Dengue Medscape

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 36

8/26/2019 https://emedicine.medscape.

com/article/215840-print

emedicine.medscape.com

Dengue
Updated: May 03, 2019
Author: Darvin Scott Smith, MD, MSc, DTM&H; Chief Editor: Michael Stuart Bronze, MD

Overview

Practice Essentials
Dengue is the most common and important arthropod-borne viral (arboviral) illness in humans. It is transmitted by mosquitoes of
the genus Aedes, which are widely distributed in subtropical and tropical areas of the world (see the image below). The
incidence of dengue has increased dramatically in recent decades, with estimates of 40%-50% of the world’s population at risk
for the disease in tropical, subtropical, and, most recently, more temperate areas.[1]

Drawing of Aedes aegypti mosquito. Courtesy of the Centers for Disease Control and Prevention (CDC).

A small percentage of persons who have previously been infected by one dengue serotype develop bleeding and endothelial
leak upon infection with another dengue serotype. This syndrome is termed severe dengue (also known as dengue hemorrhagic
fever and dengue shock syndrome).

Dengue fever is typically a self-limited disease with a mortality rate of less than 1% when detected early and with access to
proper medical care. When treated, severe dengue has a mortality rate of 2%-5%, but, when left untreated, the mortality rate is
as high as 20%.

See 7 Bug Bites You Need to Know This Summer, a Critical Images slideshow, for helpful images and information on various
bug bites.

Signs and symptoms


https://emedicine.medscape.com/article/215840-print 1/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
On average, dengue becomes symptomatic after a 4- to 10-day incubation period (range, 3-14 days). Dengue symptoms usually
last 2-7 days.

Many individuals with dengue may be asymptomatic. Many patients with dengue experience a prodrome of chills; rash, including
erythematous mottling of the skin; and facial flushing, which may last 2-3 days. Children younger than 15 years who have
dengue usually have a nonspecific febrile syndrome, which may be accompanied by a maculopapular rash. Dengue should be
suspected in individuals who present with high fever (104°F/40°C), retro-orbital headache, muscle and joint pain, nausea,
lymphadenopathy, vomiting, and rash and who have traveled within 2 weeks of symptom onset to an area where appropriate
vectors are present and dengue transmission may be occurring.

Accompanying symptoms in patients with dengue may include any of the following:

Fever
Headache
Retro-orbital pain
Severe myalgias: Especially of the lower back, arms, and legs
Arthralgias: Usually of the knees and shoulders
Nausea and vomiting (diarrhea is rare)
Rash: A maculopapular or macular confluent rash over the face, thorax, and flexor surfaces, with islands of skin sparing
Weakness, malaise, and lethargy
Altered taste sensation
Anorexia
Sore throat
Mild hemorrhagic manifestations (eg, petechiae, bleeding gums, epistaxis, menorrhagia, hematuria)
Lymphadenopathy

Severe dengue (dengue hemorrhagic fever and dengue shock syndrome)

The initial phase of severe dengue is similar to that of dengue fever and other febrile viral illnesses. Shortly after the fever
breaks (3-7 days after symptom onset or sometimes within 24 hours before), signs of plasma leakage appear, along with the
development of hemorrhagic symptoms such as bleeding from sites of trauma, gastrointestinal bleeding, and hematuria.
Patients may also present with severe abdominal pain, persistent vomiting that may contain blood, fatigue, and febrile seizures
(in children).

The subsequent 24 hours frequently prove critical. If left untreated, hemorrhagic fever most likely progresses to shock. Common
symptoms in impending shock include abdominal pain, vomiting, and restlessness. Patients also may have symptoms related to
circulatory failure, such as pallor, tachypnea, tachycardia, dizziness/lightheadedness, and a decreased level of consciousness.

See Clinical Presentation for more detail.

Diagnosis

Laboratory criteria for the diagnosis of dengue include one or more of the following, which are used to detect the virus, viral
nucleic acid, antibodies or antigens, or a combination thereof:

Demonstration of a fourfold or greater change in reciprocal immunoglobulin G (IgG) or IgM antibody titers to 1 or more
dengue virus antigens in paired serum samples
Demonstration of dengue virus antigen in autopsy tissue via immunohistochemistry or immunofluorescence or in serum
samples via enzyme immunoassay (MAC-ELISA, IgG ELISA, nonstructural protein 1 [NS1] ELISA, EIA)
Detection of viral genomic sequences in autopsy tissue, serum, or cerebral spinal fluid (CSF) samples via reverse-
transcriptase polymerase chain reaction (RT-PCR) assay: RT-PCR provides earlier and more specific diagnosis.
Less frequently, isolation of the dengue virus from serum, plasma, leukocytes, or autopsy samples

During the early phase of the disease (first 4-5 days), virus can be detected in serum, plasma, circulating blood cells, and
tissues. Virus isolation, nucleic acid detection, and antigen detection are more useful to diagnose infection. At the end of the
acute phase of illness, serology becomes the method of choice.

