Summary
Dengue, caused by four closely related viruses (DENV-1–4), is a growing global public health concern, with outbreaks capable of overwhelming healthcare systems and disrupting economies. Climate change, along with other factors, bears substantial relevance to dengue by impacting the virus, mosquitoes, and human populations. Dengue is endemic in more than 100 countries across tropical and subtropical regions worldwide, and the expanding range of the mosquito vector increases risk in new areas. Notably, locally acquired cases are increasingly documented in areas like Spain, Portugal, France and the southern United States, while emerging evidence points to silent epidemics in Africa. Recent years have witnessed significant advancement in our understanding of the virus, immune responses, and disease progression, thanks to ongoing research efforts. Nevertheless, the quest for a reliable immune correlate of protection remains a crucial challenge in the assessment of dengue vaccines. Encouragingly, novel interventions have emerged including partially effective vaccines and innovative mosquito control strategies. These developments mark the beginning of a new era in dengue prevention and control, offering promise in addressing this pressing global health issue.
Keywords: Dengue, epidemiology, classifications, diagnosis, immuno-pathogenesis, clinical manifestation, management, vaccines, anti-viral, new treatment, prevention, control
Introduction
Dengue is a systemic viral infection of increasing global importance. Epidemics of an illness compatible with dengue were first reported in 1779 (1) and the virus was first isolated in 1943 (2). Currently, dengue is endemic in more than 100 countries in tropical and subtropical regions of Southeast Asia, Africa, the West Pacific, and the Americas (3). Dengue is also seen in some regions of Europe, including France, Croatia, Portugal, and Germany, and some parts of the United States (US). Ongoing climate change, population growth, mobility, and urbanization are anticipated to exacerbate dengue burden, primarily by increasing risk in endemic areas, as well as secondarily by expanding range of the primary vector, Aedes aegypti mosquitoes, into new areas. (4) Due to the confluence of these factors, studies predict that the global population at risk will increase from 53% of the world’s population in 2015 to 63% in 2080, with high environmental suitability for dengue in tropical and subtropical areas worldwide (Figure 1). (5)
Figure 1.

Predicted global dengue risk. Means (A) and Standard deviations (SDs) (B) of Force of infection (FOI) estimates in dengue endemic countries across 200 geographically stratified bootstrap samples. Average FOI was estimated from age-stratified seroprevalence or case notification date using a set of environmental explanatory variables. Modified from Cattarino L and colleagues (3) Environmental suitability for dengue occurrence in 2080 (C). Adapted from Messina JP and colleagues (5)
Dengue has an important economic impact, resulting in estimated global healthcare costs of over $8·9 billion (95% uncertainty interval $3·7 billion–$19·7 billion) annually. (6, 7) High costs are associated with loss of productivity, as well as direct medical costs from hospitalization. (8, 9) Dengue outbreaks can overwhelm health care systems, disrupt economies, and reduce public confidence in government responses. Many commonly used vector control strategies, like insecticide spraying, have failed to curb disease incidence but continue to be employed in the absence of robust evidence for their effectiveness or optimal implementation (10). However, increased understanding of dengue epidemiology and immune mediators of symptomatic and severe disease, as well as the availability of effective clinical management, partially effective vaccines, candidate vaccines in the pipeline, and novel approaches to mosquito control, have the potential to inform and significantly improve the effectiveness of dengue control programs. This seminar reviews the latest research on dengue virus (DENV), current gaps in our knowledge, and areas for future study.
Dengue viruses
DENV-1, 2, 3 and 4 are single-stranded ribonucleic acid viruses in the genus Flavivirus, family Flaviviridae. Flavivirus include other viruses transmitted by mosquitoes and ticks such as Zika, West Nile, Japanese encephalitis, and tick-borne encephalitis viruses. The four dengue viruses are called serotypes because each has different interactions with the antibodies in human blood (2). They share approximately two thirds of their genomes (2), with different genotypes existing within each serotype, which can vary in disease severity. DENV is primarily transmitted through the bite of an infected mosquito vector, with Aedes aegypti as the most common vector, although other species including Aedes albopictus may also sustain transmission. Other rare transmission routes include perinatal transmission, blood transfusion, organ transplantation, and two cases of sexual transmission. (11–15) The incubation period from exposure to symptom development is typically four to ten days. (16)
Epidemiology
The global burden of dengue illness has continued to rise over the past decade, with large outbreaks in endemic areas and more cases of dengue in travelers. During 2007–2017, deaths from dengue increased by 65.5% to more than 40,500 (17,600–49,800) annually (17). Expansion of Aedes mosquito vectors and increasing dengue incidence in non-endemic areas is also a growing concern. New detections of local dengue transmission in areas without previous transmission and higher case numbers in areas with sporadic transmission have been documented in southern Europe and the United States, as well as unprecedented outbreaks at high altitudes as seen in Nepal. (18–20) Additionally, more DENV serotypes are co-circulating in endemic areas, producing increased case numbers and greater probability of severe disease from serotype re-introduction or replacement. (21)
DENV seroprevalence estimates vary widely across countries and regions, shaped by differences in underlying DENV transmission intensity as well as differences in methods and assays applied. High IgG antibody seroprevalence (>60%) has been reported in highly endemic areas in Southeast Asia and the Americas, mid-range (10%–60%) seroprevalence in areas with frequent or sporadic transmission such as many countries in Africa and the Middle East, and low seroprevalence in non-endemic areas such as the United States and Europe. (22) Vaccine trials have revealed high variability in DENV incidence across study sites in Latin America and Asia, ranging from 1·5 to 6·6 episodes of symptomatic dengue per 100 person-years among children 2–16 years old. (23) The estimated force of infection (FOI), which is the per capita rate at which susceptible individuals become infected, similarly varies by region, with highest FOI observed in areas near the tropics (Figure 1). However, FOI estimates are limited by surveillance data availability as well as temporal and interannual variability in dengue incidence (3).
Risk of infection is driven by susceptibility to the four DENV serotypes; therefore, DENV incidence in hyperendemic locales is concentrated in children and young adults. (24) The average age of infection in these areas has been increasing in recent decades, for reasons which may include a decrease in the FOI due to changes in the population age structure (25), effective vector control (26), or possibly increased awareness and diagnosis of dengue in adults. In areas hyperendemic for DENV transmission, the risk for enhanced disease has been suggested to be concentrated within two age-related peaks: the first in infants with possible contributions from waning maternal antibodies (27–29), and the second in individuals experiencing a second DENV infection. However, further studies of infants with severe dengue, are needed to unequivocally confirm that maternally derived anti-DENV antibodies are a critical risk factor for severe disease in infants (30). Delayed diagnosis or detection of shock can lead to severe disease and death. Risk of severe disease and death has also been shown to be higher for those with comorbidities such as diabetes or pulmonary, heart, or renal disease. A recent meta-analysis suggests that the relative risks of severe dengue associated with underlying chronic diseases and co-morbidities may be much higher than that of secondary infection alone. (31)
Disparities in dengue risk have been identified, with increased risk occurring in areas with higher population density and poor housing conditions. (32, 33) Increasing population mobility and tourism have also been linked to increased dengue transmission, while imported DENV cases have led to outbreaks in non-endemic areas. (34) DENV transmission occurs commonly within and around households, with a potential increased risk among people in endemic areas who stayed near the home compared to people with greater mobility. (35, 36) However, COVID-19-related disruptions and lockdowns in 2020 were found to result in a decrease in dengue incidence across endemic regions, with the strongest associations related to school closures and reduced time in non-residential areas, although changes in healthcare-seeking behavior could have also contributed to lower reported case numbers. (37)
Dengue is the most frequent arboviral disease encountered among travelers, with an increasing global trend during 1995 to 2020. (38) Among travelers to Southeast Asia during 1995–2020, incidence ranged from 50 to 159 dengue cases per 1000 ill travelers who sought care in the Geosentinel network of travel medicine providers during non-epidemic and epidemic years, respectively. (38) Although severe dengue often occurs during second infections in dengue-endemic regions, primary dengue infections can also be severe and result in fatal outcomes, which has been documented among travelers without previous dengue infection. (39) Additionally, asymptomatic infections have been documented to occur among travelers to dengue endemic areas in a ratio of approximately four to one, creating risks for introduction of dengue viruses or novel serotypes from asymptomatic persons into areas with competent mosquito vectors. (40)
Classification and clinical course
Dengue is a self-limiting acute febrile illness with non-specific manifestations. Among people infected with DENV, approximately 60–80% are asymptomatic or have subclinical infections, with increased risk of disease among those with older age and longer time between subsequent DENV infections. (41, 42) WHO guidelines previously classified symptomatic dengue virus infections as dengue fever, dengue hemorrhagic fever (DHF), and dengue shock syndrome (DSS). However, revised WHO guidelines in 2009 (Panel a) classified symptomatic dengue as dengue without warning signs, dengue with warning signs, and severe dengue (Figure 2). (16) (41, 42)
Figure 2.

Dengue clinical course, classification, laboratory abnormalities and management. From WHO Handbook for Clinical Management of Dengue (43).
Symptomatic dengue generally follows the following clinical course: febrile, critical, and recovery phases (Figure 3). (16, 43) During the febrile phase, which lasts from 2–7 days, there is typically an acute onset of high-grade fever (≥38·5°C) that may be accompanied by nausea, vomiting, a transient macular rash, aches, pains, and other constitutional symptoms. (44) The mucocutaneous manifestations of dengue are varied and may include transient facial erythema, petechial rash, conjunctival and scleral injection, and a maculopapular or morbilliform eruption three to six days after the onset of fever that may coalesce but with areas of sparing. (45) The tourniquet test (46) can be positive and minor bleeding, such as skin petechiae or bruises may occur. (47) Most commonly, fever resolves followed by recovery phase; if this is the case, this illness would be categorised as uncomplicated dengue.
Figure 3.

Dengue laboratory findings, virus detection and immune response
Data from WHO Dengue Guidelines for Diagnosis, Treatment, Prevention and Control (16), Hunsperger E. and colleagues (210) , Chaloemwong and colleagues (211) and Dussart and colleagues (212).
