61. Nimmannitya S, Halstead SB, Cohen S, et al. Dengue and chi-
a risk factor for dengue hemorrhagic fever/dengue shock syn-
kungunya virus infection in man in Thailand, 1962–1964. I.
drome: an historical perspective and role of antibody-dependent
Observations on hospitalized patients with hemorrhagic fever.
enhancement of infection. Arch Virol. 2013;158:1445–1459.
Am J Trop Med Hyg. 1969;18:954–971.
87. Gibbons RV, Kalanarooj S, Jarman RG, et al. Analysis of repeat
62. Halstead SB. Pathogenesis of dengue: challenges to molecular
hospital admissions for dengue to estimate the frequency of third
biology. Science. 1988;239:476–481.
or fourth dengue infections resulting in admissions and dengue
63. Ubol S, Halstead SB. How innate immune mechanisms contrib-
hemorrhagic fever, and serotype sequences. Am J Trop Med Hyg.
ute to antibody-enhanced viral infections. Clin Vaccine Immunol.
2007;77:910–913.
2010;17:1829–1835.
88. Halstead SB, Lan NT, Myint TT, et al. Infant dengue hemor-
64. Vaughn DW, Green S, Kalayanarooj S, et al. Dengue viremia titer,
rhagic fever: research opportunities ignored. Emerg Infect Dis.
antibody response pattern, and virus serotype correlate with dis-
2002;12:1474–1479.
ease severity. J Infect Dis. 2000;181:2–9.
65. Halstead SB. Neutralization and antibody dependent enhance- 89. de Alwis R, Smith SA, Olivarez NP, et al. Identification of human
ment of dengue viruses. Adv Virus Res. 2003;60:421–467. neutralizing antibodies that bind to complex epitopes on dengue
66. Halstead SB, Russell PK, Brandt WE. NS1, dengue’s dagger. J virions. Proc Natl Acad Sci USA. 2012;109:7439–7444.
Infect Dis. 2020;221:857–860. 90. Halstead SB, Palumbo NE. Studies on the immunization of mon-
67. Green S, Rothman A. Immunopathological mechanisms in keys against dengue: II. Protection following inoculation of com-
dengue and dengue hemorrhagic fever. Curr Opin Infect Dis. binations of viruses. Am J Trop Med Hyg. 1973;22:375–381.
2006;19:429–436. 91. Alcon-LePoder S, Drouet MT, Roux P, et al. The secreted form
68. Huang YH, Lei HY, Liu HS, et al. Dengue virus infects human of dengue virus nonstructural protein NS1 is endocytosed by
endothelial cells and induces IL-6 and IL-8 production. Am J Trop hepatocytes and accumulates in late endosomes: implications
Med Hyg. 2000;63:71–75. for viral infectivity. J Viol. 2005;79:11403–11411.
69. Lin CF, Lei HY, Shiau AL, et al. Antibodies from dengue patient 92. Falconar AK, Young PR. Production of dimer-specific and den-
sera cross-react with endothelial cells and induce damage. J Med gue virus group cross-reactive mouse monoclonal antibodies to
Virol. 2003;69:82–90. the dengue 2 virus non-structural glycoprotein NS1. J Gen Virol.
70. Mongkolsapaya J, Dejnirattisai W, Xu XN, et al. Original anti- 1991;72:961–965.
genic sin and apoptosis in the pathogenesis of dengue hemor- 93. Young PR, Hilditch PA, Bletchly C, et al. An antigen capture
rhagic fever. Nat Med. 2003;9:921–927. enzyme-linked immunosorbent assay reveals high levels of the
71. Rosen L. The emperor’s new clothes revisited, or reflections on dengue virus protein NS1 in the sera of infected patients. J Clin
the pathogenesis of dengue hemorrhagic fever. Am J Trop Med Microbiol. 2000;38:1053–1057.
Hyg. 1977;26:337–343. 94. Innis BL, Nisalak A, Nimmannitya S, et al. An enzyme-linked
72. Rosen L. The pathogenesis of dengue haemorrhagic fever. A critical immunosorbent assay to characterize dengue infections where
appraisal of current hypotheses. S Afr Med J. 1986;70(suppl):40–42. dengue and Japanese encephalitis co-circulate. Am J Trop Med
73. Rothman AL. T lymphocyte responses to heterologous secondary Hyg. 1989;40:418–427.
dengue virus infections. Ann NY Acad Sci. 2009;1171(suppl 1): 95. Thomas SJ. The necessity and quandaries of dengue vaccine
E36–E41. development. J Infect Dis. 2011;203:299–303.
74. Aye KS, Charngkaew K, Win N, et al. Pathologic highlights of 96. Montoya M, Gresh L, Mercado JC, et al. Symptomatic versus
dengue hemorrhagic fever in 13 autopsy cases from Myanmar. inapparent outcome in repeat dengue virus infections is influ-
Hum Pathol. 2014;45:1221–1233. enced by the time interval between infections and study year.
75. Win MM, Charngkaew K, Punyadee N, et al. Ultrastructural fea- PLoS Negl Trop Dis. 2013;7:e2357.
tures of human liver specimens from patients who died of den- 97. Alvarez M, Rodriguez-Roche R, Bernardo L, et al. Dengue hemor-
gue hemorrhagic fever. Trop Med Infect Dis. 2019;4:13. rhagic fever caused by sequential dengue 1-3 virus infections over
76. Anders KL, Nguyen NM, Van Thuy NT, et al. A birth cohort study a long time interval: Havana epidemic, 2001-2002. Am J Trop Med
of viral infections in Vietnamese infants and children: study Hyg. 2006;75:1113–1117.
design, methods and characteristics of the cohort. BMC Public 98. Guzman MG, Kouri G, Valdes L, et al. Epidemiologic studies on den-
Health. 2013;13:937. gue in Santiago de Cuba, 1997. Am J Epidemiol. 2000;152:793–799.