The following laboratory tests should also be performed in the workup of patients with possible dengue:

Complete blood cell (CBC) count


Metabolic panel
Serum protein and albumin levels
Liver panel
Coagulation panel with or without disseminated intravascular coagulation (DIC) panel

Characteristic laboratory findings in dengue are as follows:

https://emedicine.medscape.com/article/215840-print 2/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Thrombocytopenia (platelet count < 100 x 109/L)


Leukopenia
Mild to moderate elevation of aspartate aminotransferase and alanine aminotransferase values

In patients with severe dengue, the following may be present:

Increased hematocrit level secondary to plasma extravasation and/or third-space fluid loss
Hypoproteinemia
Prolonged prothrombin time
Prolonged activated partial thromboplastin time
Decreased fibrinogen
Increased amount of fibrin split products

Guaiac testing for occult blood in the stool should be performed on all patients in whom dengue virus infection is suspected.
Urinalysis identifies hematuria.

Imaging studies include the following:

Chest radiography
Head computed tomography (CT) scanning without contrast: To detect intracranial bleeding or cerebral edema due to
severe dengue
Ultrasonography: To detect fluid in the chest and abdominal cavities, pericardial effusion, and a thickened gallbladder
wall in patients with severe dengue

See Workup for more detail.

Management
Oral rehydration therapy is recommended for patients with moderate dehydration caused by high fever and vomiting.

Patients who develop signs of severe dengue warrant closer observation. Admission for close volume status monitoring and
intravenous fluid administration is indicated for patients who develop signs of dehydration, such as the following:

Tachycardia
Prolonged capillary refill time
Cool or mottled skin
Diminished pulse amplitude
Altered mental status
Decreased urine output
Rising hematocrit
Narrowed pulse pressure
Hypotension

Patients with internal or gastrointestinal bleeding may require transfusion, and patients with coagulopathy may require fresh
frozen plasma.

See Treatment and Medication for more detail.

Background
Dengue is the most common and important arthropod-borne viral (arboviral) illness in humans. Globally, 2.5-3 billion individuals
live in approximately 112 countries that experience dengue transmission. While the annual incidence is unclear owing to
incomplete global reporting and misclassification of illness, approximately 3.2 million individuals were infected globally in 2015. It
is caused by infection with 1 of the 4 serotypes of dengue virus, which is a Flavivirus (a genus of single-stranded nonsegmented
RNA viruses). Infection with one dengue serotype confers lifelong homotypic immunity to that serotype and a brief period
(approximately 2 years) of partial heterotypic immunity to other serotypes, but an individual can eventually be infected by all 4
serotypes. Several serotypes can be in circulation during an epidemic.

Dengue is transmitted by mosquitoes of the genus Aedes, which are widely distributed in subtropical and tropical areas of the
world (see the image below). An individual with dengue is capable of transmitting the virus for 4-5 days (maximum, 12 days) to a
capable vector. After an incubation period of 5-10 days, the infected mosquito can transmit virus for the rest of its life span (2

https://emedicine.medscape.com/article/215840-print 3/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
weeks to 1 month). Aedes albopictus is more cold tolerant than Aedes aegypti, so it can survive and transmit virus in the more
temperate regions of the United States and Europe.

The global incidence of dengue has increased dramatically in the last several decades, with an estimated 40%-50% of the
world’s population in 128 countries at risk.[2, 3, 4] Today, severe dengue largely affects Asian and Latin American countries,
where it is a leading cause of hospitalization and death. The World Health Organization (WHO) ranked dengue as one of the top
ten threats to global health in 2019.[5]

Initial dengue infection may be asymptomatic (50%-90%),[6] may result in a nonspecific febrile illness, or may produce the
symptom complex of classic dengue fever (DF). Classic dengue fever is marked by rapid onset of high fever, headache, retro-
orbital pain, diffuse body pain (both muscle and bone), weakness, vomiting, sore throat, altered taste sensation, and a
centrifugal maculopapular rash, among other manifestations. The severity of the pain led to the term breakbone fever to
describe dengue.

A small percentage of persons who have previously been infected by one dengue serotype develop bleeding and endothelial
leak upon infection with another dengue serotype. This syndrome is termed severe dengue (reclassified in 2009 by the WHO,
previously referred to as dengue hemorrhagic fever and dengue shock syndrome).

Severe dengue has also been termed dengue vasculopathy. Vascular leakage in these patients results in hemoconcentration
and serous effusions and can lead to circulatory collapse. This, in conjunction with severe hemorrhagic complications, can lead
to a shock syndrome, which poses a greater fatality risk than bleeding per se.[7]

Dengue virus transmission follows 2 general patterns: epidemic dengue and hyperendemic dengue. Epidemic dengue
transmission occurs when dengue virus is introduced into a region as an isolated event that involves a single viral strain. If the
number of vectors and susceptible pediatric and adult hosts is sufficient, explosive transmission can occur, with an infection
incidence of 25-50%. Mosquito-control efforts, changes in weather, and herd immunity contribute to the control of these
epidemics. Transmission appears to begin in urban centers and then spreads to the rest of the country.[8] This is the current
pattern of transmission in parts of Africa and South America, areas of Asia where the virus has reemerged, and small island
nations. Travelers to these areas are at increased risk of acquiring dengue during these periods of epidemic transmission.