Some dengue patients experience the critical phase, which generally occurs around days 4–6 of illness and often coincides with defervescence. (16, 48) The hallmark of severe dengue is plasma leakage, when the blood’s protein-rich fluid component flows from blood vessels into surrounding tissue, which can lead to shock and is sometimes associated with haemorrhage. (49, 50) There is some evidence that less severe capillary leakage may be more common in clinically diagnosed uncomplicated dengue than previously recognized. (51) Plasma leakage becomes clinically apparent near the time of defervescence and spontaneously improves after about 48 to 72 hours (52). Dengue warning signs of possible clinical deterioration can precede the critical phase. Warning signs are intended to be used to detect disease progression and include abdominal pain or tenderness, persistent vomiting, clinically detectable extra-vasal fluid accumulation, mucosal bleed, lethargy or restlessness, liver enlargement, and an increase in haematocrit usually concurrent with rapid decrease in platelet count (Figure 3). (16, 53) If the patient improves and recovers, the illness is classified as dengue with warning signs. However, the disease may continue to advance towards severe dengue, which occurs in approximately 2%–5% of dengue patients (53, 54). Rates of progression to severe disease are highly variable by age, underlying comorbidities, clinical resources, expertise in managing dengue, and possibly infecting DENV serotype and genotype. (53)
The median case fatality rate for patients with dengue is 5% (range 0.01%–39%). (55) The criteria for severe dengue include: (1) severe plasma leakage leading to shock or to fluid accumulation with respiratory distress, (2) severe bleeding as evaluated by clinician, and (3) severe organ involvement including the central nervous system, heart, or liver (indicated by an aspartate aminotransferase [AST] or alanine aminotransferase [ALT] level of 1000 IU/l or greater). (16) Shock is signalled by rising haemoconcentration followed by an increase in diastolic pressure with narrowing pulse pressure, rapid pulse, restlessness, hypotension, signs of poor peripheral perfusion (cold extremities and slow capillary refill time) and decreased urinary output. Repeat shock episodes may occur during the critical phase (56). Circulatory compromise is generally worse in the extremes of age, likely due to increased vascular permeability and lower capacity to maintain cardiovascular homeostasis in young children, while factors in the elderly include comorbidities and vascular aging. (57) Epistaxis, gum bleeding, hypermenorrhea, hemoglobinuria, and other haemorrhagic manifestations are most seen during the critical phase. (47) The risk for severe bleeding (such as from the gastrointestinal or vaginal tract) increases in profound or prolonged shock, in association with coagulation abnormalities combined with tissue hypoxia and acidosis. (58)
Involvement of other organ systems can also occur during the various phases of dengue. Hepatitis and elevated liver enzymes are common among patients with symptomatic dengue, while acute liver failure, encephalitis, myocarditis, and acute kidney injury are infrequent. (59–62) Sight-damaging ophthalmic inflammation during dengue has also been described. (63) Dengue during pregnancy has implications for the mother and the fetus. A greater risk of DHF/DSS has been reported among pregnant women compared to non-pregnant women, (64) as well as an increased risk of maternal death. (65, 66) In addition to increased maternal morbidity and mortality, dengue also poses a risk to the fetus, with an increased risk of miscarriage (67), stillbirths, and neonatal deaths. (66)
During the recovery phase, extravasated fluids are resorbed and well-being improves. The patient may develop an erythematous, sometimes pruritic, “Herman’s rash” with white islands of normal skin. (68) In adults, post-viral fatigue and depression of several weeks to months have been described. (69)
Most dengue patients recover uneventfully, but it is crucial to promptly recognise those who will require medical intervention. A systematic review confirmed that warning signs were associated with progression to severe dengue but identified potential additional markers, including low serum albumin and elevated AST and ALT concentrations. (31) Thrombocytopenia is also commonly seen in dengue patients, and lower platelet counts have been associated with progression to severe disease. (31) Ultrasound can be used to detect plasma leakage in dengue. (70) Ascites, pleural effusion and gallbladder wall thickening are the most common findings, but standard protocols for sonographic procedures, as well as improved information about the positive predictive value of early and low-volume plasma leakage for development of severe dengue, are needed. (71, 72) Investigations are ongoing to determine whether inflammatory and vascular markers in the febrile phase of dengue may be useful to predict severe outcomes. (73, 74)
Dengue management
Currently there is no effective prophylactic or therapeutic agent against dengue. (75) Chloroquine, balapiravir, celgosivir, lovastatin, corticosteroids, ivermectin, plasma infusion, recombinant activated factor VII, anti-D globulin, immunoglobulin, and interleukin 11 have not been shown to be beneficial. (76–78) However, clinical trials are limited by small sample sizes, heterogeneous populations, and difficulties in assessing outcomes, and additional work is needed to thoroughly assess potential benefits. A randomized controlled trial of montelukast, a leukotriene receptor antagonist used to reduce asthma exacerbation, is ongoing among adult patients with dengue to evaluate its efficacy in preventing dengue with warning signs (ClinicalTrials.gov Identifier: NCT04673422). The small molecule, JNJ-A07, which blocks the intracellular replication of DENV, has shown promise in pre-clinical trials. (79) There are also ongoing pre-clinical trials of therapeutic monoclonal antibodies against dengue. (80)
In the absence of specific therapy, the management of dengue remains supportive. The 2012 WHO handbook, (43) since adapted to other guidelines, (81, 82) focuses on a stepwise approach of assessment and treatment according to Groups A, B, and C (Figure 2). Group A patients (no warning signs, comorbidities, or difficult social circumstances) are sent home with daily in-person monitoring. Groups B (comorbidities or warning signs) and C (severe dengue) require hospital management and intravenous fluids (Figure 2). (83) Fluid replacement is lifesaving in severe dengue but must be administered cautiously and discontinued when plasma leakage subsides to avoid iatrogenic fluid overload. (84) Among patients with thrombocytopenia, prophylactic platelet transfusion does not prevent bleeding and may contribute to fluid overload. (85, 86) Although these guidelines are based mainly on expert opinion and small randomized controlled trials, (87–90) case fatality rates have been considerably reduced with judicious fluid replacement. Some areas of uncertainty remain, such as in the choice of colloid solution and blood products, the use of fluid boluses, and the optimal treatment of recurrent shock episodes. Steroid use is not recommended as it has not shown clinical benefit. (57, 76) Training in clinical management, including early recognition of plasma leakage, remains an essential strategy to reduce morbidity and mortality.
Dengue diagnosis
Dengue differential diagnosis is broad. During the febrile phase, it includes other viral infections (measles, rubella, enterovirus, adenovirus, influenza and other arboviruses), as well as bacterial (leptospirosis and typhoid fever) and parasitic (malaria) illnesses that may be present in dengue-endemic areas. (91). Diagnosis of dengue infection during the acute phase can be achieved using whole blood, plasma, or serum collected up to seven days after symptom onset by detection of viral RNA through nucleic acid amplification tests (NAAT) (92), detection of viral antigens such as dengue non-structural protein 1 (NS1), by enzyme-linked immunosorbent assay (ELISA), or rapid diagnostic tests, and IgM antibodies from day 4 to approximately 12 weeks post onset through serologic testing (93) (Fig 3). NAAT assays are the preferred method of dengue diagnosis; (92) in addition to their diagnostic specificity, molecular methods can be used to identify the virus serotype.
NS1 can be detected in other body fluids such as urine, saliva, and cerebrospinal fluid. (94) NS1 tests can be as sensitive as molecular tests during the first seven days of symptoms in primary infections, although sensitivity is lower in secondary infections; after seven days, although sensitivity is lower, NS1 has been detected in serum up to 12 days after symptom onset (94, 95). Dengue IgM antibodies are detectable for a longer period, from day four to approximately 12 weeks post symptom onset (96). Dengue IgG is detectable around day seven in the first infection (primary infection); the antibody concentration increases slowly thereafter and is thought to persist for life. In patients with a previous dengue infection (secondary infection), anti-dengue IgG titres rise rapidly within the first week of illness (Figure 3). Although serologic assays provide less certainty than NAAT or NS1 due to cross-reactivity with other flaviviruses and longer antibody duration, a positive anti-DENV IgM suggests recent infection. Additionally, seroconversion or a four-fold rise in titres on anti-DENV IgM or IgG assays in paired samples is strongly suggestive of recent infection. (96) Many rapid tests are available in the market and are an important tool for the early diagnosis of dengue. Meta-analyses suggest that immunochromatographic tests that combine IgM, IgG, and NS1 detection have the best performance compared to tests detecting individual analytes, with pooled sensitivity of 90%–91% and specificities of 89%–96% (97, 98). Unfortunately, these tests are not widely available in dengue endemic areas. In patients from areas with ongoing transmission of another flavivirus, plaque reduction neutralization tests (PRNT) can help distinguish DENV from other flaviviruses. PRNTs, however, are rarely available in clinical laboratories and typically do not provide results within a meaningful timeframe for clinical management. PRNTs may be valuable in circumstances such as pregnancy where differentiating between Zika and dengue may have important clinical implications. (93) NS1 antibody ELISA tests for Zika virus (ZIKV) have high specificity due to the substantial amino acid differences between DENV and ZIKV NS1 and can be useful for differential diagnosis (94).
Dengue immunology
DENV infection is initiated in the skin when an infected mosquito takes a blood meal, injecting virus along with salivary proteins that increase recruitment of susceptible immune cells to the site of infection (99). Myeloid cells are a key target of DENV infection, including monocytes, macrophages, and dendritic cells. A first DENV infection results in an early innate response characterized by stimulation of interferon gamma (IFNγ). In contrast, in subsequent DENV infections, binding but not neutralizing antibodies induced by prior exposure to DENV (or a related flavivirus) facilitate infection of myeloid cells via the fragment crystallizable gamma receptor (FcγR), producing a larger population of viruses and further exacerbating disease severity in a process called extrinsic antibody-dependent enhancement (ADE) (100–104). Entry via the FcγR also mediates intrinsic ADE, which suppresses IFNγ stimulation and innate immunity. This results in a shift instead towards a T helper 2 (Th2) response dominated by secretion of Interleukin-10, minimizing induction of other pro-inflammatory cytokines and hindering the early cellular and humoral immune response (Figure 4) (100). ADE is thought to increase replication of the virus at this key early stage and elevate the risk of progressing to severe dengue and DHF/DSS. (105, 106) However, while pre-infection binding antibody levels are associated with increased viremia and risk of DHF/DSS, the causal link from ADE to higher viremia to DHF/DSS has not been demonstrated (103).
Figure 4.

Correlates of dengue pathogenesis and protection. Types of immunological responses associated with increased dengue disease (pathogenic, generally during a secondary infection: ADE of myeloid cells (100–102), strong plasmablast response (107, 109, 110), and weak T cell response (114)) or reduced disease (protective, including broadly neutralizing antibodies (135, 136) and effective, cytotoxic CD8+ and CD4+ T cells (115–118)).
Mediators of severe disease
CD14+CD16+ monocytes increase in early DENV infection (107, 108) and trigger a strong plasmablast response characterized by secretion of high levels of anti-DENV antibodies (107, 108). The role that excess antibody production plays in acute dengue is not clear, but may contribute to disease pathogenesis by furthering ADE, increasing autoantibodies, and potentially changing glycosylation of antibodies, which has been shown to be strongly associated with DHF/DSS (109, 110). DENV can also directly infect B cells, and while B cell infection does not contribute to viremia it does drive proliferation of B cells and stimulation of cytokines (111). Higher viral load may also mediate severe disease by increasing secretion of NS1. In in vitro and animal models, NS1 levels directly and indirectly triggered vascular leakage, disrupting the glycocalyx and tight junctures between endothelial cells lining blood vessels and further facilitating dissemination of virus into tissue as well as plasma leakage (112). However, the association between NS1 levels and severe disease has not been clearly demonstrated in clinical studies. (49) Severe dengue is also associated with liver and spleen pathology, with autopsies revealing DENV tropism for liver macrophages (Kupffer cells) and splenic macrophages, which secrete high levels of cytokines and mediate damage due to deposition of complement, resulting in necrosis in liver and splenic endothelial cells (113).
T cell responses
The role of DENV-specific T cells has been debated, as they have been implicated in both pathogenic and protective immunity (Figure 4). On the side of a pathogenic role, CD8+ T cells induced by prior DENV infection have been shown to have low affinity for the new infecting serotype, proliferating but with limited cytotoxic function, resulting in delayed viral clearance and stimulation of pro-inflammatory cytokines that mediate leakage and disease (114). However, other studies suggest a protective role, with higher magnitude and more multifunctional, cytokine-producing DENV-specific CD8+ T cell responses and specific HLA alleles associated with reduced viremia, and lower probability of progression to symptomatic disease and severe dengue (115–117).
DENV-specific CD4+ cells both promote CD8+ T cells and stimulate B cells. Individuals with multiple prior DENV exposures have populations of clonally expanded cytotoxic CD4+ cells that may be protective (118). In contrast, while T follicular helper (Tfh) cells are critical to facilitating the maturation of the B cell response, Tfh expansion in acute dengue has been associated with secondary and severe dengue as well as a strong plasmablast response, suggesting a possible role in immunopathology (119, 120).
DENV serotypes and post-secondary DENV antibodies
Although all DENV serotypes can result in symptomatic or severe disease, differences have been identified in risk by serotype and infection number (primary or secondary). DENV-2 and DENV-4-associated dengue illnesses are more frequently identified as secondary DENV infections, whereas DENV-1 and DENV-3 can cause significant primary disease (121–123) DENV-4 has been associated with a reduced risk of disease compared to the other serotypes, which is supported by observations of silent DENV-4 epidemics in Thailand. (124) All sequences of infecting serotypes can cause severe disease and there is not a defined order in the sequence of DENV infections for worse outcomes, although different patterns have been described (125, 126). Antigenic differences between serotypes may be important in explaining the magnitude of dengue epidemics (127–129), and viral sequence, genotype, and changes in non-structural proteins may also help determine epidemic severity (130, 131).
Sequential infection with distinct DENV serotypes induces antibodies capable of neutralizing DENV-1–4 without triggering ADE, likely by activating cross-reactive memory B cells to undergo further affinity maturation and target quaternary epitopes conserved across serotypes, thus providing broad protection. High levels of cross-reactive binding antibodies and multiple prior DENV infections are associated with reduced risk of symptomatic disease (105, 124, 132). Consistent with this observation, Dengvaxia and the Qdenga dengue vaccines have high efficacy against symptomatic and severe disease in DENV-immune individuals but not naive individuals, further suggesting that sequential exposure to distinct DENV strains is important to inducing broad protection (133, 134). A few post-secondary broadly neutralizing antibodies have been identified such as envelope-dimer-dependent epitope (EDE) antibodies. These antibodies target conserved quaternary epitopes and neutralize by disrupting the conformational changes required for viral entry (135, 136). EDE-like broadly neutralizing antibodies may be good correlates of protection for the evaluation of new dengue vaccines.
Dengue Vaccines
The need for a tetravalent formulation that induces simultaneous and balanced protection against all 4 serotypes has slowed the development of a dengue vaccine. Among people with a DENV infection before vaccination, even a vaccine dominated by one vaccine serotype is likely to induce cross protective immunity by activating memory. However, in DENV naïve individuals with no immune memory, the protective immune response will strongly depend on the immunogenicity of each serotype-specific vaccine component (137).