77. Duyen HT, Ngoc TV, Ha DT, et al. Kinetics of plasma viremia and 99. Weiskopf D, Angelo MA, de Azeredo EL, et al. Comprehensive
soluble nonstructural protein 1 concentrations in dengue: dif- analysis of dengue virus-specific responses supports an HLA-
ferential effects according to serotype and immune status. J Infect linked protective role for CD8+ T cells. Proc Natl Acad Sci USA.
Dis. 2011;203(9):1292–1300. 2013;110:E2046–E2053.
78. Thai KT, Phuong HL, Thanh Nga TT, et al. Clinical, epidemio- 100. Yauch LE, Zellweger RM, Kotturi MF, et al. A protective role for den-
logical and virological features of Dengue virus infections in Viet- gue virus-specific CD8+ T cells. J Immunol. 2009;182:4865–4873.
namese patients presenting to primary care facilities with acute 101. Kyle JL, Balsitis SJ, Zhang L, et al. Antibodies play a greater
undifferentiated fever. J Infect. 2010;60:229–237. role than immune cells in heterologous protection against
79. WHO. Handbook for Clinical Managent of Dengue. Geneva: secondary dengue virus infection in a mouse model. Virology.
WHO; 2012:111. 2008;380:296–303.
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Dengue Vaccines 285.e3
102. Pulendran B. Learning immunology from the yellow fever vac- 131. Bhamarapravati N, Yoksan S. Live attenuated tetravalent dengue
cine: innate immunity to systems vaccinology. Nat Rev Immunol. vaccine. In: Gubler DJ, Kuno G, eds. Dengue and Dengue Hemor- 19
2009;9:741–747. rhagic Fever. New York: CAB International; 1997:367–377.
103. Querec TD, Akondy RS, Lee EK, et al. Systems biology approach 132. Bhamarapravati N, Yoksan S, Chayaniyayothin T, et al. Immu-
predicts immunogenicity of the yellow fever vaccine in humans. nization with a live attenuated dengue-2-virus candidate
Nat Immunol. 2009;10:116–125. vaccine (16681-PDK 53): clinical, immunological and bio-
104. Tandan JB, Ohrr H, Sohn YM, et al. Single dose of SA 14-14-2 vac- logical responses in adult volunteers. Bull World Health Organ.
cine provides long-term protection against Japanese encephalitis: 1987;65:189–195.
a case-control study in Nepalese children 5 years after immuniza- 133. Vaughn DW, Green S, Kalayanarooj S, et al. Peak dengue 2 virus
tion. Vaccine. 2007;25:5041–5045. titer early in secondary infection correlates with plasma leakage. In:
105. Yu Y. Phenotypic and genotypic characteristics of Japanese Paper Presented at: 45th Meeting of the American Society of Tropical
encephalitis attenuated live vaccine virus SA14-14-2 and their Medicine and Hygiene; 1-5 December,1996; Baltimore, MD .
stabilities. Vaccine. 2010;28:3635–3641. 134. Kanesa-thasan N, Sun W, Kim-Ahn G, et al. Safety and immuno-
genicity of attenuated dengue virus vaccines (Aventis Pasteur) in
106. Blanc G, Caminopetros J. Recherches experimentales sur la den-
human volunteers. Vaccine. 2001;19:3179–3188.
gue. Ann Inst Pasteur (Paris). 1930;44:367–436.
135. Bhamarapravati N, Yoksan S. Study of bivalent dengue vaccine in
107. Hotta S. Experimental studies in dengue I. Isolation, identifica-
volunteers. Lancet. 1989;1:1077.
tion and modification of the virus. J Infect Dis. 1952;90:1–9.
136. Sun W, Nisalak A, Gettayacamin M, et al. Protection of Rhe-
108. Sabin AB, Schlesinger RW. Production of immunity to dengue with sus monkeys against dengue virus challenge after tetrava-
virus modified by propagation in mice. Science. 1945;101:640–642. lent live attenuated dengue virus vaccination. J Infect Dis.
109. Schlesinger RW, Gordon I, Frankel JW, et al. Clinical and sero- 2006;193:1658–1665.
logic response of man to immunization with attenuated dengue 137. Chanthavanich P, Luxemburger C, Sirivichayakul C, et al. Short
and yellow fever viruses. J Immunol. 1956;77:352–364. report: immune response and occurrence of dengue infec-
110. Wisseman Jr CL, Sweet BH, Rosenzweig EC, et al. Attenuated liv- tion in Thai children three to eight years after vaccination with
ing type 1 dengue vaccines. Am J Trop Med Hyg. 1963;12:620–623. live attenuated tetravalent dengue vaccine. Am J Trop Med Hyg.
111. Blaney Jr JE, Durbin AP, Murphy BR, et al. Targeted mutagen- 2006;75:26–28.
esis as a rational approach to dengue virus vaccine development. 138. Chokephaibulkit K, Sirivichayakul C, Thisyakorn U, et al. Safety
Curr Top Microbiol Immunol. 2010;338:145–158. and immunogenicity of a single administration of live-attenu-
112. Chavez JH, Silva JR, Amarilla AA, et al. Domain III peptides from ated Japanese encephalitis vaccine in previously primed 2- to
flavivirus envelope protein are useful antigens for serologic diag- 5-year-olds and naive 12- to 24-month-olds: multicenter ran-
nosis and targets for immunization. Biologicals. 2010;38:613–618. domized controlled trial. Pediatr Infect Dis J. 2010;29:1111–1117.
113. Durbin AP, Whitehead SS. Dengue vaccine candidates in devel- 139. Sabchareon A, Lang J, Chanthavanich P, et al. Safety and immu-
opment. Curr Top Microbiol Immunol. 2010;338:129–143. nogenicity of a three dose regimen of two tetravalent live-atten-
114. Faheem M, Raheel U, Riaz MN, et al. A molecular evaluation of uated dengue vaccines in five- to twelve-year-old Thai children.
dengue virus pathogenesis and its latest vaccine strategies. Mol Pediatr Infect Dis J. 2004;23:99–109.