Hyperendemic dengue transmission is characterized by the continuous circulation of multiple viral serotypes in an area where a
large pool of susceptible hosts and a competent vector (with or without seasonal variation) are constantly present. This is the
predominant pattern of global transmission. In areas of hyperendemic dengue, antibody prevalence increases with age, and
most adults are immune. Hyperendemic transmission appears to be a major risk for dengue hemorrhagic fever. Travelers to
these areas are more likely to be infected than are travelers to areas that experience only epidemic transmission.[9]

Because the signs and symptoms of dengue fever are nonspecific, attempting laboratory confirmation of dengue infection by
serodiagnosis, reverse-transcriptase polymerase chain reaction (RT-PCR), or culture is important. Serodiagnosis is made on the
basis of a rise in antibody titer in paired IgG or IgM specimens. Results vary depending on whether the infection is primary or
secondary (see Presentation and Workup). Dengue is a reportable disease in the United States; known or suspected cases
should be reported to public health authorities.

Dengue fever is usually a self-limited illness. Supportive care with analgesics, judicious fluid replacement, and bed rest is
usually sufficient. Successful management of severe dengue requires intravascular volume replacement, with careful attention
to fluid management and proactive treatment of hemorrhage. Admission to an intensive care unit is indicated for patients with
severe dengue (see Treatment).

Historical background

The earliest known documentation of dengue fever–like illness was in the Chinese Encyclopedia of Symptoms during the Chin
Dynasty (CE 265-420). The illness was called "the water poison" and was associated with flying insects near water.

Earliest recorded outbreaks

Outbreaks of febrile illnesses compatible with dengue fever have been recorded throughout history, with the first epidemic
described in 1635 in the West Indies.

In 1779-1780, the first confirmed, reported outbreak of dengue fever occurred almost simultaneously in Asia, North America,
and Africa. In 1789, the American physician Benjamin Rush published an account of a probable dengue fever epidemic that had
occurred in Philadelphia in 1780. Rush coined the term breakbone fever to describe the intense symptoms reported by one of
his patients.

A denguelike epidemic in East Africa in the early 1820s was called, in Swahili, ki denga pepo ("it is a sudden overtaking by a
spirit"). The English version of this term, “Dandy fever,” was applied to an 1827-28 Caribbean outbreak, and in the Spanish
Caribbean colonies, that term was altered to “dengue.”

Increased distribution after World War II


https://emedicine.medscape.com/article/215840-print 4/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
Probable outbreaks of dengue fever occurred sporadically every 10-30 years until after World War II. The socioeconomic
disruptions caused by World War II resulted in increased worldwide spread of dengue viruses and capable vectors. The first
epidemic of dengue hemorrhagic fever in the modern era was described in Manila in 1953. After that, outbreaks of dengue fever
became more common.

A pattern developed in which dengue fever epidemics occurred with increasing frequency and were associated with occasional
dengue hemorrhagic fever cases. Subsequently, dengue hemorrhagic fever epidemics occurred every few years. Eventually,
dengue hemorrhagic fever epidemics occurred yearly, with major outbreaks occurring approximately every 3 years. This pattern
has repeated itself as dengue fever has spread to new regions.

Although initial epidemics were located in urban areas, increased dengue spread has involved suburban and rural locales in
Asia and Latin America. The only continents that do not experience dengue transmission are Europe and Antarctica. In the
1950s, 9 countries reported dengue outbreaks; currently, the geographic distribution includes more than 100 countries
worldwide. Several of these countries had not previously reported dengue, and many had not reported dengue in 20 years.

Dengue transmission spread from Southeast Asia into surrounding subtropical and tropical Asian countries, southern China and
southern Taiwan, the Indian subcontinent and Sri Lanka, and down the island nations of Malaysia, the Philippines, New Guinea,
northeastern Australia, and several Pacific islands, including Tahiti, Palau, Tonga, and the Cook Islands. Hyperendemic
transmission is reported in Vietnam, Thailand, Indonesia, Pakistan, India, Malaysia, and the Philippines. Dengue continues to
extend its range.

In the Americas, dengue epidemics were rare post war because Aedes mosquitoes had been eradicated from most of the region
through coordinated vector-control efforts. Systematic spraying was halted in the early 1970s because of environmental
concerns. By the 1990s, A aegypti mosquitoes repopulated most of the countries in which they had been eliminated.

In 2014, increased cases of dengue were reported to the WHO in the Peoples Republic of China, Cook Island, Fiji, Malaysia,
and Vanuatu, which experienced an outbreak of dengue serotype 3 (DENV-3) after a 10-year hiatus. In 2015, large outbreaks of
dengue were reported in the Philippines (>169,000 cases), Malaysia (>111,000 suspected cases), and Brazil (>1.5 million
cases). Delhi, India, experienced its worse outbreak since 2006.

DENV-1 and DENV-2

Serotype 1 dengue (DENV-1) was first reported in French Polynesia and Japan in 1943, followed by Hawaii.[10] DENV-1 was
introduced into a largely susceptible population in Cuba in 1977. Serosurveys indicated that more than 44% of the population
was infected, with only mild disease reported. The first dengue hemorrhagic fever epidemic in the Americas occurred in Cuba in
1981 and involved serotype 2 dengue (DENV-2), with hundreds of thousands of cases of dengue in both children and adults,
24,000 cases of dengue hemorrhagic fever, 10,000 cases of dengue shock syndrome, and 158 reported deaths.

In 1997, Asian genotype DENV-2 was reintroduced, and dengue shock syndrome and dengue hemorrhagic fever were seen
only in adults who had previously been infected with DENV-1 in 1977. Disease and case-fatality rates were higher in those who
had been infected with DENV-2 20 years after their initial DENV-1 infection than those who were infected 4 years apart.