There are three leading dengue vaccines. Dengvaxia (CYD-TDV), developed by Sanofi-Pasteur, was the first licensed dengue vaccine (138). Qdenga (TAK-003), developed by Takeda, was approved by the European Commission in December 2020 and is licensed in several countries including Indonesia, Brazil, Argentina, the United Kingdom, and Germany (139–141). The third, TV003, was developed by the National Institutes of Health (NIH) and is in late-stage trials (142). All three are live vaccines and contain 4 different attenuated vaccine viruses (tetravalent) targeting each of the DENV serotypes. However, they differ in the number of doses required (1 to 3) and time to complete the series (up to 1 year), which could affect feasibility and preferences for use in different settings. Additionally, the need for a dengue test to determine eligibility (pre-vaccination screening) poses a logistical barrier for vaccines only recommended for use among people with a previous DENV infection. There are several other dengue vaccine candidates undergoing clinical trials or pre-clinical evaluation, including other live-attenuated vaccines, inactivated vaccines, recombinant vaccines, and DNA vaccines (143). The successful mRNA vaccine technology used for SARS-CoV-2 is also being evaluated for dengue and could provide dengue vaccine candidates in the future. (144)
Dengvaxia (CYD-TDV)
Dengvaxia uses a three-dose schedule, with doses administered six months apart. It was first recommended by WHO in 2016 for persons nine years of age and older living in highly endemic areas (145). Long term follow-up data over five years from the phase three trials and further analyses of the efficacy results (146) demonstrated that children with evidence of previous DENV infection (seropositive) were protected from severe dengue if they were vaccinated with Dengvaxia. However, risk of hospitalization for dengue and severe dengue was increased among children two to 16 years of age without previous DENV infection (seronegative) who were vaccinated with Dengvaxia and had a subsequent infection in the years after vaccination (hazard ratio for hospitalization, 1·75, 95% Confidence Intervals [CI]: 1·14–2·70, severe dengue 2·87, 95% CI: 1·09, 7·61). After these findings, WHO revised the recommendations for the vaccine to only be given to children with laboratory-confirmed evidence of a past DENV infection (147).
For children aged nine to 16 years with evidence of previous DENV infection, Dengvaxia had an efficacy of about 80% against the outcomes of symptomatic virologically confirmed dengue (VCD), hospitalization for dengue, and severe dengue. (146, 148) Among seropositive children the efficacy by serotype varied (149), with highest protection against DENV-4 (89%), followed by DENV-3 (80%), and lowest against DENV-1 (67%) and DENV-2 (67%) (Table 1). (146)
Table 1.
Comparison of vaccine efficacy for the target use population for live attenuated tetravalent dengue vaccines licensed or in phase three trials
| Vaccine | Dengvaxia/CYD-TDV146 | Qdenga/TAK-003152 | TV003161 |
|---|---|---|---|
| Manufacturer | Sanofi Pasteur | Takeda | NIH/Butantan/Merck |
| Status | Recommended by World Health Organization (WHO) with two options: 1) seropositive persons ages 9–45y; or 2) all persons regardless of serostatus in high seroprevalence areas (>80% seropositivity). Licensed in 20 countries. In U.S. recommended for seropositive children ages 9–16y living in endemic areas.138 | Recommended by WHO to be considered for introduction among children ages 6–16 years in settings with high transmission intensity. Licensed in several countries including Indonesia, Brazil, Argentina, United Kingdom and Germany. | Ongoing phase three trial |
| Platform | 4 chimeric viruses for each DENV serotype on a yellow fever virus (YFV) backbone | Attenuated DENV-2 and 3 chimeric viruses each one of the four DENV serotypes | Attenuated DENV-1, DENV-3 and DENV-4 and a chimeric virus for DENV-2 on a DENV-4 backbone |
| Ages of trial participants | 9–16y | 6–16y | 2–59y |
| Doses | 3 doses 6 months apart | 2 doses 3 months apart | 1 dose |
| Pre-vaccination antibody screening recommended | Yes | No | Unknown |
| Timeframe for efficacy endpoint | 25 months for virologically confirmed dengue (VCD and 60 months for hospitalization) | 54 months for VCD and hospitalization | 24 months for VCD |
| Efficacy among seropositive persons | Efficacy and 95% Confidence Interval (CI) | Efficacy and 95% CI | Efficacy, 95% CI |
| Virologically Confirmed Dengue | |||
| Overall | 76 (64, 84) | 64 (58, 69) | 89 (78, 96) |
| By serotype | |||
| DENV-1 | 67 (46, 80) | 56 (45, 65) | 97 (81, 100) |
| DENV-2 | 67 (47, 80) | 80 (73, 86) | 84 (63, 94) |
| DENV-3 | 80 (67, 88) | 52 (37, 64) | NR |
| DENV-4 | 89 (80, 94) | 71 (40, 86) | NR |
| Hospitalization | |||
| Overall | 79 (46, 80) | 86 (79, 91) | NR |
| By serotype | |||
| DENV-1 | 78 (55, 90) | 67 (37, 82) | NR |
| DENV-2 | 82 (66, 90) | 96 (90, 98) | NR |
| DENV-3 | 63 (18, 83) | 74 (39, 89) | NR |
| DENV-4 | 93 (62, 99) | 100 (NE, NE) | NR |
| Efficacy among seronegative persons | |||
| Virologically confirmed disease | |||
| Overall | 39 (−1, 63) | 54 (42, 63) | 74 (58, 84) |
| By serotype | |||
| DENV-1 | 41 (−7, 67) | 45 (26, 60) | 86 (69, 94) |
| DENV-2 | −21 (−136, 38) | 88 (79, 93) | 58 (21, 78) |
| DENV-3 | 52 (−6, 78) | −16 (−108, 36) | NR |
| DENV-4 | 65 (24, 84) | −106 (−629, 42) | NR |
| Hospitalization | |||
| Overall | −41 (−168, 93) | 79 (64, 88) | NR |
| By serotype | |||
| DENV-1 | −37 (−219, 41) | 78 (44, 92) | NR |
| DENV-2 | −141 (−795, 35) | 100 (NE, NE) | NR |
| DENV-3 | 15 (−225, 78) | −88 (−573, 48) | NR |
| DENV-4 | 7 (−712, 89) | 100 (NE, NE) | NR |
Note: Dengvaxia trial included participants ages 2–16 years of age but only licensed for seropositive individuals ≥9 years. Estimates of efficacy against hospitalization by serotype in seronegative participants ages 2–8 years are: DENV-1 –42 (34, −205), DENV-2 –436 (−58, −1723), DENV-3 –141 (9, −540), DENV-4 –16 (70, −344). NE=Not possible to estimate due to zero cell in one of the groups, NR= not reported. Y=years
The requirement for a laboratory test prior to vaccine administration creates a unique challenge for Dengvaxia implementation. Qualifying laboratory tests include a positive NAAT or NS1 test performed during an episode of acute dengue, or a positive result on pre-vaccination screening tests for serologic evidence of previous infection (presence of IgG antibodies) that meet specific performance characteristics. To reduce the risk of vaccinating someone without previous DENV infection, high specificity in a pre-vaccination screening test is a priority. International working groups and the CDC recommend using tests with a minimum sensitivity of 75% to 85% and minimum specificity of 95% to 98% (138, 150). Few commercially available tests currently meet these requirements. (151)
Qdenga (TAK-003)
Qdenga, developed by Takeda, consists of two doses given three months apart. Among children aged four to 16 years, efficacy against VCD was 64% among seropositive and 54% among seronegative children at three years after vaccination. Efficacy against hospitalization for dengue was higher, at 86% among seropositive and 79% among seronegative children. (152) Differences in efficacy were observed by serotype. Among seronegatives, there was no efficacy against DENV-3 and DENV-4 (Table 1). Notably, estimates indicated a potential increased risk for hospitalization after infection with DENV-3, although numbers were small (three cases in the placebo group and eleven cases in the vaccine group), and mainly observed at one site (134). In December 2022, the European Commission approved the use of Qdenga regardless of serostatus following a positive opinion from the European Medicines Agency (139). The next step for its use in Europe is official recommendations from each European Union country (153). Qdenga has been approved in several countries and Germany has started vaccination among travelers (154). In September 2023, it received a recommendation from the Strategic Advisory Group of Experts (SAGE) on immunization that recommended that Qdenga be considered for introduction in settings with high transmission intensity to maximize the public health impact and minimize any potential risk in seronegative persons. SAGE recommended that the vaccine be introduced to children aged 6 to 16 years of age, and that post-authorization studies should be conducted to further study vaccine effectiveness and safety against serotypes 3 and 4. (155)
The company also plans to submit filings to other regulatory agencies (156).
TV003
TV003 was developed by NIH and was formulated by selecting serotype-specific components to provide a balanced safety and immunogenicity profile based on an evaluation of multiple monovalent and tetravalent candidates (Table 1). (157, 158) TV003 consists of a single dose and has been licensed to several manufacturers globally, including Merck & Co in the U.S. and the Instituto Butantan in Brazil. Phase three trials in Brazil are underway (159, 160). Preliminary results from two-year follow-up of the phase three trial were recently released. Through 2 years of follow-up, the efficacy against VCD was 89% among seropositives and 74% among seronegatives. Results by serotype were available for DENV-1 and DENV-2, with higher efficacy among seropositive participants compared to seronegative (DENV-1 97% and 86%, DENV-2 84% and 58%, respectively) (161). Efficacy for other serotypes is not currently available but is expected as part of the ongoing phase three trial. (162)
Vector control
People who live in or travel to dengue endemic areas can prevent mosquito bites by using EPA-approved insect repellents and wearing clothing that covers arms and legs. The use of screened windows and doors and air conditioning have been shown to be protective (163–165).Bednets can reduce mosquito populations and may have an impact on dengue transmission. (166) Chemical control of Aedes species mosquitoes is limited by widespread insecticide resistance in endemic areas. (167) Novel vector control methods have been developed, including the use of genetically modified mosquitoes (GMM) and Wolbachia-based methods (168). GMM carry a gene that is passed to their offspring and kills females in the larval stage. Male offspring, however, survive and pass this gene to future generations. As a result, mosquito populations decrease over time. (168, 169)
Strategies utilizing Wolbachia, an intracellular bacterium found in about 60% of all insects (170) but not commonly found in wild Aedes mosquitos, (171) have been used for vector control. Wolbachia-mediated suppression refers to a reduction in wild populations of Aedes mosquitoes and is achieved by releasing Wolbachia-infected males into the environment to mate with uninfected wild females, as the resulting eggs do not hatch. (172) In Wolbachia replacement, both Wolbachia-infected males and female mosquitoes are released, which pass the bacteria to their offspring and gradually replace the wild population. (173, 174) In mosquitoes, Wolbachia infection reduces transmission of arboviruses, including dengue, chikungunya, and Zika viruses, when infected female mosquitoes take a bloodmeal. This method has demonstrated reductions of nearly 80% in dengue cases and related hospitalizations in areas where it has been implemented (175) and is currently being deployed in Brazil and Indonesia (176).
Dengue controversies, gaps, and opportunities (panel b)
Many of the key questions in dengue research revolve around the role and behavior of antibodies in protective immunity and ADE, as well as how these change at different time points after infection. It was previously thought that a first DENV infection induced antibodies that wane over two years to titers that can subsequently enhance severe dengue disease, and that secondary DENV infection with a different serotype induced stable, cross-serotype protective antibodies. (41, 42, 177) However, instead of waning over two years, cross-reactive binding antibodies associated with protection or enhancement are stable by ~8 months after primary infection and are maintained at that ‘set point’ for many years after (124, 178). In contrast, anti-DENV antibodies induced after secondary DENV are less stable, wane rapidly for 8 months and then gradually decay over longer periods. One study showed vaccine efficacy waned much faster than geometric mean antibody titers to DENV-1–4, suggesting a component of immunity other than waning antibodies may explain loss of protection (179).
While serotype-specific immunity (i.e., ‘homotypic’ immunity) has classically been thought to impart lifelong protection, there is some evidence that reinfections with a given DENV serotype may occur (180). This bears further evaluation; if proven, the absence of lifelong homotypic immunity would have important implications for dengue vaccines and our understanding of DENV epidemiology.
Additionally, factors shaping the variability observed in severity of epidemics remain poorly understood. Investigators in Taiwan have reported increasing severity throughout the time course of a given epidemic, associated with increased viral diversity, which they hypothesize to be driven by cross-protective immunity (181, 182). It remains to be seen whether these same phenomena are replicated in other regions, with different levels of population immunity and transmission patterns.
Identification of a satisfactory immune correlate of protection — a biomarker measuring immune response to vaccination that is associated with vaccine efficacy — remains an important challenge for DENV epidemiologic studies and assessments of vaccine immunogenicity. This is mainly due to the dominance of immunity to cross-reactive epitopes which do not provide effective protection. In the Dengvaxia pediatric vaccine trial, the discordance between vaccine efficacy and neutralization response rates indicated that PRNT neutralization response is not a completely valid correlate of protection. However, higher PRNT titers after three doses of Dengvaxia were associated with a lower rate of VCD and hospitalization overall and for each infecting serotype (183). Following vaccination, geometric neutralizing antibodies by PRNT to DENV-1–4 of ≥1:100 were associated with ~50% protection against symptomatic dengue, while titers of ≥1:500 were associated with 80% vaccine efficacy; high titers were also associated with protection against hospitalized dengue (183, 184). In the Qdenga vaccine trials, neutralizing antibody titers were lower in participants who experienced VCD (i.e., cases) compared to control, largely driven by data from seropositive participants (134). Techniques such as antibody depletion of cross-reactive antibodies provide improved information about serotype-specific immunity and have shown associations with serotype-specific vaccine efficacy (134, 183–185). Depletion assays have shown that Dengvaxia is dominated by DENV-4 type-specific antibodies and the Takeda dengue vaccine by DENV-2 type-specific antibodies, with mostly cross-reactive antibodies against other serotypes (186, 187). Many of these serotype-specific antibodies bind quaternary epitopes (i.e., two adjacent E proteins simultaneously). The role of neutralizing antibodies binding quaternary epitopes is an ongoing area of research into correlates of protection.