Biol Rep. 2011;38:3731–3740. 140. Sabchareon A, Lang J, Chanthavanich P, et al. Safety and immu-
115. Konishi E. Issues related to recent dengue vaccine development. nogenicity of tetravalent live-attenuated dengue vaccines in Thai
Trop Med Health. 2012;39(4 suppl):63–71. adult volunteers: role of serotype concentration, ratio, and mul-
116. Malabadi RB, Ganguly A, Silva JA, et al. Overview of plant- tiple doses. Am J Trop Med Hyg. 2002;66:264–272.
derived vaccine antigens: dengue virus. J Pharm Pharm Sci. 141. Sabchareon A, Wallace D, Sirivichayakul C, et al. Protective effi-
2012;14:400–413. cacy of the recombinant, live-attenuated, CYD tetravalent dengue
117. Murphy BR, Whitehead SS. Immune response to dengue virus vaccine in Thai schoolchildren: a randomised, controlled phase
and prospects for a vaccine. Annu Rev Immunol. 2011;29:587–619. 2b trial. Lancet. 2012;380:1559–1567.
118. Schmitz J, Roehrig J, Barrett A, et al. Next generation dengue vac- 142. Sanchez V, Gimenez S, Tomlinson B, et al. Innate and adaptive
cines: a review of candidates in preclinical development. Vaccine. cellular immunity in flavivirus-naive human recipients of a live-
2011;29:7276–7284. attenuated dengue serotype 3 vaccine produced in Vero cells
119. Thomas SJ. Developing a dengue vaccine: progress and future (VDV3). Vaccine. 2006;24:4914–4926.
challenges. Ann N Y Acad Sci. 2014;1323:140–159. 143. Bauer K, Esquilin IO, Cornier AS, et al. A Phase II, randomized,
120. Thomas SJ, Endy TP. Critical issues in dengue vaccine develop- safety and immunogenicity trial of a re-derived, live-attenuated
ment. Curr Opin Infect Dis. 2011;24:442–450. dengue virus vaccine in healthy children and adults living in
121. Thomas SJ, Endy TP. Current issues in dengue vaccination. Curr Puerto Rico. Am J Trop Med Hyg. 2015;93:441–453.
Opin Infect Dis. 2013;26:429–434. 144. Briggs CM, Smith KM, Piper A, et al. Live attenuated tetrava-
122. Thomas SJ, Endy TP. Vaccines for the prevention of dengue: lent dengue virus host range vaccine is immunogenic in Afri-
development update. Hum Vaccin. 2011;7:674–684. can green monkeys following a single vaccination. J Virol.
123. Webster DP, Farrar J, Rowland-Jones S. Progress towards a dengue 2014;88:6729–6742.
vaccine. Lancet Infect Dis. 2009;9:678–687. 145. Simasathien S, Thomas SJ, Watanaveeradej V, et al. Safety and
124. Halstead SB, Sukhavachana P, Nisalak A. In vitro recovery of den- immunogenicity of a tetravalent live-attenuated dengue vac-
gue viruses from naturally infected human beings and arthro- cine in flavivirus naive children. Am J Trop Med Hyg. 2008;78:
pods. Nature. 1964;202:931–932. 426–433.
125. Halstead SB. Studies on the attenuation of dengue 4. Asian J Infect 146. Sun W, Cunningham D, Wasserman SS, et al. Phase 2 clinical
Dis. 1978;2:112–117. trial of three formulations of tetravalent live-attenuated dengue
126. Halstead SB, Marchette NJ. Biologic properties of dengue viruses vaccine in flavivirus-naive adults. Hum Vaccin. 2009;5:33–40.
following serial passage in primary dog kidney cells: studies at the 147. Thomas SJ, Eckels KH, Carletti I, et al. A Phase II, randomized,
University of Hawaii. Am J Trop Med Hyg. 2003;69(6 suppl):5–11. safety and immunogenicity study of a re-derived, live-attenu-
127. Innis BL, Eckels KH. Progress in development of a live-attenu- ated dengue virus vaccine in healthy adults. Am J Trop Med Hyg.
ated, tetravalent dengue virus vaccine by the United States Army 2013;88:73–88.
Medical Research and Materal Command. Am J Trop Med Hyg. 148. Watanaveeradej V, Gibbons RV, Simasathien S, et al. Safety and
2003;69:1–4. immunogenicity of a rederived, live-attenuated dengue virus vac-
128. Halstead SB. Neutralization and antibody-dependent enhance- cine in healthy adults living in Thailand: a randomized trial. Am
ment of dengue viruses. In: Chambers TJ, Monath TP, eds. The J Trop Med Hyg. 2014;91:119–128.
Flaviviruses: Pathogenesis and Immunity. Vol 60 New York: Elsevier 149. Watanaveeradej V, Simasathien S, Nisalak A, et al. Safety and
Academic Press; 2003:422–467. immunogenicity of a tetravalent live-attenuated dengue vac-
129. Yoksan S. Short history of dengue and Mahidol dengue vaccine. cine in flavivirus-naive infants. Am J Trop Med Hyg. 2011;85:
South East Asian J Trop Med Pub Health. 2017;48(suppl 1):20–32. 341–351.
130. Bhamarapravati N, Sutee Y. Live attenuated tetravalent dengue 150. Mammen MP, Lyons A, Innis BL, et al. Evaluation of dengue virus
vaccine. Vaccine. 2000;18(suppl 2):44–47. strains for human challenge studies. Vaccine. 2014;14:1488–1494.