Data from other countries supports the finding that the severity of secondary dengue infections appears to intensify with longer
intervals between infections.[11, 12] Since then, dengue fever and dengue hemorrhagic fever cases have progressively
increased.

United States

In 1986, the first clearly identified local transmission of dengue in the United States occurred in Texas. Carriers of the virus were
believed to have crossed the border from Mexico; the local vector population was then infected. Since then, seasonal
autochthonous infection has been reported in both Texas and Hawaii.

In 2001-2002, Hawaii experienced its first outbreak of dengue since World War II ended. The outbreak involved 2 variants of
DENV-1 that were transmitted by A albopictus. Predominantly affecting young adults and adults, 122 cases of dengue fever
spread slowly on Maui, Oahu, and Kauai. The epidemic was traced to viremic visitors from Tahiti, which was then experiencing a
severe outbreak of the infection. In 2015, Hawaii reported more than 65,000 cases, with ongoing transmission reported in 2016.

Two competent vectors, A aegypti and A albopictus, are currently seasonally abundant in some areas of the southwestern and
southeastern United States, including Texas, Arizona, New Mexico, Louisiana, Mississippi, Alabama, Georgia, and mid to south
Florida. A aegypti has also been reported sporadically in portions of North Carolina, South Carolina, Tennessee, Arkansas,
Maryland, and New Jersey. The range of A albopictus extends almost as far north as the Great Lakes.

As suggested by a reported case of a woman aged 63 years who died from complications of dengue acquired in New Mexico or
Texas in 2012, the disease may not be adequately recognized in the United States as a source of potentially fatal acute febrile
illness. The patient had initially been diagnosed with West Nile virus, but a postmortem bone marrow biopsy revealed the
presence of dengue virus.[13, 14]

https://emedicine.medscape.com/article/215840-print 5/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
In addition, the patient’s records revealed that she met the clinical case definition for hemophagocytic lymphohistiocytosis, a
hyperinflammatory syndrome that is sometimes associated with dengue and that, in this instance, was the cause of death.

Europe

Dengue fever did not naturally occur in the European Union and in continental Europe because these areas did not have an
appropriate vector population to allow further spread of dengue from viremic individuals returning from other countries. However,
dengue does occur in several overseas territories of European Union members. In recent decades, reports of dengue infections
in long-term expatriates, aid workers, military personnel, immigrants, and travelers returning from the tropics and subtropics
have been increasing. In 2010, local transmission was reported in France and Croatia. Another outbreak with more than 2000
cases occurred in Madeira in 2012.

Factors believed to be responsible for the spread of dengue include the following:

Explosive population growth


Unplanned urban overpopulation with inadequate public health systems
Poor control of standing water and vectors
Viral evolution
Increased international recreational, business, and military travel to endemic areas

All of these factors must be addressed to control the spread of dengue and other mosquito-borne infections. Unplanned
urbanization is believed to have had the largest impact on disease amplification in individual countries, whereas travel is
believed to have had the largest impact on global spread.[6, 8, 9, 12, 15]

Liu-Helmersson et al (2016) found that, over time, the duration and intensity of dengue transmission in Europe have increased.
With increasing levels of greenhouse gas emissions, the authors predict that changes in intensity and duration will occur more
rapidly.[16]

Travel surveillance

Over the past decades, the GeoSentinel Network of Travel Medicine providers has demonstrated that dengue has become more
frequently diagnosed than malaria in travelers returning from tropical areas other than Africa. Such sentinel travel surveillance
can augment global and national public health surveillance. More recent studies have not supported an earlier suggestion that
climate change is also directly responsible for increased transmission.[11, 9, 12]

Pathophysiology
Dengue fever is a mosquito-borne viral disease caused by 1 of 4 closely related but antigenically distinct serotypes of dengue
virus, serotypes DENV-1 through DEN-4.[17] Infection with one dengue serotype confers lifelong homotypic immunity and a brief
period of partial heterotypic immunity (2 years), but each individual can eventually be infected by all 4 serotypes. Several
serotypes can be in circulation during an epidemic.

The Aedes mosquito

Dengue viruses are transmitted by the bite of an infected female Aedes (subgenus Stegomyia) mosquito.[18] Both males and
females require nectar for energy. Females require a blood meal as a source of appropriate protein for egg development.
Globally, Aedes aegypti is the predominant highly efficient mosquito vector for dengue infection, but the Asian tiger mosquito,
Aedes albopictus, and other Aedes species can also transmit dengue with varying degrees of efficiency (see the images below).

https://emedicine.medscape.com/article/215840-print 6/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Drawing of Aedes aegypti mosquito. Courtesy of the Centers for Disease Control and Prevention (CDC).

Aedes albopictus. Courtesy of the Centers for Disease Control and Prevention (CDC).

https://emedicine.medscape.com/article/215840-print 7/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Aedes aegypti mosquito. Courtesy of Wikimedia Commons (https://commons.wikimedia.org/wiki/File:Aedes_aegypti.jpg;


author Muhammad Mahdi Karim).

Aedes mosquito species have adapted well to human habitation, often breeding around dwellings in small amounts of stagnant
water found in old tires or other small containers discarded by humans. Even a bottle cap filled with water can serve as to
incubate and hatch Aedes eggs. Eggs can survive periods of drying and will hatch when exposed to water. Humans are the
preferred hosts.