The role of immunological ‘boosting’, defined here as qualitative or quantitative changes in immunity associated with re-exposure to DENV in an individual already exposed to that serotype, remains poorly understood. The effects of boosting are proposed to be evident in dynamic antibody patterns among individuals residing in hyperendemic locales over time. (188) If boosting were a significant contributor to maintenance of DENV immunity and durability of protection, interventions that decrease the force of infection (such as incompletely-protective vaccination programs or partially-effective mosquito control programs) could yield paradoxical effects by lowering levels of boosting and increasing the susceptible population. However, partially effective vaccines and mosquito control interventions complement each other, contributing effectiveness when the other is lacking. Mathematical modeling suggests that combining interventions may yield consistent high effectiveness. (189) Designing and implementing a dengue control program that uses a combination of available interventions is a public health priority.
The immunological and clinical interactions between DENV and non-DENV flaviviruses remain poorly understood, but existing data suggest that the relationship may be highly context-dependent and not bidirectional. ZIKV infection has been suggested to predispose to DHF with a subsequent infection with some, but not necessarily all, serotypes (104). In reverse, higher levels of DENV immunity are associated with protection against Zika in adults and children (190), while lower levels may increase risk of Zika microcephaly. (191, 192)Japanese encephalitis virus (JEV) immunity has been variably associated with risk of dengue illness (193) but also with possible protection (194). Given significant immunological cross-reactivity observed between DENV and non-DENV flaviviruses, it is plausible that these interactions may have consequences for the clinical outcomes of DENV infection as well as the immunological outcomes of DENV vaccination. Further research on these interactions is needed, across a range of flavivirus-endemic regions of the world, to inform DENV vaccine development and evaluation efforts, as well as diagnostics.
Dengue surveillance remains a key challenge in assessing global dengue burden and temporal and geospatial trends. The large proportion of asymptomatic and subclinical cases contribute substantially to transmission but complicate detection, and the non-specific presentation of acute febrile illness can easily be mistaken for other etiologies. This is particularly challenging in malaria-endemic regions where cases have similar presentations, in areas with limited diagnostic test availability, and regions with infrequent or sporadic dengue transmission. National dengue surveillance systems also vary widely in surveillance and laboratory capacity, as well as case definitions used; future efforts should work toward strengthening country-level surveillance and laboratory capacity, harmonizing case definitions, and encouraging public data sharing to better inform dengue preparedness and response efforts.
Search strategy and selection criteria
We searched Pubmed for the years September 30, 2012, to October 10, 2022. We used the search terms “dengue” or “dengue virus”. We also included references cited in these publications and relevant older references from the authors’ personal files. We consulted international dengue control, prevention and treatment guidelines and WHO policy documents.
Panel a: The 1997 versus the 2009 WHO dengue classification and case definitions.
The 1997 WHO classification and case definitions of dengue, dengue haemorrhagic fever (DHF) and dengue shock syndrome (DSS) (195) originated from a clinical study in the 1960s of 123 children in Thailand. (196) A case definition of DHF is met when all four criteria of fever, haemorrhagic manifestations, thrombocytopenia and evidence of plasma leakage are present. DHF has four increasing grades of severity towards DSS (Grades III and IV). DHF definition is required to classify cases as dengue shock syndrome (DHF plus circulatory failure). The 1997 WHO classifications offer distinct advantages for research studies, as the clinical phenomena described relate to underlying mechanisms of immuno-pathogenesis (i.e., ADE), and may guide treatment pathways (e.g., timing and degree of fluid replacement with plasma leakage). Disadvantages of the 1997 criteria include failure to capture severe disease beyond DHF (e.g., cardiac or hepatic end-organ damage) and limited capacity to facilitate triage of DENV-infected patients. The WHO regional office for South-East Asia proposed the term Expanded Dengue Syndrome to describe cases with atypical yet serious manifestations. (197)
The 2009 WHO classification system evolved to facilitate triage and management of dengue patients (53), as well as to capture a broader spectrum of dengue-related disease. Individuals are classified as having dengue, dengue with warning signs and severe dengue. (16)
The 2009 WHO criteria have been widely implemented to guide dengue clinical management decisions, but they have also been criticized due to the broad recommendations for hospitalization of patients with dengue warning signs. Multiple studies indicate the general approach of hospitalizing for dengue with warning signs, as per the 2009 WHO criteria, may increase the identification of individuals who will progress to severe disease (i.e., improved sensitivity), at the expense of increasing hospitalization of individuals who will not progress to severe disease (198, 199). The positive predictive value of individual warning signs for severe dengue has been reported to range from 12–58%, with some warning signs (e.g., clinical fluid accumulation) performing better than others (200). The availability and increasing use of dengue rapid diagnostic tests may facilitate the use of the 2009 simpler dengue classification scheme. However, rapid diagnostic tests are not available in most endemic countries. More precise definitions of the 2009 warning signs and severe dengue are needed, as well as improved risk prediction tools for clinical and research use (201–204).
Currently, the 2009 classification of dengue without warning signs, dengue with warning signs and severe dengue (16) has been adapted in several countries and international guidelines, (82, 83, 205, 206) while the 1997 dengue, DHF, DSS classification (195) continues to be used in others. (207–209)
Panel b: Priorities for future dengue virus (DENV) research.
DENV prevention and control:
Effectiveness of combined vaccine/vector control programs in decreasing or eliminating DENV infection and illness.
Long term data on effectiveness of interventions including vaccines and Wolbachia-based vector control.
Immune correlates of protection:
Immune signatures durably associated with immunopathogenesis and immunoprotection for DENV.
Clinical and immunological interactions between DENV and non-DENV flaviviruses.
Durability of homotypic protection – is it lifelong?
Importance of immune boosting in maintaining protective immunity for DENV.
Gaps in vaccine efficacy for specific sub-groups that could lead to enhancement of severe disease.
DENV epidemiology:
Homotypic DENV reinfections – confirmation, frequency, and risk factors.
Increases in DENV severity within the course of an epidemic –confirmation and features of settings at risk.
Harmonization of surveillance and laboratory methods across regions experiencing DENV transmission; increased data sharing, establish a coordinated genomic surveillance strategy.
Management and diagnosis
Improved point of care diagnostics.
Clinical evaluation of the impact of antivirals.
Improved triage and risk assessment tools (biomarkers, ultrasound, other).
Online Summary.
Dengue Overview
DENV-1, 2, 3 and 4 are single-stranded ribonucleic acid viruses in the genus Flavivirus, family Flaviviridae. DENV-1–4 are called serotypes because each has different interactions with the antibodies in human blood. They share approximately two thirds of their genomes, with different genotypes existing within each serotype, which vary in disease severity.
DENV is primarily transmitted through the bite of an infected mosquito vector, with Aedes aegypti as the most common vector, although other species including Aedes albopictus may also sustain transmission.
Other rare transmission routes include perinatal transmission, blood transfusion, organ transplantation, and two cases of sexual transmission. The incubation period from exposure to symptom development is typically 4–10 days.
Dengue affects more than 100 countries in tropical and subtropical areas. Ongoing climate change, population growth, mobility and urbanization are anticipated to exacerbate dengue burden, primarily by increasing risk in endemic areas, as well as secondarily by expanding range of Aedes species mosquitoes, into new areas.
Presentation and diagnosis
Dengue is a self-limiting acute febrile illness with non-specific manifestations. Warning signs (e.g. abdominal pain, persisting vomiting, fluid accumulation) are used to detect disease progression. The hallmark of severe dengue is plasma leakage that may lead to shock and less frequently, haemorrhage and there may be liver, central nervous system, heart and other organ involvement.
Currently there is no effective prophylactic or therapeutic agent against dengue. The management of dengue remains supportive with an emphasis on maintaining hydration and, when indicated by the clinical manifestations, providing intravenous fluids.
Diagnosis of dengue infection during the acute phase can be achieved using blood collected ≤7 days after symptom onset by detection of viral RNA through nucleic acid amplification tests (NAAT), detection of viral antigens such as dengue non-structural protein 1 (NS1), by enzyme-linked immunosorbent assay (ELISA), or rapid diagnostic tests, and IgM antibodies through serologic testing.
Prevention
Persons who live in or travel to dengue endemic areas can prevent mosquito bites by using EPA-approved insect repellents and wearing clothing that covers arms and legs. The use of screened windows and doors, air conditioning, and bed nets have been shown to be protective. Chemical control of Aedes species mosquitoes is limited by widespread insecticide resistance in endemic areas.
Novel vector control methods have been developed, including the use of genetically modified mosquitoes (GMM) and Wolbachia-based methods (168). There are three leading dengue vaccines. Dengvaxia (CYD-TDV), developed by Sanofi-Pasteur, was the first licensed dengue vaccine. Qdenga (TAK-003), developed by Takeda, was recommended by SAGE in September 2023 to be considered for introduction in settings with high transmission intensity. The third, TV003, was developed by the National Institutes of Health (NIH) and is in late-stage trials.
Acknowledgements:
We would like to thank Jorge Munoz and Liliana Sanchez for their expertise and advice on the preparation of Figure 2 and 3, and Mike Johansson and Lyle Petersen for their review of the manuscript. Figure 4 was made using BioRender (BioRender.com).
Disclaimer:
The findings and conclusions in this report are those of the authors and do not necessarily represent the official position of the Centers for Disease Control and Prevention.
Footnotes
Conflicts of Interest and Source of Funding: No funding was used for this publication. GPB, LA, LK, and KA have no conflict of interest. GPB and LA are employees of the Centers for Disease Control and Prevention. LK is supported by the Intramural Research Program of NIAID.