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
285.e4 SECTION II Licenced Vaccines and Vaccines in Development
151. Sun W, Eckels KH, Putnak JR, et al. Experimental dengue administered on varying schedules to healthy U.S. adults: a Phase
virus challenge of human subjects previously vaccinated 1/2 randomized study. Am J Trop Med Hyg. 2020;103:132–141.
with live attenuated tetravalent dengue vaccines. J Infect Dis. 173. Martinez LJ, Lin L, Blaylock JM, et al. Safety and Immunogenicity
2013;207:700–708. of a dengue virus serotype-1 purified-inactivated vaccine: results
152. Porter KR, Kochel TJ, Wu SJ, et al. Protective efficacy of a dengue 2 of a Phase 1 clinical trial. Am J Trop Med Hyg. 2015;93:454–460.
DNA vaccine in mice and the effect of CpG immuno-stimulatory 174. Borges MB, Marchevsky RS, Carvalho Pereira R, et al. Detection of
motifs on antibody responses. Arch Virol. 1998;143:997–1003. post-vaccination enhanced dengue virus infection in macaques:
153. Raviprakash K, Ewing D, Simmons M, et al. Needle-free biojec- an improved model for early assessment of dengue vaccines.
tor injection of a dengue virus type 1 DNA vaccine with human PLoS Pathog. 2019;15:e1007721.
immunostimulatory sequences and the GM-CSF gene increases 175. Butrapet S, Huang CY, Pierro DJ, et al. Attenuation markers of a
immunogenicity and protection from virus challenge in Aotus candidate dengue type 2 vaccine virus, strain 16681 (PDK-53),
monkeys. Virology. 2003;315:345–352. are defined by mutations in the 5′ noncoding region and non-
154. Raviprakash K, Kochel TJ, Ewing D, et al. Immunogenicity of structural proteins 1 and 3. J Virol. 2000;74:3011–3019.
dengue virus type 1 DNA vaccines expressing truncated and full 176. Huang CY, Kinney RM, Livengood JA, et al. Genetic and pheno-
length envelope protein. Vaccine. 2000;18:2426–2434. typic characterization of manufacturing seeds for a tetravalent
155. Raviprakash K, Porter KR, Kochel TJ, et al. Dengue virus type 1 dengue vaccine (DENVax). PLoS Negl Trop Dis. 2013;7:e2243.
DNA vaccine induces protective immune responses in rhesus 177. Biswal S, Reynales H, Saez-Llorens X, et al. Efficacy of a tetrava-
macaques. J Gen Virol. 2000;81(Pt 7):1659–1667. lent dengue vaccine in healthy children and adolescents. N Engl J
156. Blair PJ, Kochel TJ, Raviprakash K, et al. Evaluation of immu- Med. 2019;381:2009–2019.
nity and protective efficacy of a dengue-3 pre-membrane and 178. Sirivichayakul C, Barranco-Santana EA, Rivera IE, et al. Long-term
envelope DNA vaccine in Aotus nancymae monkeys. Vaccine. safety and immunogenicity of a tetravalent dengue vaccine can-
2006;24:1427–1432. didate in children and adults: a randomized, placebo-controlled,
157. Wu SJ, Grouard-Vogel G, Sun W, et al. Human skin Lang- phase 2 study. J Infect Dis. 2020;243. doi:10.1083/jiaa.406.
erhans cells are targets of dengue virus infection. Nat Med. 179. Tricou V, Sáez-Llorens X, Yu D, et al. Safety and immunoge-
2000;6:816–820. nicity of a tetravalent dengue vaccine in children aged 2-17
158. Porter KR, Teneza-Mora N, Raviprakash K. Tetravalent DNA vac- years: a randomised, placebo-controlled, phase 2 trial. Lancet.
cine product as a vaccine candidate against dengue. Methods Mol 2020;395:1434–1443.
Biol. 2014;1143:283–295. 180. López-Medina E, Biswal S, Saez-Llorens X, et al. Efficacy of a den-
159. Beckett CG, Tjaden J, Burgess T, et al. Evaluation of a proto- gue vaccine candidate (TAK-003) in healthy children and adoles-
type dengue-1 DNA vaccine in a Phase 1 clinical trial. Vaccine. cents two years after vaccination. J Infect Dis. 2021. doi:1-.1093/
2011;29:960–968. infdis/jiaa761.
160. Porter KR, Ewing D, Chen L, et al. Immunogenicity and protec- 181. Durbin AP, Kirkpatrick BD, Pierce KK, et al. A single dose of any
tive efficacy of a vaxfectin-adjuvanted tetravalent dengue DNA of four different live attenuated tetravalent dengue vaccines is
vaccine. Vaccine. 2012;30:336–341. safe and immunogenic in flavivirus-naive adults: a randomized,
161. Danko JR, Kochel T, Teneza-Mora N, et al. Safety and immunoge- double-blind clinical trial. J Infect Dis. 2013;207:957–965.
nicity of a tetravalent dengue DNA vaccine administered with a 182. Durbin AP, Whitehead SS. Next-generation dengue vaccines:
cationic lipid-based adjuvant in a Phase 1 clinical trial. Am J Trop novel strategies currently under development. Viruses. 2011;3:
Med Hyg. 2018;98:849–856. 1800–1814.
162. Williams M, Ewing D, Blevins M, et al. Enhanced immunoge- 183. Kirkpatrick BD, Durbin AP, Pierce KK, et al. Robust and balanced
nicity and protective efficacy of a tetravalent dengue DNA vac- immune responses to all 4 dengue virus serotypes following
cine using electroporation and intradermal delivery. Vaccine. administration of a single dose of a live attenuated tetravalent
2019;37:4444–4453. dengue vaccine to healthy, flavivirus-naive adults. J Infect Dis.