Female Aedes mosquitoes are daytime feeders. They inflict an innocuous bite, usually on the back of the neck and the ankles,
and are easily disturbed during a blood meal, causing them to move on to finish a meal on another individual, making them
efficient vectors. Not uncommonly, entire families develop infection within a 24- to 36-hour period, presumably from the bites of a
single infected mosquito.

Hosts for transmission

Humans serve as the primary reservoir for dengue. Certain nonhuman primates in Africa and Asia also serve as hosts but do
not develop dengue hemorrhagic fever. Mosquitoes acquire the virus when they feed on a carrier of the virus. Persons with
dengue viruses in their blood can transmit the viruses to the mosquito 1 day before the onset of the febrile period. The patient
usually remains infectious for the subsequent 4-5 days (up to 12 days).

The mosquito can transmit dengue if it immediately bites another host. In addition, transmission occurs after 8-12 days of viral
replication in the mosquito's salivary glands (extrinsic incubation period). The virus does not adversely affect the mosquito. The
mosquito remains infected for the remainder of its life. The life span of A aegypti is usually 21 days but ranges from 15 to 65
days. Vertical transmission of dengue virus in mosquitoes has been documented.[19] The eggs of Aedes mosquitoes withstand
long periods of desiccation, reportedly as long as 1 year, but are killed by temperatures of less than 10°C. Rare cases of vertical
dengue transmission have been reported. In addition, rare reports of human-to-human transmission via needle-stick injuries
have been published.[20]

Once inoculated into a human host, dengue has an incubation period of 3-14 days (average 4-7 days) while viral replication
takes place in target dendritic cells. Infection of target cells, primarily those of the reticuloendothelial system, such as dendritic
cells, macrophages, hepatocytes, and endothelial cells,[21, 22, 23, 24] result in the production of immune mediators that serve
to shape the quantity, type, and duration of cellular and humoral immune response to both the initial and subsequent virus
infections.[21, 25, 26, 27, 28, 29, 30]

Dengue viral infections frequently are not apparent. In most cases, especially in children younger than 15 years, the patient is
asymptomatic or has a mild undifferentiated febrile illness lasting 5-7 days. Classic dengue fever primarily occurs in nonimmune,
nonindigenous adults and children and is typically self-limiting. Recovery is usually complete by 7-10 days. Severe dengue
(dengue hemorrhagic fever/dengue shock syndrome) usually occur around the third to seventh day of illness during a second
dengue infection in persons with preexisting actively or passively (maternally) acquired immunity to a heterologous dengue virus
serotype.

Dengue fever

Dengue presents in a nonspecific manner similarly to that of many other viral and bacterial illnesses. Fever typically begins on
the third day of illness and persists 5-7 days, abating with the cessation of viremia. Fever may reach 41C°. Occasionally, and
more frequently in children, the fever abates for a day and recurs, a pattern that is termed a saddleback fever; however, this
pattern is more commonly seen in dengue hemorrhagic fever.

Leukopenia, lymphopenia near the end of the febrile phase, and thrombocytopenia are common findings in dengue fever and
are believed to be caused by direct destructive actions of the virus on bone marrow precursor cells. The resulting active viral
replication and cellular destruction in the bone marrow are believed to cause the bone pain. Approximately one third of patients
with dengue fever may have mild hemorrhagic symptoms, including petechiae, gingival bleeding, and a positive tourniquet test
(>20 petechiae in an area of 2.5 X 2.5 cm). Dengue fever is rarely fatal.

Severe dengue (dengue hemorrhagic fever)


Severe dengue occurs less frequently than dengue fever but has a more dramatic clinical presentation. In most of Asia, where it
first was described, severe dengue is primarily a disease of children. However, in the Americas, and more recently reported in
Taiwan, severe dengue has an equal distribution in all ages.

Severe dengue typically begins with the initial manifestations of dengue fever. The acute febrile illness (temperatures ≤40°C),
like that of dengue fever, lasts approximately 2-7 days. However, in persons with severe dengue, the fever reappears, giving a
biphasic or saddleback fever curve.

https://emedicine.medscape.com/article/215840-print 8/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Along with biphasic fever, patients with severe dengue have progressive thrombocytopenia, increasing hematocrit (20%
absolute rise from baseline) and low albumin (signs of hemoconcentration preceding shock), more obvious hemorrhagic
manifestations (>50% of patients have a positive tourniquet test), and progressive effusions (pleural or peritoneal).
Lymphocytosis, often with atypical lymphocytes, commonly develops before defervescence or the onset of shock. Transaminase
levels may be mildly elevated or present in the several thousands associated with hepatomegaly in those patients with acute
hepatitis. Low fibrinogen and elevated fibrin split products are signs of disseminated intravascular coagulation. Severe metabolic
acidosis and circulatory failure can occur.

The critical feature of severe dengue is plasma leakage. Plasma leakage is caused by increased capillary permeability and may
manifest as hemoconcentration, as well as pleural effusion and ascites. Bleeding is caused by capillary fragility and
thrombocytopenia and may manifest in various forms, ranging from petechial skin hemorrhages to life-threatening
gastrointestinal bleeding.