References
- 1.Rigau-Perez JG, Clark GG, Gubler DJ, Reiter P, Sanders EJ, Vorndam AV. Dengue and dengue haemorrhagic fever. Lancet. 1998;352(9132):971–7. [DOI] [PubMed] [Google Scholar]
- 2.Nature Education. Dengue viruses Cambridge, MA: Nature Education; 2014. [Available from: https://www.nature.com/scitable/topicpage/dengue-viruses-22400925/. [Google Scholar]
- 3.Cattarino L, Rodriguez-Barraquer I, Imai N, Cummings DAT, Ferguson NM. Mapping global variation in dengue transmission intensity. Sci Transl Med. 2020;12(528). [DOI] [PubMed] [Google Scholar]
- 4.Xu Z, Bambrick H, Frentiu FD, Devine G, Yakob L, Williams G, Hu W. Projecting the future of dengue under climate change scenarios: Progress, uncertainties and research needs. PLoS Negl Trop Dis. 2020;14(3):e0008118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Messina JP, Brady OJ, Golding N, Kraemer MUG, Wint GRW, Ray SE, et al. The current and future global distribution and population at risk of dengue. Nat Microbiol. 2019;4(9):1508–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Shepard DS, Undurraga EA, Halasa YA, Stanaway JD. The global economic burden of dengue: a systematic analysis. The Lancet Infectious Diseases. 2016;16(8):935–41. [DOI] [PubMed] [Google Scholar]
- 7.Oliveira L, Itria A, Lima EC. Cost of illness and program of dengue: A systematic review. PLoS One. 2019;14(2):e0211401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hung TM, Shepard DS, Bettis AA, Nguyen HA, McBride A, Clapham HE, Turner HC. Productivity costs from a dengue episode in Asia: a systematic literature review. BMC Infect Dis. 2020;20(1):393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Laserna A, Barahona-Correa J, Baquero L, Castañeda-Cardona C, Rosselli D. Economic impact of dengue fever in Latin America and the Caribbean: a systematic review. Rev Panam Salud Publica. 2018;42:e111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Achee NL, Gould F, Perkins TA, Reiner RC Jr., Morrison AC, Ritchie SA, et al. A critical assessment of vector control for dengue prevention. PLoS Negl Trop Dis. 2015;9(5):e0003655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lee C, Lee H. Probable female to male sexual transmission of dengue virus infection. Infect Dis (Lond). 2019;51(2):150–2. [DOI] [PubMed] [Google Scholar]
- 12.Cedano JA, Mora BL, Parra-Lara LG, Manzano-Nuñez R, Rosso F. A scoping review of transmission of dengue virus from donors to recipients after solid organ transplantation. Trans R Soc Trop Med Hyg. 2019;113(8):431–6. [DOI] [PubMed] [Google Scholar]
- 13.Sabino EC, Loureiro P, Lopes ME, Capuani L, McClure C, Chowdhury D, et al. Transfusion-Transmitted Dengue and Associated Clinical Symptoms During the 2012 Epidemic in Brazil. J Infect Dis. 2016;213(5):694–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Basurko C, Matheus S, Hildéral H, Everhard S, Restrepo M, Cuadro-Alvarez E, et al. Estimating the Risk of Vertical Transmission of Dengue: A Prospective Study. Am J Trop Med Hyg. 2018;98(6):1826–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Phongsamart W, Yoksan S, Vanaprapa N, Chokephaibulkit K. Dengue virus infection in late pregnancy and transmission to the infants. Pediatr Infect Dis J. 2008;27(6):500–4. [DOI] [PubMed] [Google Scholar]
- 16.WHO/TDR. Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control: New Edition. Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control: New Edition. WHO Guidelines Approved by the Guidelines Review Committee. Geneva: 2009. [Google Scholar]
- 17.Zeng Z, Zhan J, Chen L, Chen H, Cheng S. Global, regional, and national dengue burden from 1990 to 2017: A systematic analysis based on the global burden of disease study 2017. EClinicalMedicine. 2021;32:100712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cochet A, Calba C, Jourdain F, Grard G, Durand GA, Guinard A, et al. Autochthonous dengue in mainland France, 2022: geographical extension and incidence increase. Euro Surveill. 2022;27(44). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kretschmer M, Collins J, Dale AP, Garrett B, Koski L, Zabel K, et al. Notes From the Field: First Evidence of Locally Acquired Dengue Virus Infection - Maricopa County, Arizona, November 2022. MMWR Morb Mortal Wkly Rep. 2023;72(11):290–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Pandey BD, Costello A. The dengue epidemic and climate change in Nepal. Lancet. 2019;394(10215):2150–1. [DOI] [PubMed] [Google Scholar]
- 21.Harapan H, Michie A, Yohan B, Shu PY, Mudatsir M, Sasmono RT, Imrie A. Dengue viruses circulating in Indonesia: A systematic review and phylogenetic analysis of data from five decades. Rev Med Virol. 2019;29(4):e2037. [DOI] [PubMed] [Google Scholar]
- 22.Li Z, Wang J, Cheng X, Hu H, Guo C, Huang J, et al. The worldwide seroprevalence of DENV, CHIKV and ZIKV infection: A systematic review and meta-analysis. PLoS Negl Trop Dis. 2021;15(4):e0009337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.L’Azou M, Moureau A, Sarti E, Nealon J, Zambrano B, Wartel TA, et al. Symptomatic Dengue in Children in 10 Asian and Latin American Countries. N Engl J Med. 2016;374(12):1155–66. [DOI] [PubMed] [Google Scholar]
- 24.Rodriguez-Barraquer I, Salje H, Cummings DA. Opportunities for improved surveillance and control of dengue from age-specific case data. Elife. 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cummings DA, Iamsirithaworn S, Lessler JT, McDermott A, Prasanthong R, Nisalak A, et al. The impact of the demographic transition on dengue in Thailand: insights from a statistical analysis and mathematical modeling. PLoS Med. 2009;6(9):e1000139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ooi EE, Goh KT, Gubler DJ. Dengue prevention and 35 years of vector control in Singapore. Emerg Infect Dis. 2006;12(6):887–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Halstead SB, Lan NT, Myint TT, Shwe TN, Nisalak A, Kalyanarooj S, et al. Dengue hemorrhagic fever in infants: research opportunities ignored. Emerg Infect Dis. 2002;8(12):1474–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kliks SC, Nimmanitya S, Nisalak A, Burke DS. Evidence that maternal dengue antibodies are important in the development of dengue hemorrhagic fever in infants. Am J Trop Med Hyg. 1988;38(2):411–9. [DOI] [PubMed] [Google Scholar]
- 29.O’Driscoll M, Buddhari D, Huang AT, Waickman A, Kaewhirun S, Iamsirithaworn S, et al. Maternally derived antibody titer dynamics and risk of hospitalized infant dengue disease. Proc Natl Acad Sci U S A. 2023;120(41):e2308221120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Halstead SB, Yamarat C. Recent Epidemics of Hemorrhagic Fever in Thailand. Observations Related to Pathogenesis of a “New” Dengue Disease. Am J Public Health Nations Health. 1965;55(9):1386–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sangkaew S, Ming D, Boonyasiri A, Honeyford K, Kalayanarooj S, Yacoub S, et al. Risk predictors of progression to severe disease during the febrile phase of dengue: a systematic review and meta-analysis. Lancet Infect Dis. 2021;21(7):1014–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Olson MF, Juarez JG, Kraemer MUG, Messina JP, Hamer GL. Global patterns of aegyptism without arbovirus. PLoS Negl Trop Dis. 2021;15(5):e0009397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Louis VR, Phalkey R, Horstick O, Ratanawong P, Wilder-Smith A, Tozan Y, Dambach P. Modeling tools for dengue risk mapping - a systematic review. Int J Health Geogr. 2014;13:50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Eder M, Cortes F, Teixeira de Siqueira Filha N, Araújo de França GV, Degroote S, Braga C, et al. Scoping review on vector-borne diseases in urban areas: transmission dynamics, vectorial capacity and co-infection. Infect Dis Poverty. 2018;7(1):90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Berry IM, Melendrez MC, Pollett S, Figueroa K, Buddhari D, Klungthong C, et al. Precision Tracing of Household Dengue Spread Using Inter- and Intra-Host Viral Variation Data, Kamphaeng Phet, Thailand. Emerg Infect Dis. 2021;27(6):1637–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Salje H, Wesolowski A, Brown TS, Kiang MV, Berry IM, Lefrancq N, et al. Reconstructing unseen transmission events to infer dengue dynamics from viral sequences. Nat Commun. 2021;12(1):1810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chen Y, Li N, Lourenço J, Wang L, Cazelles B, Dong L, et al. Measuring the effects of COVID-19-related disruption on dengue transmission in southeast Asia and Latin America: a statistical modelling study. Lancet Infect Dis. 2022;22(5):657–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Osman S, Preet R. Dengue, chikungunya and Zika in GeoSentinel surveillance of international travellers: a literature review from 1995 to 2020. J Travel Med. 2020;27(8). [DOI] [PubMed] [Google Scholar]
- 39.Huits R, Schwartz E. Fatal outcomes of imported dengue fever in adult travelers from non-endemic areas are associated with primary infections. J Travel Med. 2021;28(5). [DOI] [PubMed] [Google Scholar]
- 40.Olivero RM, Hamer DH, MacLeod WB, Benoit CM, Sanchez-Vegas C, Jentes ES, et al. Dengue Virus Seroconversion in Travelers to Dengue-Endemic Areas. Am J Trop Med Hyg. 2016;95(5):1130–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Montoya M, Gresh L, Mercado JC, Williams KL, Vargas MJ, Gutierrez G, et al. Symptomatic versus inapparent outcome in repeat dengue virus infections is influenced by the time interval between infections and study year. PLoS Negl Trop Dis. 2013;7(8):e2357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Anderson KB, Gibbons RV, Cummings DA, Nisalak A, Green S, Libraty DH, et al. A shorter time interval between first and second dengue infections is associated with protection from clinical illness in a school-based cohort in Thailand. J Infect Dis. 2014;209(3):360–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.WHO. Handbook for clinical management of dengue. Geneva: 2012. [Google Scholar]
- 44.WHO. Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control. Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control: New Edition. Geneva: World Health Organization Copyright © 2009, World Health Organization.; 2009. [PubMed] [Google Scholar]
- 45.Thomas EA, John M, Kanish B. Mucocutaneous manifestations of Dengue fever. Indian J Dermatol. 2010;55(1):79–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kalayanarooj S, Vaughn DW, Nimmannitya S, Green S, Suntayakorn S, Kunentrasai N, et al. Early clinical and laboratory indicators of acute dengue illness. J Infect Dis. 1997;176(2):313–21. [DOI] [PubMed] [Google Scholar]
- 47.Simmons CP, Farrar JJ, Nguyen v V, Wills B. Dengue. N Engl J Med. 2012;366(15):1423–32. [DOI] [PubMed] [Google Scholar]
- 48.Lam PK, Tam DT, Diet TV, Tam CT, Tien NT, Kieu NT, et al. Clinical characteristics of Dengue shock syndrome in Vietnamese children: a 10-year prospective study in a single hospital. Clin Infect Dis. 2013;57(11):1577–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Malavige GN, Ogg GS. Pathogenesis of vascular leak in dengue virus infection. Immunology. 2017;151(3):261–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Yacoub S, Wertheim H, Simmons CP, Screaton G, Wills B. Microvascular and endothelial function for risk prediction in dengue: an observational study. Lancet. 2015;385 Suppl 1:S102. [DOI] [PubMed] [Google Scholar]
- 51.Meltzer E, Heyman Z, Bin H, Schwartz E. Capillary leakage in travelers with dengue infection: implications for pathogenesis. Am J Trop Med Hyg. 2012;86(3):536–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Yacoub S, Wertheim H, Simmons CP, Screaton G, Wills B. Cardiovascular manifestations of the emerging dengue pandemic. Nat Rev Cardiol. 2014;11(6):335–45. [DOI] [PubMed] [Google Scholar]
- 53.Alexander N, Balmaseda A, Coelho IC, Dimaano E, Hien TT, Hung NT, et al. Multicentre prospective study on dengue classification in four South-east Asian and three Latin American countries. Trop Med Int Health. 2011;16(8):936–48. [DOI] [PubMed] [Google Scholar]
- 54.Jayarajah U, Dissanayake U, Abeysuriya V, De Silva PK, Jayawardena P, Kulatunga A, et al. Comparing the 2009 and 1997 World Health Organization dengue case classifications in a large cohort of South Asian patients. J Infect Dev Ctries. 2020;14(7):781–7. [DOI] [PubMed] [Google Scholar]
- 55.Chagas GCL, Rangel AR, Noronha LM, Veloso FCS, Kassar SB, Oliveira MJC, et al. Risk factors for mortality in patients with dengue: A systematic review and meta-analysis. Trop Med Int Health. 2022;27(8):656–68. [DOI] [PubMed] [Google Scholar]