163. Simmons M, Burgess T, Lynch J, et al. Protection against den- 2015;212:702–710.
gue virus by non-replicating and live attenuated vaccines 184. Whitehead SS. Development of TV003/TV005, a single dose,
used together in a prime boost vaccination strategy. Virology. highly immunogenic live attenuated dengue vaccine; what
2010;396:280–288. makes this vaccine different from the Sanofi-Pasteur CYD vac-
164. Simmons M, Porter KR, Hayes CG, et al. Characterization of anti- cine? Expert Rev Vaccines. 2016;15:509–517.
body responses to combinations of a dengue virus type 2 DNA 185. Kirkpatrick BD, Whitehead SS, Pierce KK, et al. The live attenu-
vaccine and two dengue virus type 2 protein vaccines in rhesus ated dengue vaccine TV003 elicits complete protection against
macaques. J Virol. 2006;80:9577–9585. dengue in a human challenge model. Sci Transl Med. 2016;8:
165. Apt D, Raviprakash K, Brinkman A, et al. Tetravalent neutralizing 330ra336.
antibody response against four dengue serotypes by a single chi- 186. Durbin AP, Kirkpatrick BD, Pierce KK, et al. A 12-month interval
meric dengue envelope antigen. Vaccine. 2006;24:335–344. dosing study in adults indicates that a single dose of the NIAID
166. Raviprakash K, Apt D, Brinkman A, et al. A chimeric tetravalent tetravalent dengue vaccine induces a robust neutralizing anti-
dengue DNA vaccine elicits neutralizing antibody to all four body response. J Infect Dis. 2016;214(6):832–835. doi:10.1093/
virus serotypes in rhesus macaques. Virology. 2006;353:166–173. infdis/jiw067.
167. Porter KR, Raviprakash K. Nucleic acid (DNA) immuniza- 187. Smith SA, de Alwis R, Kose N, et al. Human monoclonal anti-
tion as a platform for dengue vaccine development. Vaccine. bodies derived from memory B cells following live attenuated
2015;29(42):7261–7266. dengue virus vaccination or natural infection exhibit similar
168. Fernandez S, Thomas SJ, De La Barrera R, et al. An adjuvanted, characteristics. J Infect Dis. 2013;207:1898–1908.
tetravalent dengue virus purified inactivated vaccine candidate 188. Swanstrom JA, Nivarthi UK, Patel B, et al. Beyond neutral-
induces long-lasting and protective antibody responses against izing antibody levels: the epitope specificity of antibodies
dengue challenge in rhesus macaques. Am J Trop Med Hyg. induced by NIH monovalent dengue virus vaccines. J Infect Dis.
2015;92:698–708. 2019:20(2):219–227.
169. Putnak R, Cassidy K, Conforti N, et al. Immunogenic and pro- 189. Tsai WY, Durbin A, Tsai JJ, et al. Complexity of neutralization
tective response in mice immunized with a purified, inactivated, antibodies against multiple dengue viral serotypes after hetero-
Dengue-2 virus vaccine prototype made in fetal rhesus lung cells. typic immunization and secondary infection revealed by in-depth
Am J Trop Med Hyg. 1996;55:504–510. analysis of cross-reactive antibodies. J Virol. 2015;89:7348–7362.
170. Putnak RJ, Coller BA, Voss G, et al. An evaluation of dengue type-2 190. Angelo MA, Grifoni A, O’Rourke PH, et al. Human CD4+ T cell
inactivated, recombinant subunit, and live-attenuated vaccine can- responses to an attenuated tetravalent dengue vaccine parallel
didates in the rhesus macaque model. Vaccine. 2005;23:4442–4452. those induced by natural infection in magnitude, HLA restric-
171. Diaz C, Lin L, Martinez LJ, et al. Phase 1 randomized study of a tion, and antigen specificity. J Virol. 2017:91(5):e02147–16.
tetravalent dengue purified inactivated vaccine in healthy adults 191. Grifoni A, Voic H, Dhanda SK, et al. T cell responses induced
from Puerto Rico. Am J Trop Med Hyg. 2018;98(5):1435–1443. by attenuated flavivirus vaccination are specific and show lim-
172. Lin L, Lyke KE, Koren M, et al. Safety and immunogenicity of an ited cross-reactivity with other flavivirus species. J Virol. 2020;89.
AS03(B)-adjuvanted inactivated tetravalent dengue virus vaccine doi:10.1128/JVI0089-20.
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Dengue Vaccines 285.e5
192. Weiskopf D, Angelo MA, Bangs DJ, et al. The human CD8+ T 214. WHO. A Toolkit for National Dengue Burden Estimation. Geneva:
cell responses induced by a live attenuated tetravalent dengue World Health Organization; 2018. 19
vaccine are directed against highly conserved epitopes. J Virol. 215. Halstead SB, Russell PK. Protective and immunological behavior
2015;89:120–128. of chimeric yellow fever dengue vaccine. Vaccine. 2016;34:
193. Elong Ngono A, Chen HW, Tang WW, et al. Protective role of 1643–1647.
cross-reactive CD8 T cells against dengue virus infection. EBio- 216. Sridhar S, Luedtke A, Langevin E, et al. Effect of dengue
Medicine. 2016;13:284–293. serostatus on dengue vaccine safety and efficacy. N Engl J Med.
194. Tian Y, Sette A, Weiskopf D. Cytotoxic CD4 T Cells: Differentia- 2018;379:327–340.
tion, Function, and Application to Dengue Virus Infection. Front 217. Halstead SB, Russell PK, Katzelnick LC, et al. Ethics of a partially
Immunol. 2019;10. doi:10.3389/fimmu/2016.00531. effective dengue vaccine: lessons from the Philippines. Vaccine.
195. Weiskopf D, Cerpas C, Angelo MA, et al. Human CD8+ T-cell 2020;38:5572–5576.
responses against the 4 dengue virus serotypes are associated with 218. Lok SM, Kostyuchenko V, Nybakken GE, et al. Binding of a neu-
distinct patterns of protein targets. J Infect Dis. 2015;212:1743–1751. tralizing antibody to dengue virus alters the arrangement of sur-
196. Schlesinger JJ, Brandriss MW, Walsh EE. Protection of mice face glycoproteins. Nat Struct Mol Biol. 2008;15:312–317.
against dengue 2 virus encephalitis by immunization with the 219. Pierson TC, Kuhn RJ. Capturing a virus while it catches its breath.
dengue 2 virus non-structural glycoprotein NS1. J Gen Virol. Structure. 2012;20:200–202.