Liver damage manifests as increases in levels of alanine aminotransferase and aspartate aminotransferase, low albumin levels,
and deranged coagulation parameters (prothrombin time, partial thromboplastin time).[31, 32] In persons with fatal dengue
hepatitis, infection was demonstrated in more than 90% of hepatocytes and Kupffer cells with minimal cytokine response (tumor
necrosis factor [TNF]–alpha, interleukin [IL]–2). This is similar to that seen with fatal yellow fever and Ebola infections.[31]

As the term implies, severe dengue shock is essentially dengue hemorrhagic fever with progression into circulatory failure, with
ensuing hypotension, narrow pulse pressure (< 20 mm Hg), and, ultimately, shock and death if left untreated. Death may occur
8-24 hours after onset of signs of circulatory failure. The most common clinical findings in impending shock include hypothermia,
abdominal pain, vomiting, and restlessness.

Secondary infection

The immunopathology of severe dengue remains incompletely understood. Most patients who develop severe dengue have had
prior infection with one or more dengue serotypes. When an individual is infected with another serotype (ie, secondary infection)
and produces low levels of nonneutralizing antibodies, these antibodies, directed against 1 of 2 surface proteins (precursor
membrane protein and envelope protein), when bound by macrophage and monocyte Fc receptors, have been proposed to fail
to neutralize virus and instead form an antigen-antibody complex.

This results in increased viral entry into macrophages bearing IgG receptors, allowing unchecked viral replication with higher
viral titers and increased cytokine production and complement activation, a phenomenon called antibody-dependent
enhancement.[33, 34]

The affected macrophages release vasoactive mediators that increase vascular permeability, leading to vascular leakage,
hypovolemia, and shock. This mechanism, along with individual host and viral genome variations, plays an active role in
pathogenesis. Infants born to mothers who have had dengue, as maternally derived dengue neutralizing IgGs wane, are also
thought to be at risk for enhanced disease.[33, 34]

Some researchers suggest that T-cell immunopathology may play a role, with increased T-cell activation and apoptosis.
Increased concentrations of interferon have been recorded 1-2 days following fever onset during symptomatic secondary
dengue infections.[35] The activation of cytokines, including TNF-alpha, TNF receptors, soluble CD8, and soluble IL-2 receptors,
has been correlated with disease severity.[21]

Cuban studies have shown that stored serum sample analysis demonstrated progressive loss of cross-reactive neutralizing
antibodies to DENV-2 as the interval since DENV-1 infection increased.[28] In addition, certain dengue strains, particularly those
of DENV-2, have been proposed to be more virulent, in part because more epidemics of dengue hemorrhagic fever have been
associated with DENV-2 than with the other serotypes.

DENV-2–activated platelets were phagocytized in large numbers when the platelet activation inhibitor prostacyclin was added.
[36]

Several recent studies have investigated the causes of thrombocytopenia in dengue. Laboratory and human studies have
suggested a direct correlation between activation and depletion of platelets, with a sharp drop occurring on day 4 of fever. A
high number of dengue virus genome copies have been found in these activated platelets. Increased binding of complement C3
and IgG have also been found on the surface of these platelets. In addition to platelet activation, dengue infection has been
found to activate the intrinsic pathway of apoptosis, with increased surface phosphatidylserine exposure, mitochondrial
depletion, and activation of caspase 3 and 9.[37]

Etiology
https://emedicine.medscape.com/article/215840-print 9/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Dengue infection is caused by dengue virus (DENV), which is a single-stranded RNA virus (approximately 11 kilobases long)
with an icosahedral nucleocapsid and covered by a lipid envelope. The virus is in the family Flaviviridae, genus Flavivirus, and
the type-specific virus is yellow fever.

The dengue virus has 4 related but antigenically distinct serotypes: DENV-1, DENV-2, DENV-3, and DENV-4. Genetic studies of
sylvatic strains suggest that the 4 serotypes evolved from a common ancestor in primate populations approximately 1000 years
ago and that all 4 separately emerged into a human urban transmission cycle 500 years ago in either Asia or Africa.[6, 38]
Albert Sabin speciated these viruses in 1944. Each serotype is known to have several different genotypes. Viral genotype and
serotype, and the sequence of infection with different serotypes, appear to affect disease severity.

Living in endemic areas of the tropics (or warm, moist climates such as the southern United States) where the vector mosquito
thrives is an important risk factor for infection.[17, 39, 40, 41, 42] Poorly planned urbanization combined with explosive global
population growth brings the mosquito and the human host into close proximity. Increased air travel easily transports infectious
diseases between populations.

Epidemiology
United States statistics

In the United States, of the 100-200 reported cases per year, dengue occurs principally in travelers returning from endemic
transmission areas. During 2006–2008, an average of 244 confirmed and probable travel-associated dengue cases were
reported in the United States, according to the US Centers for Disease Control and Prevention (CDC).[43] The CDC reports that
cases of dengue in returning US travelers have increased steadily during the past 20 years, and dengue has become the
leading cause of acute febrile illness in US travelers returning from the Caribbean, South America, and Asia.[44]

Dengue was once epidemic in the southeastern United States, and the potential exists for its reemergence. The principal
mosquito vector for dengue, A aegypti, is found in the southern and southeastern United States, along with A albopictus, a less
efficient vector species introduced in 1985. A aegypti breeds year-round in southern Florida.