- 56.Trieu HT, Khanh LP, Ming DKY, Quang CH, Phan TQ, Van VCN, et al. The compensatory reserve index predicts recurrent shock in patients with severe dengue. BMC Med. 2022;20(1):109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Trung DT, Trieu HT, Wills BA. Microvascular Fluid Exchange: Implications of the Revised Starling Model for Resuscitation of Dengue Shock Syndrome. Front Med (Lausanne). 2020;7:601520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wills BA, Oragui EE, Stephens AC, Daramola OA, Dung NM, Loan HT, et al. Coagulation abnormalities in dengue hemorrhagic Fever: serial investigations in 167 Vietnamese children with Dengue shock syndrome. Clin Infect Dis. 2002;35(3):277–85. [DOI] [PubMed] [Google Scholar]
- 59.Chia PY, Thein TL, Ong SWX, Lye DC, Leo YS. Severe dengue and liver involvement: an overview and review of the literature. Expert Rev Anti Infect Ther. 2020;18(3):181–9. [DOI] [PubMed] [Google Scholar]
- 60.Carod-Artal FJ, Wichmann O, Farrar J, Gascon J. Neurological complications of dengue virus infection. Lancet Neurol. 2013;12(9):906–19. [DOI] [PubMed] [Google Scholar]
- 61.Nicacio JM, Gomes OV, Carmo RFD, Nunes SLP, Rocha J, Souza CDF, et al. Heart Disease and Arboviruses: A Systematic Review and Meta-Analysis. Viruses. 2022;14(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Diptyanusa A, Phumratanaprapin W. Predictors and Outcomes of Dengue-Associated Acute Kidney Injury. Am J Trop Med Hyg. 2021;105(1):24–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Vijitha V, Dave TV, Murthy SI, Ali MJ, Dave VP, Pappuru RR, Narayanan R. Severe ocular and adnexal complications in dengue hemorrhagic fever: A report of 29 eyes. Indian Journal of Ophthalmology. 2021;69(3):617–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Machado CR, Machado ES, Rohloff RD, Azevedo M, Campos DP, de Oliveira RB, Brasil P. Is pregnancy associated with severe dengue? A review of data from the Rio de Janeiro surveillance information system. PLoS Negl Trop Dis. 2013;7(5):e2217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Paixao ES, Harron K, Campbell O, Teixeira MG, Costa M, Barreto ML, Rodrigues LC. Dengue in pregnancy and maternal mortality: a cohort analysis using routine data. Sci Rep. 2018;8(1):9938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Rathore SS, Oberoi S, Hilliard J, Raja R, Ahmed NK, Vishwakarma Y, et al. Maternal and foetal-neonatal outcomes of dengue virus infection during pregnancy. Trop Med Int Health. 2022;27(7):619–29. [DOI] [PubMed] [Google Scholar]
- 67.Paixao ES, Teixeira MG, Costa M, Rodrigues LC. Dengue during pregnancy and adverse fetal outcomes: a systematic review and meta-analysis. Lancet Infect Dis. 2016;16(7):857–65. [DOI] [PubMed] [Google Scholar]
- 68.Bothra A, Maheswari A, Singh M, Pawar M, Jodhani K. Cutaneous manifestations of viral outbreaks. Australas J Dermatol. 2021;62(1):27–36. [DOI] [PubMed] [Google Scholar]
- 69.Sigera PC, Rajapakse S, Weeratunga P, De Silva NL, Gomes L, Malavige GN, et al. Dengue and post-infection fatigue: findings from a prospective cohort-the Colombo Dengue Study. Trans R Soc Trop Med Hyg. 2021;115(6):669–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Srikiatkhachorn A, Krautrachue A, Ratanaprakarn W, Wongtapradit L, Nithipanya N, Kalayanarooj S, et al. Natural history of plasma leakage in dengue hemorrhagic fever: a serial ultrasonographic study. Pediatr Infect Dis J. 2007;26(4):283–90; discussion 91–2. [DOI] [PubMed] [Google Scholar]
- 71.Dewan N, Zuluaga D, Osorio L, Krienke ME, Bakker C, Kirsch J. Ultrasound in Dengue: A Scoping Review. Am J Trop Med Hyg. 2021;104(3):826–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Halstead SB, Tissera H, Kumarasinghe M, Fernando L. Comments on “Dengue management in triage using ultrasound in children from Cambodia: A prospective cohort study”. Lancet Reg Health West Pac. 2022;25:100527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Vuong NL, Lam PK, Ming DKY, Duyen HTL, Nguyen NM, Tam DTH, et al. Combination of inflammatory and vascular markers in the febrile phase of dengue is associated with more severe outcomes. Elife. 2021;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Yuan K, Chen Y, Zhong M, Lin Y, Liu L. Risk and predictive factors for severe dengue infection: A systematic review and meta-analysis. PLoS One. 2022;17(4):e0267186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Obi JO, Gutierrez-Barbosa H, Chua JV, Deredge DJ. Current Trends and Limitations in Dengue Antiviral Research. Trop Med Infect Dis. 2021;6(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Wilder-Smith A, Ooi EE, Horstick O, Wills B. Dengue. Lancet. 2019;393(10169):350–63. [DOI] [PubMed] [Google Scholar]
- 77.Suputtamongkol Y, Avirutnan P, Mairiang D, Angkasekwinai N, Niwattayakul K, Yamasmith E, et al. Ivermectin Accelerates Circulating Nonstructural Protein 1 (NS1) Clearance in Adult Dengue Patients: A Combined Phase 2/3 Randomized Double-blinded Placebo Controlled Trial. Clin Infect Dis. 2021;72(10):e586–e93. [DOI] [PubMed] [Google Scholar]
- 78.Rajapakse S, de Silva NL, Weeratunga P, Rodrigo C, Fernando SD. Prophylactic and therapeutic interventions for bleeding in dengue: a systematic review. Trans R Soc Trop Med Hyg. 2017;111(10):433–9. [DOI] [PubMed] [Google Scholar]
- 79.Kaptein SJF, Goethals O, Kiemel D, Marchand A, Kesteleyn B, Bonfanti JF, et al. A pan-serotype dengue virus inhibitor targeting the NS3-NS4B interaction. Nature. 2021;598(7881):504–9. [DOI] [PubMed] [Google Scholar]
- 80.Tien SM, Chang PC, Lai YC, Chuang YC, Tseng CK, Kao YS, et al. Therapeutic efficacy of humanized monoclonal antibodies targeting dengue virus nonstructural protein 1 in the mouse model. PLoS Pathog. 2022;18(4):e1010469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Pan American Health Organization. Dengue: guidelines for patient care in the Region of the Americas, 2nd edition. PAHO, Washington, D.C., 2016. https://iris.paho.org/handle/10665.2/31207 Accessed 25 October 2022. [Google Scholar]
- 82.Médecins Sans Frontières. MSF medical guidelines: Dengue. https://medicalguidelines.msf.org/en/viewport/CG/english/dengue-16690007.html Accessed 25 October 2022 [
- 83.Pan American Health Organization. Guidelines for the Clinical Diagnosis and Treatment of Dengue, Chikungunya, and Zika. Washington, D.C.: PAHO, 2022. 10.37774/9789275124871 Accessed 25 October 2022. [DOI] [Google Scholar]
- 84.Rosenberger KD, Lum L, Alexander N, Junghanss T, Wills B, Jaenisch T, Group DCS. Vascular leakage in dengue--clinical spectrum and influence of parenteral fluid therapy. Trop Med Int Health. 2016;21(3):445–53. [DOI] [PubMed] [Google Scholar]
- 85.Lum LC, Abdel-Latif Mel A, Goh AY, Chan PW, Lam SK. Preventive transfusion in Dengue shock syndrome-is it necessary? J Pediatr. 2003;143(5):682–4. [DOI] [PubMed] [Google Scholar]
- 86.Lye DC, Archuleta S, Syed-Omar SF, Low JG, Oh HM, Wei Y, et al. Prophylactic platelet transfusion plus supportive care versus supportive care alone in adults with dengue and thrombocytopenia: a multicentre, open-label, randomised, superiority trial. Lancet. 2017;389(10079):1611–8. [DOI] [PubMed] [Google Scholar]
- 87.Dung NM, Day NP, Tam DT, Loan HT, Chau HT, Minh LN, et al. Fluid replacement in dengue shock syndrome: a randomized, double-blind comparison of four intravenous-fluid regimens. Clin Infect Dis. 1999;29(4):787–94. [DOI] [PubMed] [Google Scholar]
- 88.Kalayanarooj S. Choice of colloidal solutions in dengue hemorrhagic fever patients. J Med Assoc Thai. 2008;91 Suppl 3:S97–103. [PubMed] [Google Scholar]
- 89.Ngo NT, Cao XT, Kneen R, Wills B, Nguyen VM, Nguyen TQ, et al. Acute management of dengue shock syndrome: a randomized double-blind comparison of 4 intravenous fluid regimens in the first hour. Clin Infect Dis. 2001;32(2):204–13. [DOI] [PubMed] [Google Scholar]
- 90.Wills BA, Nguyen MD, Ha TL, Dong TH, Tran TN, Le TT, et al. Comparison of three fluid solutions for resuscitation in dengue shock syndrome. N Engl J Med. 2005;353(9):877–89. [DOI] [PubMed] [Google Scholar]
- 91.Sharp TM, Anderson KB, Katzelnick LC, Clapham H, Johansson MA, Morrison AC, et al. Knowledge gaps in the epidemiology of severe dengue impede vaccine evaluation. Lancet Infect Dis. 2022;22(2):e42–e51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Centers for Disease Control and Prevention (CDC). Molecular Tests for Dengue Virus Atlanta, GA: 2019. [Available from: https://www.cdc.gov/dengue/healthcare-providers/testing/molecular-tests/index.html. [Google Scholar]
- 93.Sharp TM, Fischer M, Munoz-Jordan JL, Paz-Bailey G, Staples JE, Gregory CJ, Waterman SH. Dengue and Zika Virus Diagnostic Testing for Patients with a Clinically Compatible Illness and Risk for Infection with Both Viruses. MMWR Recomm Rep. 2019;68(1):1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Fisher R, Lustig Y, Sklan EH, Schwartz E. The Role of NS1 Protein in the Diagnosis of Flavivirus Infections. Viruses. 2023;15(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Centers for Disease Control and Prevention (CDC). Dengue Virus Antigen Detection 2019. [Available from: https://www.cdc.gov/dengue/healthcare-providers/testing/antigen-detection.html.
- 96.Centers for Disease Control and Prevention (CDC). Serologic Tests for Dengue Virus 2022. [Available from: https://www.cdc.gov/dengue/healthcare-providers/testing/serologic-tests.html.
- 97.Macedo JVL, Frias IAM, Oliveira MDL, Zanghelini F, Andrade CAS. A systematic review and meta-analysis on the accuracy of rapid immunochromatographic tests for dengue diagnosis. Eur J Clin Microbiol Infect Dis. 2022;41(9):1191–201. [DOI] [PubMed] [Google Scholar]
- 98.Mata VE, Andrade CAF, Passos SRL, Hokerberg YHM, Fukuoka LVB, Silva SAD. Rapid immunochromatographic tests for the diagnosis of dengue: a systematic review and meta-analysis. Cad Saude Publica. 2020;36(6):e00225618. [DOI] [PubMed] [Google Scholar]
- 99.King CA, Wegman AD, Endy TP. Mobilization and Activation of the Innate Immune Response to Dengue Virus. Front Cell Infect Microbiol. 2020;10:574417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Halstead SB, Mahalingam S, Marovich MA, Ubol S, Mosser DM. Intrinsic antibody-dependent enhancement of microbial infection in macrophages: disease regulation by immune complexes. Lancet Infect Dis. 2010;10(10):712–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Halstead SB. In vivo enhancement of dengue virus infection in rhesus monkeys by passively transferred antibody. J Infect Dis. 1979;140(4):527–33. [DOI] [PubMed] [Google Scholar]
- 102.Halstead SB, O’Rourke EJ. Antibody-enhanced dengue virus infection in primate leukocytes. Nature. 1977;265(5596):739–41. [DOI] [PubMed] [Google Scholar]
- 103.Waggoner JJ, Katzelnick LC, Burger-Calderon R, Gallini J, Moore RH, Kuan G, et al. Antibody-Dependent Enhancement of Severe Disease Is Mediated by Serum Viral Load in Pediatric Dengue Virus Infections. J Infect Dis. 2020;221(11):1846–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Katzelnick LC, Narvaez C, Arguello S, Lopez Mercado B, Collado D, Ampie O, et al. Zika virus infection enhances future risk of severe dengue disease. Science. 2020;369(6507):1123–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Katzelnick LC, Gresh L, Halloran ME, Mercado JC, Kuan G, Gordon A, et al. Antibody-dependent enhancement of severe dengue disease in humans. Science. 2017;358(6365):929–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Mizumoto K, Ejima K, Yamamoto T, Nishiura H. On the risk of severe dengue during secondary infection: a systematic review coupled with mathematical modeling. J Vector Borne Dis. 2014;51(3):153–64. [PubMed] [Google Scholar]
- 107.Kwissa M, Nakaya HI, Onlamoon N, Wrammert J, Villinger F, Perng GC, et al. Dengue virus infection induces expansion of a CD14(+)CD16(+) monocyte population that stimulates plasmablast differentiation. Cell Host Microbe. 2014;16(1):115–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Michlmayr D, Andrade P, Gonzalez K, Balmaseda A, Harris E. CD14(+)CD16(+) monocytes are the main target of Zika virus infection in peripheral blood mononuclear cells in a paediatric study in Nicaragua. Nat Microbiol. 2017;2(11):1462–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Wang TT, Sewatanon J, Memoli MJ, Wrammert J, Bournazos S, Bhaumik SK, et al. IgG antibodies to dengue enhanced for FcgammaRIIIA binding determine disease severity. Science. 2017;355(6323):395–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Bournazos S, Vo HTM, Duong V, Auerswald H, Ly S, Sakuntabhai A, et al. Antibody fucosylation predicts disease severity in secondary dengue infection. Science. 2021;372(6546):1102–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Upasani V, Vo HTM, Auerswald H, Laurent D, Heng S, Duong V, et al. Direct Infection of B Cells by Dengue Virus Modulates B Cell Responses in a Cambodian Pediatric Cohort. Front Immunol. 2020;11:594813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Glasner DR, Puerta-Guardo H, Beatty PR, Harris E. The Good, the Bad, and the Shocking: The Multiple Roles of Dengue Virus Nonstructural Protein 1 in Protection and Pathogenesis. Annu Rev Virol. 2018;5(1):227–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Aye KS, Charngkaew K, Win N, Wai KZ, Moe K, Punyadee N, et al. Pathologic highlights of dengue hemorrhagic fever in 13 autopsy cases from Myanmar. Hum Pathol. 2014;45(6):1221–33. [DOI] [PubMed] [Google Scholar]