1987;68:853–857. 220. Beltramello M, Williams KL, Simmons CP, et al. The human
197. Kallas EG, Precioso AR, Palacios R, et al. Safety and immunogenic- immune response to Dengue virus is dominated by highly cross-
ity of the tetravalent, live-attenuated dengue vaccine Butantan-DV reactive antibodies endowed with neutralizing and enhancing
in adults in Brazil: a two-step, double-blind, randomised placebo- activity. Cell Host Microbe. 2010;8:271–283.
controlled phase 2 trial. Lancet Infect Dis. 2020;20:839–850. 221. de Alwis R, Beltramello M, Messer WB, et al. In-depth analysis of
198. Lopes TRR, Silva CS, Pastor AF, et al. Dengue in Brazil in 2017: the antibody response of individuals exposed to primary dengue
what happened? Rev Inst Med Trop Sao Paulo. 2018;60:e43. virus infection. PLoS Negl Trop Dis. 2011;5:e1188.
198a. Mohanty L, Prabhu M, Kumar Mishra A, et al. Safety and immu- 222. Teoh EP, Kukkaro P, Teo EW, et al. The structural basis for sero-
nogenicity of a single dose, live-attenuated ‘tetravalent dengue type-specific neutralization of dengue virus by a human anti-
vaccine’ in healthy Indian adults; a randomized, double-blind, body. Sci Transl Med. 2012;4:139ra183.
placebo controlled phase I/II trial. Vaccine: X 2022; 10: 100142. 223. Snow GE, Haaland B, Ooi EE, et al. Review article: Research
199. Chambers TJ, Nestorowicz A, Mason PW, et al. Yellow fever/Japa- on dengue during World War II revisited. Am J Trop Med Hyg.
nese encephalitis chimeric viruses: construction and biological 2014;91:1203–1217.
properties. J Virol. 1999;73:3095–3101. 224. Corbett KS, Katzelnick L, Tissera H, et al. Preexisting neutralizing
200. Rice CM, Lenches EM, Eddy SR, et al. Nucleotide sequence of yel- antibody responses distinguish clinically inapparent and appar-
low fever viruses: implications for flavivirus gene expression and ent dengue virus infections in a Sri Lankan pediatric cohort.
evolution. Science. 1985;229:726–733. J Infect Dis. 2015;211:590–599.
201. Guirakhoo F, Weltzin R, Chambers TJ, et al. Recombinant chime- 225. Weiskopf D, Angelo MA, Sidney J, et al. Immunodominance
ric yellow fever-dengue type 2 virus is immunogenic and protec- changes as a function of the infecting dengue virus serotype and
tive in nonhuman primates. J Virol. 2000;74:5477–5485. primary versus secondary infection. J Virol. 2014;88:11383–11394.
202. Pugachev KV, Guirakhoo F, Ocran SW, et al. High fidelity of yel- 226. Cardim LL, Pinho STR, Teixeira MG, et al. Heterogeneities in
low fever virus RNA polymerase. J Virol. 2004;78:1032–1038. dengue spatial-temporal transmission in Brazilian cities and its
203. Arroyo J, Guirakhoo F, Fenner S, et al. Molecular basis for influence on the optimal age of vaccination. BMC Public Health.
attenuation of neurovirulence of a yellow fever virus/Japanese 2019;19:155.
encephalitis virus chimera vaccine (ChimeriVax-JE). J Virol. 227. Carvalho SA, da Silva SO, Charret IDC. Mathematical mod-
2001;75:934–942. eling of dengue epidemic: control methods and vaccination
204. McGee CE, Lewis MG, Claire MS, et al. Recombinant chimeric strategies. Theory Biosci = Theorie Biowissenschaften. 2019;138:
virus with wild-type dengue 4 virus premembrane and enve- 223–239.
lope and virulent yellow fever virus Asibi backbone sequences 228. Iboi EA, Gumel AB. Mathematical assessment of the role of
is dramatically attenuated in nonhuman primates. J Infect Dis. Dengvaxia vaccine on the transmission dynamics of dengue sero-
2008;197:693–697. types. Math Biosci. 2018;304:25–47.
205. McGee CE, Tsetsarkin K, Vanlandingham DL, et al. Substitu- 229. Kurauchi A, Struchiner CJ, Wilder-Smith A, et al. Modelling the
tion of wild-type yellow fever Asibi sequences for 17D vaccine effect of a dengue vaccine on reducing the evolution of resistance
sequences in ChimeriVax-dengue 4 does not enhance infection against antibiotic due to misuse in dengue cases. Theor Biol Med
of Aedes aegypti mosquitoes. J Infect Dis. 2008;197:686–692. Modell. 2020;17:7.
206. Guirakhoo F, Zhang ZX, Chambers TJ, et al. Immunogenicity, 230. Pearson CAB, Abbas KM, Clifford S, et al. Serostatus testing
genetic stability, and protective efficacy of a recombinant, chime- and dengue vaccine cost-benefit thresholds. J R Soc Interface.
ric yellow fever-Japanese encephalitis virus (ChimeriVax-JE) as a 2019;16:20190234.
live, attenuated vaccine candidate against Japanese encephalitis. 231. Perera S, John D, Senanayaka B. Cost effectiveness of dengue vac-
Virology. 1999;257:363–372. cination following pre-vaccination serological screening in Sri
207. Lai CJ, Monath TP. Chimeric flaviviruses: novel vaccines against Lanka. Int J Technol Assessment Health Care. 2019;35:427–435.
dengue fever, tick-borne encephalitis, and Japanese encephalitis. 232. Shim E. Optimal dengue vaccination strategies of seropositive
Adv Virus Res. 2003;61:469–509. individuals. Math Biosci Eng: MBE. 2019;16:1171–1189.