Most dengue cases in US citizens occur in Puerto Rico, the US Virgin Islands, Guam, and Samoa. Puerto Rico has experienced
several seasonal outbreaks since 2015, with more significant transmission occurring from August to November. The last dengue
epidemic in Florida (in the Tampa and Miami areas) occurred in 1934-1935 and affected an estimated 15,000 people of the
population of 135,000 in Miami. The last recorded epidemic in the southeastern United States occurred in Louisiana in 1945.
Outbreaks of dengue also occurred in Laredo, Texas, in 1998. Dengue reemerged in Florida in 2009-2010, however, with 27
locally acquired cases in Key West.[44] The index case in this outbreak was diagnosed after returning home to New York from a
visit to Key West. This illustrates the importance of awareness of dengue among physicians outside endemic areas. In 2013, 23
cases were reported in Martin County, Florida. During that year, 120 cases were imported into Florida from various countries.
[45] Since January 2010, dengue has been a reportable disease in the United States.[44]

International statistics

The overall incidence of dengue, as well as the explosive outbreaks of dengue, has been increasing dramatically over the last
several years. Older data suggested an estimated 50-100 million cases of dengue fever and 500,000 cases of dengue
hemorrhagic fever occur worldwide, with 22,000 deaths (mainly in children).[46, 47, 48] In 2015, official data from WHO member
states reported more than 3.2 million cases, with 2.35 million cases in the Americas alone, including 10,200 cases of severe
dengue and 1181 deaths. One study estimates that approximately 390 million dengue infections occur per year (95% CI; 284-
528 million), with 96 million of these presenting clinically.[49] An estimated 2.5-3 billion people (approximately 40%-50% of the
world’s population) are estimated to be at risk for dengue infection. Recent estimates find that 128 countries worldwide are at
risk for dengue infection, which includes 36 that had once been classified as dengue-free.[50] The only continent that has not
experienced dengue transmission is Antarctica.

https://emedicine.medscape.com/article/215840-print 10/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Global map of dengue risk. Frequent or continuous risk = frequent outbreaks or ongoing transmission. Sporadic or uncertain
risk = variable and unpredictable risk, country-level data are unavailable. Courtesy of the Centers for Disease Control and
Prevention (CDC) (https://www.cdc.gov/dengue/areaswithrisk/around-the-world.html).

Worldwide distribution of dengue in 2000. Courtesy of the Centers for Disease Control and Prevention (CDC).

https://emedicine.medscape.com/article/215840-print 11/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Worldwide distribution of dengue in 2003. Courtesy of the Centers for Disease Control and Prevention (CDC).

Worldwide distribution of dengue in 2005. Courtesy of the Centers for Disease Control and Prevention (CDC).

According to the World Health Organization, dengue ranks as the most important mosquito-borne viral disease in the world. In
the last 50 years, the incidence of dengue has increased 30-fold worldwide.[48] In the Americas alone, the incidence rose from
250,000 cases of dengue fever and 7,000 cases of dengue hemorrhagic fever in 1995 to more than 890,000 cases of dengue
fever and 26,000 cases of dengue hemorrhagic fever in 2007.

The world's largest known epidemic of dengue occurred in Cuba in 1981, with more than 116,000 persons hospitalized and as
many as 11,000 cases reported in a single day. Current outbreaks can be monitored via the ProMed listserve by contacting
owner-promed@promedmail.org.

Since 2000, at least 8 areas previously without dengue have reported outbreaks, including Nepal, Bhutan, Macau, Hong Kong,
Taiwan,[51] Madagascar, the Galapagos, and Easter Island. The Pan American Health Organization (PAHO) reported that 2007
saw the highest number of dengue fever and dengue hemorrhagic fever cases (918,495) in the Americas since 1985.

Southeast Asia

Currently, dengue hemorrhagic fever is one of the leading causes of hospitalization and death in children in many Southeast
Asian countries, with Indonesia reporting the majority of dengue hemorrhagic fever cases. Of interest and significance in
prevention and control, 3 surveillance studies in Asia report an increasing age among infected patients and increasing mortality
rate.

A 5-year prospective study in Thai children examined the relative economic burden of dengue infection in children on the local
population. Most disability-adjusted life years (DALYs) lost to dengue resulted from long-duration illness in children who had not
been hospitalized. The infecting serotype appeared to be a determining factor of DALYs lost, with DENV-2 and DENV-3

https://emedicine.medscape.com/article/215840-print 12/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
responsible for 30% and 29%, respectively. The mean cost of illness from dengue was significantly higher than that from other
febrile illnesses.[52]

Since 1982 in Singapore, more than 50% of deaths have occurred in individuals older than 15 years. In Indonesia, young adults
in Jakarta and provincial areas make up a larger percentage of infected patients. During the 2000 epidemic in Bangladesh, up to
82% of hospitalized patients were adults, and all deaths occurred in patients older than 5 years.

Africa

The epidemiology of dengue fever in Africa is more poorly characterized. Aedes aegypti is present in a large portion of the
Middle East and sub-Saharan Africa. Dengue fever is present in 19 countries on the African continent. In a 1993 epidemic in the
Comoros, an estimated 60,000 persons were infected with dengue. Of note, no major dengue hemorrhagic fever epidemics
have occurred in Africa, despite the fact that all 4 dengue serotypes circulate in the continent. This may be explained by a
genetic factor in these populations.