- 114.Mongkolsapaya J, Dejnirattisai W, Xu XN, Vasanawathana S, Tangthawornchaikul N, Chairunsri A, et al. Original antigenic sin and apoptosis in the pathogenesis of dengue hemorrhagic fever. Nat Med. 2003;9(7):921–7. [DOI] [PubMed] [Google Scholar]
- 115.Hatch S, Endy TP, Thomas S, Mathew A, Potts J, Pazoles P, et al. Intracellular cytokine production by dengue virus-specific T cells correlates with subclinical secondary infection. J Infect Dis. 2011;203(9):1282–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Weiskopf D, Angelo MA, de Azeredo EL, Sidney J, Greenbaum JA, Fernando AN, et al. Comprehensive analysis of dengue virus-specific responses supports an HLA-linked protective role for CD8+ T cells. Proc Natl Acad Sci U S A. 2013;110(22):E2046–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Wijeratne DT, Fernando S, Gomes L, Jeewandara C, Ginneliya A, Samarasekara S, et al. Quantification of dengue virus specific T cell responses and correlation with viral load and clinical disease severity in acute dengue infection. PLoS Negl Trop Dis. 2018;12(10):e0006540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Weiskopf D, Bangs DJ, Sidney J, Kolla RV, De Silva AD, de Silva AM, et al. Dengue virus infection elicits highly polarized CX3CR1+ cytotoxic CD4+ T cells associated with protective immunity. Proc Natl Acad Sci U S A. 2015;112(31):E4256–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Haltaufderhyde K, Srikiatkhachorn A, Green S, Macareo L, Park S, Kalayanarooj S, et al. Activation of Peripheral T Follicular Helper Cells During Acute Dengue Virus Infection. J Infect Dis. 2018;218(10):1675–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Rouers A, Chng MHY, Lee B, Rajapakse MP, Kaur K, Toh YX, et al. Immune cell phenotypes associated with disease severity and long-term neutralizing antibody titers after natural dengue virus infection. Cell Rep Med. 2021;2(5):100278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Clapham HE, Cummings DAT, Johansson MA. Immune status alters the probability of apparent illness due to dengue virus infection: Evidence from a pooled analysis across multiple cohort and cluster studies. PLoS Negl Trop Dis. 2017;11(9):e0005926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Nisalak A, Clapham HE, Kalayanarooj S, Klungthong C, Thaisomboonsuk B, Fernandez S, et al. Forty Years of Dengue Surveillance at a Tertiary Pediatric Hospital in Bangkok, Thailand, 1973–2012. Am J Trop Med Hyg. 2016;94(6):1342–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Soo KM, Khalid B, Ching SM, Chee HY. Meta-Analysis of Dengue Severity during Infection by Different Dengue Virus Serotypes in Primary and Secondary Infections. PLoS One. 2016;11(5):e0154760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Salje H, Cummings DAT, Rodriguez-Barraquer I, Katzelnick LC, Lessler J, Klungthong C, et al. Reconstruction of antibody dynamics and infection histories to evaluate dengue risk. Nature. 2018;557(7707):719–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Aguas R, Dorigatti I, Coudeville L, Luxemburger C, Ferguson NM. Cross-serotype interactions and disease outcome prediction of dengue infections in Vietnam. Sci Rep. 2019;9(1):9395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Halstead SB. Neutralization and antibody-dependent enhancement of dengue viruses. Adv Virus Res. 2003;60:421–67. [DOI] [PubMed] [Google Scholar]
- 127.Katzelnick LC, Coello Escoto A, Huang AT, Garcia-Carreras B, Chowdhury N, Maljkovic Berry I, et al. Antigenic evolution of dengue viruses over 20 years. Science. 2021;374(6570):999–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Forshey BM, Reiner RC, Olkowski S, Morrison AC, Espinoza A, Long KC, et al. Incomplete Protection against Dengue Virus Type 2 Re-infection in Peru. PLoS Negl Trop Dis. 2016;10(2):e0004398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Kochel TJ, Watts DM, Halstead SB, Hayes CG, Espinoza A, Felices V, et al. Effect of dengue-1 antibodies on American dengue-2 viral infection and dengue haemorrhagic fever. Lancet. 2002;360(9329):310–2. [DOI] [PubMed] [Google Scholar]
- 130.OhAinle M, Balmaseda A, Macalalad AR, Tellez Y, Zody MC, Saborio S, et al. Dynamics of dengue disease severity determined by the interplay between viral genetics and serotype-specific immunity. Sci Transl Med. 2011;3(114):114ra28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Rodriguez-Roche R, Sanchez L, Burgher Y, Rosario D, Alvarez M, Kouri G, et al. Virus role during intraepidemic increase in dengue disease severity. Vector Borne Zoonotic Dis. 2011;11(6):675–81. [DOI] [PubMed] [Google Scholar]
- 132.Olkowski S, Forshey BM, Morrison AC, Rocha C, Vilcarromero S, Halsey ES, et al. Reduced risk of disease during postsecondary dengue virus infections. J Infect Dis. 2013;208(6):1026–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Hadinegoro SR, Arredondo-Garcia JL, Capeding MR, Deseda C, Chotpitayasunondh T, Dietze R, et al. Efficacy and Long-Term Safety of a Dengue Vaccine in Regions of Endemic Disease. N Engl J Med. 2015;373(13):1195–206. [DOI] [PubMed] [Google Scholar]
- 134.Rivera L, Biswal S, Saez-Llorens X, Reynales H, Lopez-Medina E, Borja-Tabora C, et al. Three-year Efficacy and Safety of Takeda’s Dengue Vaccine Candidate (TAK-003). Clin Infect Dis. 2022;75(1):107–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Barba-Spaeth G, Dejnirattisai W, Rouvinski A, Vaney MC, Medits I, Sharma A, et al. Structural basis of potent Zika-dengue virus antibody cross-neutralization. Nature. 2016;536(7614):48–53. [DOI] [PubMed] [Google Scholar]
- 136.Sharma A, Zhang X, Dejnirattisai W, Dai X, Gong D, Wongwiwat W, et al. The epitope arrangement on flavivirus particles contributes to Mab C10’s extraordinary neutralization breadth across Zika and dengue viruses. Cell. 2021;184(25):6052–66 e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.White LJ, Young EF, Stoops MJ, Henein SR, Adams EC, Baric RS, de Silva AM. Defining levels of dengue virus serotype-specific neutralizing antibodies induced by a live attenuated tetravalent dengue vaccine (TAK-003). PLoS Negl Trop Dis. 2021;15(3):e0009258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Paz-Bailey G, Adams L, Wong JM, Poehling KA, Chen WH, McNally V, et al. Dengue Vaccine: Recommendations of the Advisory Committee on Immunization Practices, United States, 2021. MMWR Recomm Rep. 2021;70(6):1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.European Medicines Agency. Dengue Tetravalent Vaccine (Live, Attenuated) Takeda: Opinion on medicine for use outside EU 2022. [updated October 10, 2022. Available from: https://www.ema.europa.eu/en/opinion-medicine-use-outside-EU/human/dengue-tetravalent-vaccine-live-attenuated-takeda.
- 140.Takeda. Takeda Receives Positive CHMP Opinion Recommending Approval of Dengue Vaccine Candidate in EU and Dengue-Endemic Countries 2022. [Available from: https://www.takeda.com/newsroom/newsreleases/2022/Positive-CHMP-Opinion-Recommending-Approval-of-Dengue-vaccine/.
- 141.Matsuyama K, Huang G. Japanese pharmaceutical firm Takeda lowers price of dengue shot in Brazil and Indonesia. Japan Times. 2022. [Google Scholar]
- 142.Whitehead SS. Development of TV003/TV005, a single dose, highly immunogenic live attenuated dengue vaccine; what makes this vaccine different from the Sanofi-Pasteur CYD vaccine? Expert Rev Vaccines. 2016;15(4):509–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Huang CH, Tsai YT, Wang SF, Wang WH, Chen YH. Dengue vaccine: an update. Expert Rev Anti Infect Ther. 2021;19(12):1495–502. [DOI] [PubMed] [Google Scholar]
- 144.Park J, Kim J, Jang YS. Current status and perspectives on vaccine development against dengue virus infection. J Microbiol. 2022;60(3):247–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.World Health O Dengue vaccine: WHO position paper, July 2016 - recommendations. Vaccine. 2017;35(9):1200–1. [DOI] [PubMed] [Google Scholar]
- 146.Sridhar S, Luedtke A, Langevin E, Zhu M, Bonaparte M, Machabert T, et al. Effect of Dengue Serostatus on Dengue Vaccine Safety and Efficacy. New England Journal of Medicine. 2018;379(4):327–40. [DOI] [PubMed] [Google Scholar]
- 147.World Health Organization. Dengue vaccine: WHO position paper, September 2018 - Recommendations. Vaccine. 2019;37(35):4848–9. [DOI] [PubMed] [Google Scholar]
- 148.Hadinegoro SR, Arredondo-García JL, Capeding MR, Deseda C, Chotpitayasunondh T, Dietze R, et al. Efficacy and Long-Term Safety of a Dengue Vaccine in Regions of Endemic Disease. New England Journal of Medicine. 2015;373(13):1195–206. [DOI] [PubMed] [Google Scholar]
- 149.Henein S, Swanstrom J, Byers AM, Moser JM, Shaik SF, Bonaparte M, et al. Dissecting Antibodies Induced by a Chimeric Yellow Fever-Dengue, Live-Attenuated, Tetravalent Dengue Vaccine (CYD-TDV) in Naive and Dengue-Exposed Individuals. J Infect Dis. 2017;215(3):351–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Fongwen N, Wilder-Smith A, Gubler DJ, Ooi EE, EM TS, de Lamballerie X, et al. Target product profile for a dengue pre-vaccination screening test. PLoS Negl Trop Dis. 2021;15(7):e0009557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Centers for Disease Control and Prevention (CDC). Laboratory Testing Requirements for Vaccination with Dengvaxia Dengue Vaccine Atlanta, GA: 2022. [Available from: https://www.cdc.gov/dengue/vaccine/hcp/testing.html. [Google Scholar]
- 152.Sharma M, Tricou V, Rauscher M, Watkins H, Messere N, Cox L, et al. Efficacy, safety and immunogenicity of Takeda’s tetravalent dengue vaccine candidate (TAK-003) after 4.5 years of follow-up. American Society of Tropical Medicine and Hygiene (ASTMH) Annual Meeting; October 30-November 3, 2022; Seattle2022. [Google Scholar]
- 153.Dal-Re R, Launay O. Public trust on regulatory decisions: The European Medicines Agency and the AstraZeneca COVID-19 vaccine label. Vaccine. 2021;39(30):4029–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Huang KMaG. Japanese pharmaceutical firm Takeda lowers price of dengue shot in Brazil and Indonesia. Japan Times. 2022. [Google Scholar]
- 155.SAGE. Highlights from the Meeting of the Strategic Advisory Group of Experts (SAGE) on Immunization, 25–29 September 2023. Geneva, Switzerland: 2023. [Available from: https://cdn.who.int/media/docs/default-source/2021-dha-docs/highlights-3.pdf?sfvrsn=9237c77d_1. [Google Scholar]
- 156.Takeda. Takeda’s Biologics License Application (BLA) for Dengue Vaccine Candidate (TAK-003) Granted Priority Review by U.S. Food and Drug Administration 2022. [Available from: https://www.takeda.com/newsroom/newsreleases/2022/takedas-biologics-license-application-bla-for-dengue-vaccine-candidate-tak-003-granted-priority-review-by-us-food-and-drug-administration/.
- 157.Durbin AP, Kirkpatrick BD, Pierce KK, Schmidt AC, Whitehead SS. Development and clinical evaluation of multiple investigational monovalent DENV vaccines to identify components for inclusion in a live attenuated tetravalent DENV vaccine. Vaccine. 2011;29(42):7242–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Durbin AP, Kirkpatrick BD, Pierce KK, Elwood D, Larsson CJ, Lindow JC, et al. A Single Dose of Any of Four Different Live Attenuated Tetravalent Dengue Vaccines Is Safe and Immunogenic in Flavivirus-naive Adults: A Randomized, Double-blind Clinical Trial. The Journal of Infectious Diseases. 2013;207(6):957–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Butantan Institute. Phase III Trial to Evaluate Efficacy and Safety of a Tetravalent Dengue Vaccine 2015. [Available from: https://clinicaltrials.gov/ct2/show/NCT02406729.
- 160.Merck. Merck and Instituto Butantan Announce Collaboration Agreement to Develop Vaccines to Protect Against Dengue Infections 2018. [Available from: https://www.merck.com/news/merck-and-instituto-butantan-announce-collaboration-agreement-to-develop-vaccines-to-protect-against-dengue-infections/. [Google Scholar]
- 161.Nogueira ML, Cintra AT, Moreira JA, et al. Efficacy and safety of Butantan-DV Live-attenuated tetravalent dengue vaccine from a phase 3 clinical trial i children, adolescent and adults. In: Hygiene AAoTMa, editor. ASTMH; October/20/2023; Chicaco, IL: 2023. [Google Scholar]
- 162.Butantan Institute. Butantan’s dengue vaccine has 79.6% efficacy, partial results from 2-year follow-up show Sao Paulo, Brazil 2022. [Available from: https://butantan.gov.br/noticias/butantan’s-dengue-vaccine-has-79.6-efficacy-partial-results-from-2-year-follow-up-show.