208. Monath TP, Levenbook I, Soike K, et al. Chimeric yellow fever 233. Yang MH, Freitas ARR. Biological view of vaccination described
virus 17D-Japanese encephalitis virus vaccine: dose-response by mathematical modellings: from rubella to dengue vaccines.
effectiveness and extended safety testing in rhesus monkeys. J Math Biosci Eng: MBE. 2019;16:3195–3214.
Virol. 2000;74:1742–1751. 234. Ashburn PM, Craig CF. Experimental investigations regarding the
209. Hadinegoro SR, Arredondo-Garcia JL, Capeding MR, et al. Effi- etiology of dengue fever. J Infect Dis. 1907;4:440–475.
cacy and long-term safety of a dengue vaccine in regions of 235. Cleland JB, Bradley B, MacDonald W. Further experiments in the
endemic disease. N Engl J Med. 2015;373:1195–1206. etiology of dengue fever. J Hyg. 1919;18:217–254.
210. Guy B, Jackson N. Dengue vaccine: hypotheses to understand 236. Graham H. Dengue: a study of its mode of propagation and
CYD-TDV-induced protection. Nat Rev Microbiol. 2016;14:45–54. pathology. Med Rec New York. 1902;61:204–207.
211. WHO. Global Advisory Committee on Vaccine Safety, 15-16 June 237. Graham H. The dengue: a study of its pathology and mode of
2016. Wkly Epidemiol Rec. 2016;91:341–348. propagation. J Trop Med. 1903;6:209–214.
212. WHO. Meeting of the Strategic Advisory Group of Experts on 238. Sawada T, Sato H, Sai S. On the experimental dengue infection in
Immunization, April 2016, conclusions and recommendations. man. Nippon Igaku. 1943;3325:529–531.
Wkly Epidemiol Rec. 2016;91:265–284. 239. Siler JF, Hall MW, Hitchens AP. Dengue: its history, epidemi-
213. WHO. Informing Vaccination Programs: A Guide to the Design and ology, mechanism of transmission, etiology, clinical manifes-
Conduct of Dengue Serosurveys. Geneva: World Health Organiza- tations, immunity, and prevention. Philippine J Sci. 1926;29:
tion; 2017. 1–304.
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
285.e6 SECTION II Licenced Vaccines and Vaccines in Development
240. Snijders EP, Dinger EJ, Schuffner WAP. On the transmission of 263. Deubel V, Kinney RM, Esposito JJ, et al. Dengue 2 virus envelope
dengue in Sumatra. Am J Trop Med. 1931;11:171–197. protein expressed by a recombinant vaccinia virus fails to protect
241. Yaoi H. A summary of out studies on dengue. Yokohama Med Bull. monkeys against dengue. J Gen Virol. 1988;69:1921–1929.
1958;9:1–20. 264. Men R, Wyatt L, Tokimatsu I, et al. Immunization of rhesus
242. Endy TP, Wang D, Polhemus ME, et al. A Phase 1, open label- monkeys with a recombinant of modified vaccinia virus Ankara
assessment of a dengue virus-1 live virus human challenge – expressing a truncated envelope glycoprotein of dengue type 2
(DENV-1-LVHC) strain. J Infect Dis. 2021;223(2):258–267. virus induced resistance to dengue type 2 virus challenge. Vaccine.
243. Cassetti MC, Thomas SJ. Dengue human infection model: intro- 2000;18:3113–3122.
duction. J Infect Dis. 2014;209(suppl 2):S37–S39. 265. Zhao BT, Prince G, Horswood R, et al. Expression of dengue virus
244. Thomas SJ. Dengue human infection model: re-establishing a structural proteins and nonstructural protein NS1 by a recombi-
tool for understanding dengue immunology and advancing vac- nant vaccinia virus. J Virol. 1987;61:4019–4022.
cine development. Hum Vaccin Immunother. 2013;9:1587–1590. 266. Coller BA, Clements DE, Bett AJ, et al. The development of
245. Dayan GH, Garbes P, Noriega F, et al. Immunogenicity and recombinant subunit envelope-based vaccines to protect against
safety of a recombinant tetravalent dengue vaccine in children dengue virus induced disease. Vaccine. 2011;29:7267–7275.
and adolescents ages 9-16 years in Brazil. Am J Trop Med Hyg. 267. Putnak R, Coller BA, Voss G, et al. Dengue type-2 vaccine formu-
2013;89:1058–1065. lations evaluated in rhesus monkeys are potential candidates for
246. Gailhardou S, Skipetrova A, Dayan GH, et al. Safety overview human use. Paper Presented at: Annual Meeting of the, Washington,
of a recombinant live-attenuated tetravalent dengue vaccine: DC: American Society of Tropical Medicine and Hygiene; 1999
pooled analysis of data from 18 clinical trials. PLoS Negl Trop Dis. 28 November–2 December.
2016;10:e0004821.
268. Govindarajan D, Meschino S, Guan L, et al. Preclinical devel-
247. Russell PK, Halstead SB. Challenges to the design of clinical tri-
opment of a dengue tetravalent recombinant subunit vaccine:
als for live-attenuated tetravalent dengue vaccines. PLoS Negl Trop
immunogenicity and protective efficacy in nonhuman primates.
Dis. 2016;10:e0004854.
Vaccine. 2015;33:4105–4116.
248. Secretariat. Background Paper on Dengue Vaccines, SAGE Working
Group on Dengue Vaccine. Geneva: World Health Organization; 2016. 269. Manoff SB, George SL, Bett AJ, et al. Preclinical and clinical
249. WHO. Dengue vaccine: WHO position paper – July 2016. Wkly development of a dengue recombinant subunit vaccine. Vaccine.
Epidemiol Rec. 2016;91:349–364. 2015;33:7126–7134.
250. de Silva AM, Harris E. Which dengue vaccine approach is the 270. Hermida L, Bernardo L, Martin J, et al. A recombinant fusion pro-
most promising, and should we be concerned about enhanced tein containing the domain III of the dengue-2 envelope protein
disease after vaccination? The path to a dengue vaccine: learning is immunogenic and protective in nonhuman primates. Vaccine.
from human natural dengue infection studies and vaccine trials. 2006;24:3165–3171.
Cold Spring Harbor Perspect Biol. 2018:10. 271. Valdes I, Hermida L, Martin J, et al. Immunological evaluation
251. Henein S, Swanstrom J, Byers AM, et al. Dissecting antibodies in nonhuman primates of formulations based on the chimeric
induced by a chimeric yellow fever-dengue, live-attenuated, tetra- protein P64k-domain III of dengue 2 and two components of
valent dengue vaccine (CYD-TDV) in naive and dengue-exposed Neisseria meningitidis. Vaccine. 2009;27:995–1001.
individuals. J Infect Dis. 2017;215:351–358. 272. Suzarte E, Marcos E, Gil L, et al. Generation and characterization
252. Halstead SB. Identifying protective dengue vaccines: guide to of potential dengue vaccine candidates based on domain III of
mastering an empirical process. Vaccine. 2013;31:4501–4507. the envelope protein and the capsid protein of the four serotypes
253. Weiskopf D, Sette A. T-cell immunity to infection with dengue of dengue virus. Arch Virol. 2014;159(7):1629–1640.
virus in humans. Front Immunol. 2014;5:93. 273. Valdés I, Izquierdo A, Cobas K, et al. A heterologous prime-boost
254. Lopez AL, Adams C, Ylade M, et al. Determining dengue virus strategy for immunization against Dengue virus combining the
serostatus by indirect IgG ELISA compared with focus reduction Tetra DIIIC subunit vaccine candidate with the TV005 live-atten-
neutralisation test in children in Cebu, Philippines: a prospective uated tetravalent vaccine. J Gen Virol. 2019;100:975–984.
population-based study. Lancet Global Health. 2021;9:e44–e51. 274. Grunwald T, Ulbert S. Improvement of DNA vaccination by
255. Brinton MA, Kurane I, Mathew A, et al. Immune mediated adjuvants and sophisticated delivery devices: vaccine-platforms
and inherited defences against flaviviruses. Clin Diagn Virol. for the battle against infectious diseases. Clin Exp Vaccine Res.
1998;10:129–139. 2015;4:1–10.
256. Trent DW, Kinney RM, Huang CY. Recombinant dengue virus 275. Mason PW, Shustov AV, Frolov I. Production and characteriza-
vaccines. In: Gubler DJ, Kuno G, eds. Dengue and Dengue Hemor- tion of vaccines based on flaviviruses defective in replication.
rhagic Fever. New York: CAB International; 1997:379–404. Virology. 2006;351:432–443.
257. Fonseca BAL, Khoshnood K, Shope RE, et al. Flavivirus type-specific 276. Suzuki R, Winkelmann ER, Mason PW. Construction and charac-
antigens produced from fusions of a portion of the E protein terization of a single-cycle chimeric flavivirus vaccine candidate
gene with the Escherichia coli TRPE gene. Am J Trop Med Hyg. that protects mice against lethal challenge with dengue virus
1991;44:500–508. type 2. J Virol. 2009;83:1870–1880.
258. Srivastava AK, Putnak JR, Warren RL, et al. Mice immunized with 277. Becker Y. Dengue fever virus and Japanese encephalitis virus syn-
a dengue type 2 virus E and NS1 fusion protein made in Esch- thetic peptides, with motifs to fit HLA class I haplotypes preva-
erichia coli are protected against lethal dengue virus infection. lent in human populations in endemic regions, can be used for
Vaccine. 1995;13:1251–1258. application to skin Langerhans cells to prime antiviral CD8(+)
259. Bielefeldt Ohmann H, Beasley DW, Fitzpatrick DR, et al. Analysis cytotoxic T cells (CTLs) – a novel approach to the protection of
of a recombinant dengue-2 virus-dengue-3 virus hybrid envelope humans. Virus Genes. 1994;9:33–45.
protein expressed in a secretory baculovirus system. J Gen Virol. 278. Roehrig JT, Johnson AJ, Hunt AR, et al. Enhancement of the anti-
1997;78(Pt 11):2723–2733. body response to flavivirus B-cell epitopes by using homologous
260. Delenda C, Staropoli I, Frenkiel MP, et al. Analysis of C-termi- or heterologous T-cell epitopes. J Virol. 1992;66:3385–3390.
nally truncated dengue 2 and dengue 3 virus envelope glycopro- 279. Huang JH, Wey JJ, Sun YC, et al. Antibody responses to an
teins: processing in insect cells and immunogenic properties in immunodominant nonstructural 1 synthetic peptide in patients
mice. J Gen Virol. 1994;75:1569–1578. with dengue fever and dengue hemorrhagic fever. J Med Virol.
261. Velzing J, Groen J, Drouet MT, et al. Induction of protective immu- 1999;57:1–8.
nity against Dengue virus type 2: comparison of candidate live 280. Garcia G, Vaughn DW, Del Angel RM. Recognition of synthetic
attenuated and recombinant vaccines. Vaccine. 1999;17:1312–1320. oligopeptides from nonstructural proteins NS1 and NS3 of den-
262. Sugrue RJ, Fu J, Howe J, et al. Expression of the dengue virus gue-4 virus by sera from dengue virus-infected children. Am J Trop
structural proteins in Pichia pastoris leads to the generation of Med Hyg. 1997;56:466–470.
virus-like particles. J Gen Virol. 1997;78(Pt 8):1861–1866.
Descargado para Alonso Humberto Bolbaran Castillo ([email protected]) en University of Chile de ClinicalKey.es por Elsevier en mayo 06,
2024. Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.