- 163.Lindsay SW, Davies M, Alabaster G, Altamirano H, Jatta E, Jawara M, et al. Recommendations for building out mosquito-transmitted diseases in sub-Saharan Africa: the DELIVER mnemonic. Philos Trans R Soc Lond B Biol Sci. 2021;376(1818):20190814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.CDC, 2019. Dengue—Prevent Mosquito Bites. Atlanta, GA: Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Division of Vector-Borne Diseases (DVBD). Available at: https://www.cdc.gov/dengue/prevention/prevent-mosquito-bites.html#:∼:text=Use%20air%20conditioning%2C%20if%20available,%2C%20flowerpots%2C%20or%20trash%20containers. [Accessed 10 October 2021]. [Google Scholar]
- 165.Manrique-Saide P Use of insecticide-treated house screens to reduce infestations of dengue virus vectors, Mexico. Emerg Infect Dis. 2015;21:308–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Lenhart A, Orelus N, Maskill R, Alexander N, Streit T, McCall PJ. Insecticide-treated bednets to control dengue vectors: preliminary evidence from a controlled trial in Haiti. Trop Med Int Health. 2008;13(1):56–67. [DOI] [PubMed] [Google Scholar]
- 167.Moyes CL, Vontas J, Martins AJ, Ng LC, Koou SY, Dusfour I, et al. Contemporary status of insecticide resistance in the major Aedes vectors of arboviruses infecting humans. PLoS neglected tropical diseases. 2017;11(7):e0005625–e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Wang G-H, Gamez S, Raban RR, Marshall JM, Alphey L, Li M, et al. Combating mosquito-borne diseases using genetic control technologies. Nature communications. 2021;12(1):4388-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Dobson SL. When More is Less: Mosquito Population Suppression Using Sterile, Incompatible and Genetically Modified Male Mosquitoes. Journal of Medical Entomology. 2021;58(5):1980–6. [DOI] [PubMed] [Google Scholar]
- 170.Hilgenboecker K, Hammerstein P, Schlattmann P, Telschow A, Werren JH. How many species are infected with Wolbachia?--A statistical analysis of current data. FEMS Microbiol Lett. 2008;281(2):215–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.McGraw EA, O’Neill SL. Beyond insecticides: new thinking on an ancient problem. Nat Rev Microbiol. 2013;11(3):181–93. [DOI] [PubMed] [Google Scholar]
- 172.O’Connor L, Plichart C, Sang AC, Brelsfoard CL, Bossin HC, Dobson SL. Open release of male mosquitoes infected with a wolbachia biopesticide: field performance and infection containment. PLoS Negl Trop Dis. 2012;6(11):e1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Hoffmann AA, Montgomery BL, Popovici J, Iturbe-Ormaetxe I, Johnson PH, Muzzi F, et al. Successful establishment of Wolbachia in Aedes populations to suppress dengue transmission. Nature. 2011;476(7361):454–7. [DOI] [PubMed] [Google Scholar]
- 174.O’Neill SL, Ryan PA, Turley AP, Wilson G, Retzki K, Iturbe-Ormaetxe I, et al. Scaled deployment of Wolbachia to protect the community from dengue and other Aedes transmitted arboviruses. Gates Open Res. 2018;2:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Utarini A, Indriani C, Ahmad RA, Tantowijoyo W, Arguni E, Ansari MR, et al. Efficacy of Wolbachia-Infected Mosquito Deployments for the Control of Dengue. New England Journal of Medicine. 2021;384(23):2177–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Brazil expands implementation of innovative Wolbachia method [press release]. July/3/2023. 2023. [Google Scholar]
- 177.Reich NG, Shrestha S, King AA, Rohani P, Lessler J, Kalayanarooj S, et al. Interactions between serotypes of dengue highlight epidemiological impact of cross-immunity. J R Soc Interface. 2013;10(86):20130414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Katzelnick LC, Zambrana JV, Elizondo D, Collado D, Garcia N, Arguello S, et al. Dengue and Zika virus infections in children elicit cross-reactive protective and enhancing antibodies that persist long term. Sci Transl Med. 2021;13(614):eabg9478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Salje H, Alera MT, Chua MN, Hunsawong T, Ellison D, Srikiatkhachorn A, et al. Evaluation of the extended efficacy of the Dengvaxia vaccine against symptomatic and subclinical dengue infection. Nat Med. 2021;27(8):1395–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Waggoner JJ, Balmaseda A, Gresh L, Sahoo MK, Montoya M, Wang C, et al. Homotypic Dengue Virus Reinfections in Nicaraguan Children. J Infect Dis. 2016;214(7):986–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Chen HL, Lin SR, Liu HF, King CC, Hsieh SC, Wang WK. Evolution of dengue virus type 2 during two consecutive outbreaks with an increase in severity in southern Taiwan in 2001–2002. Am J Trop Med Hyg. 2008;79(4):495–505. [PubMed] [Google Scholar]
- 182.Ko HY, Li YT, Chao DY, Chang YC, Li ZT, Wang M, et al. Inter- and intra-host sequence diversity reveal the emergence of viral variants during an overwintering epidemic caused by dengue virus serotype 2 in southern Taiwan. PLoS Negl Trop Dis. 2018;12(10):e0006827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Moodie Z, Juraska M, Huang Y, Zhuang Y, Fong Y, Carpp LN, et al. Neutralizing Antibody Correlates Analysis of Tetravalent Dengue Vaccine Efficacy Trials in Asia and Latin America. J Infect Dis. 2018;217(5):742–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Huang Y, Moodie Z, Juraska M, Fong Y, Carpp LN, Chambonneau L, et al. Immunobridging efficacy of a tetravalent dengue vaccine against dengue and against hospitalized dengue from children/adolescents to adults in highly endemic countries. Trans R Soc Trop Med Hyg. 2021;115(7):750–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Juraska M, Magaret CA, Shao J, Carpp LN, Fiore-Gartland AJ, Benkeser D, et al. Viral genetic diversity and protective efficacy of a tetravalent dengue vaccine in two phase 3 trials. Proc Natl Acad Sci U S A. 2018;115(36):E8378–E87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Henein S, Adams C, Bonaparte M, Moser JM, Munteanu A, Baric R, de Silva AM. Dengue vaccine breakthrough infections reveal properties of neutralizing antibodies linked to protection. J Clin Invest. 2021;131(13). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.White LJ, Young EF, Stoops MJ, Henein SR, Adams EC, Baric RS, de Silva AM. Defining levels of dengue virus serotype-specific neutralizing antibodies induced by a live attenuated tetravalent dengue vaccine (TAK-003). PLOS Neglected Tropical Diseases. 2021;15(3):e0009258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Clapham HE, Rodriguez-Barraquer I, Azman AS, Althouse BM, Salje H, Gibbons RV, et al. Dengue Virus (DENV) Neutralizing Antibody Kinetics in Children After Symptomatic Primary and Postprimary DENV Infection. J Infect Dis. 2016;213(9):1428–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Hladish TJ, Pearson CAB, Toh KB, Rojas DP, Manrique-Saide P, Vazquez-Prokopec GM, et al. Designing effective control of dengue with combined interventions. Proc Natl Acad Sci U S A. 2020;117(6):3319–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Rodriguez-Barraquer I, Costa F, Nascimento EJM, Nery NJ, Castanha PMS, Sacramento GA, et al. Impact of preexisting dengue immunity on Zika virus emergence in a dengue endemic region. Science. 2019;363(6427):607–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Carvalho MS, Freitas LP, Cruz OG, Brasil P, Bastos LS. Association of past dengue fever epidemics with the risk of Zika microcephaly at the population level in Brazil. Sci Rep. 2020;10(1):1752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Pedroso C, Fischer C, Feldmann M, Sarno M, Luz E, Moreira-Soto A, et al. Cross-Protection of Dengue Virus Infection against Congenital Zika Syndrome, Northeastern Brazil. Emerg Infect Dis. 2019;25(8):1485–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Anderson KB, Gibbons RV, Thomas SJ, Rothman AL, Nisalak A, Berkelman RL, et al. Preexisting Japanese encephalitis virus neutralizing antibodies and increased symptomatic dengue illness in a school-based cohort in Thailand. PLoS Negl Trop Dis. 2011;5(10):e1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Hoke CH, Nisalak A, Sangawhipa N, Jatanasen S, Laorakapongse T, Innis BL, et al. Protection against Japanese encephalitis by inactivated vaccines. N Engl J Med. 1988;319(10):608–14. [DOI] [PubMed] [Google Scholar]
- 195.WHO. Dengue haemorrhagic fever : diagnosis, treatment, prevention and control, 2nd ed. Geneva: 1997. [Google Scholar]
- 196.Halstead SB. Dengue—The Case Definition Dilemma: A Commentary. The Pediatric Infectious Disease Journal. 2007;26(4):291–2. [Google Scholar]
- 197.WHO. Comprehensive Guidelines for Prevention and Control of Dengue and Dengue Haemorrhagic Fever: Revised and Expanded Edition. Comprehensive Guidelines for Prevention and Control of Dengue and Dengue Haemorrhagic Fever: Revised and Expanded Edition. WHO Guidelines New Delhi: WHO; 2011. [Google Scholar]
- 198.Arora SK, Nandan D, Sharma A, Benerjee P, Singh DP. Predictors of severe dengue amongst children as per the revised WHO classification. J Vector Borne Dis. 2021;58(4):329–34. [DOI] [PubMed] [Google Scholar]
- 199.Bodinayake CK, Tillekeratne LG, Nagahawatte A, Devasiri V, Kodikara Arachchi W, Strouse JJ, et al. Evaluation of the WHO 2009 classification for diagnosis of acute dengue in a large cohort of adults and children in Sri Lanka during a dengue-1 epidemic. PLoS Negl Trop Dis. 2018;12(2):e0006258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Ahmad MH, Ibrahim MI, Mohamed Z, Ismail N, Abdullah MA, Shueb RH, Shafei MN. The Sensitivity, Specificity and Accuracy of Warning Signs in Predicting Severe Dengue, the Severe Dengue Prevalence and Its Associated Factors. Int J Environ Res Public Health. 2018;15(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Hadinegoro SR. The revised WHO dengue case classification: does the system need to be modified? Paediatr Int Child Health. 2012;32 Suppl 1:33–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Tomashek KM, Wills B, See Lum LC, Thomas L, Durbin A, Leo YS, et al. Development of standard clinical endpoints for use in dengue interventional trials. PLoS Negl Trop Dis. 2018;12(10):e0006497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Morra ME, Altibi AMA, Iqtadar S, Minh LHN, Elawady SS, Hallab A, et al. Definitions for warning signs and signs of severe dengue according to the WHO 2009 classification: Systematic review of literature. Rev Med Virol. 2018;28(4):e1979. [DOI] [PubMed] [Google Scholar]
- 204.Srikiatkhachorn A, Rothman AL, Gibbons RV, Sittisombut N, Malasit P, Ennis FA, et al. Dengue--how best to classify it. Clin Infect Dis. 2011;53(6):563–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.World Health Organization Western Pacific Regional Office. Dengue clinical management: facilitator’s training manual. WHO WPRO, Manila, Philippines, 2013. https://apps.who.int/iris/handle/10665/207673 Accessed 25 October 2022. [Google Scholar]
- 206.Zhang FC, He JF, Peng J, Tang XP, Qin CF, Lu HZ, et al. [Guidelines for diagnosis and treatment of dengue in China]. Zhonghua Nei Ke Za Zhi. 2018;57(9):642–8. [DOI] [PubMed] [Google Scholar]
- 207.World Health Organization. Regional Office for South-East Asia. (2011). Comprehensive Guideline for Prevention and Control of Dengue and Dengue Haemorrhagic Fever. Revised and expanded edition. WHO Regional Office for South-East Asia. https://apps.who.int/iris/handle/10665/204894 Accessed 25 October 2022. [Google Scholar]
- 208.Sri Lanka Ministry of Health. Guidelines on management of dengue fever & dengue haemorrhagic fever in adults. Colombo, 2012. https://www.epid.gov.lk/web/images/pdf/Publication/guidelines_for_the_management_of_df_and_dhf_in_adults.pdf Accessed 25 October 2022. [Google Scholar]
- 209.Country Office for India, World Health Organization. (2015). National guidelines for clinical management of dengue fever. WHO Country Office for India. https://apps.who.int/iris/handle/10665/208893 Accessed 25 October 2022. [Google Scholar]
- 210.Hunsperger EA, Munoz-Jordan J, Beltran M, Colon C, Carrion J, Vazquez J, et al. Performance of Dengue Diagnostic Tests in a Single-Specimen Diagnostic Algorithm. J Infect Dis. 2016;214(6):836–44. [DOI] [PubMed] [Google Scholar]
- 211.Chaloemwong J, Tantiworawit A, Rattanathammethee T, Hantrakool S, Chai-Adisaksopha C, Rattarittamrong E, Norasetthada L. Useful clinical features and hematological parameters for the diagnosis of dengue infection in patients with acute febrile illness: a retrospective study. BMC Hematol. 2018;18:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Dussart P, Duong V, Bleakley K, Fortas C, Lorn Try P, Kim KS, et al. Comparison of dengue case classification schemes and evaluation of biological changes in different dengue clinical patterns in a longitudinal follow-up of hospitalized children in Cambodia. PLoS Negl Trop Dis. 2020;14(9):e0008603. [DOI] [PMC free article] [PubMed] [Google Scholar]