South America

Hyperendemic circulation of all 4 dengue serotypes is present in the northern countries of South America. Brazil (700,000 cases
in 2002), Colombia, and Venezuela report the most cases of dengue and dengue hemorrhagic fever, with low-level transmission
occurring year-round but with most occurring during periods of epidemic transmission. Since the 1970s, outbreaks of dengue
fever have increased in frequency and severity in the Caribbean. Significant outbreaks of dengue have been reported in 2005
and 2006 in Puerto Rico, the US Virgin Islands, the Dominican Republic, Barbados, Curacao, Cuba, Guadeloupe, and
Martinique.

Race-, sex-, and age-related demographics

The distribution of dengue is geographically determined. Dengue affects all races. Some African and Haitian data demonstrate a
relative dearth of dengue hemorrhagic fever and dengue shock syndrome during dengue fever epidemics, suggesting that these
populations may share a genetic advantage to the virus. This merits further study.

A study of dengue incidence in six different Asian countries reported a higher incidence in males than in females. The authors
speculated that the difference may result from different gender roles and subsequent differences in exposure risks.[53]

Dengue affects people of all ages. However, children younger than 15 years typically present with only a nonspecific, self-
limited, febrile illness. In endemic areas, a high prevalence of immunity in adults may limit outbreaks to children.

In Southeast Asia, where dengue is hyperendemic, dengue hemorrhagic fever usually affects children younger than 15 years.
However, in the Americas, where dengue is becoming progressively hyperendemic, dengue hemorrhagic fever shows no age
predilection.

Prognosis
Dengue fever is typically a self-limiting disease with a mortality rate of less than 1%. When treated, dengue hemorrhagic fever
has a mortality rate of 2-5%. When left untreated, dengue hemorrhagic fever has a mortality rate as high as 50%. Survivors
usually recover without sequelae and develop immunity to the infecting serotype.

The fatality rate associated with severe dengue varies by country, from 12-44%. In a 1997 Cuban epidemic, the fatality rate in
patients who met criteria for severe dengue was approximately 6%. The mortality rate associated with dengue fever is less than
1%. Data from the 1997 Cuban epidemic suggest that, for every clinically apparent case of dengue fever, 13.9 cases of dengue
infection went unrecognized because of absent or minimal symptoms.

A 2005 review from Singapore of 14,209 patients found that useful predictors of death included the following[54] :

Atypical presentations
Significant comorbid illness
Abnormal serum markers (including albumin and coagulation studies)
Secondary bacterial infections

Factors that affect disease severity include the following:

Patient age
Pregnancy

https://emedicine.medscape.com/article/215840-print 13/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
Nutritional status
Ethnicity
Sequence of infection with different dengue serotypes
Virus genotype
Quality and extent of available medical care

Complications and sequelae of dengue virus infections are rare but may include the following:

Cardiomyopathy
Seizures, encephalopathy, and viral encephalitis
Hepatic injury
Depression
Pneumonia
Iritis
Orchitis
Oophoritis

In 20-30% of dengue hemorrhagic fever cases, the patient develops shock, known as the dengue shock syndrome. Worldwide,
children younger than 15 years constitute 90% of dengue hemorrhagic fever patients[46] ; however, in the Americas, dengue
hemorrhagic fever occurs in both adults and children.

Although dengue is an extremely important arboviral illness globally, literature evaluating the economic impact is fairly sparse,
with some conflicting findings. A recent expert panel assessment and 2 studies in the Americas recommended additional
research to fill important information gaps, including disease outcomes and accurate statistics regarding disease burden, that
could better inform future decision making regarding control and prevention.[55, 56, 57]

A 5-year prospective study in Thai children examined the relative economic burden of dengue infection in children on the local
population. Most disability-adjusted life years (DALYs) lost to dengue resulted from long-term illness in children who had not
been hospitalized. The infecting serotype appeared to be the major determinant of DALYs lost, with DEN-2 and DEN-3
responsible for 59%. The mean cost of illness from dengue was significantly higher than that from other febrile illnesses studied.
[52]

A prospective study examined the direct and indirect costs of dengue infection in 1695 pediatric and adult patients in 8
countries. The average illness lasted 11.9 days for ambulatory patients and 11 days for hospitalized patients. Hospitalized
students lost 5.6 days of school. Those at work lost 9.9 work days. Overall mean costs were more than double (1394
international dollars [I$]) for hospitalized cases. With an annual average of 594,000 cases the aggregate economic cost was
estimated to be at least I$587 million, without factoring in underreporting of disease and dengue surveillance and vector control
costs. This represents a significant global economic burden in low-income countries.[57]

Patient Education
Educate patients, especially those who have experienced prior dengue fever, to avoid mosquito bites, including the use of
appropriate mosquito repellants and peridomestic vector control, when traveling to dengue-endemic areas. Current evidence
suggests that those with a history of dengue fever are at highest risk for dengue hemorrhagic fever or dengue shock syndrome if
they are infected with a different dengue strain.

Information for reducing risk of contracting dengue while traveling, as well as current information on dengue outbreaks, is
available at the US Centers for Disease Control and Prevention Travel & Dengue Outbreaks Web page. Information on dengue
and alerts on current outbreaks are also available through the World Health Organization Web site.

Presentation

History
Patients with dengue will have a history of living in, or recent travel to, a region where the disease is endemic. The incubation
period is 3-14 days (average, 4-7 days); symptoms that begin more than 2 weeks after a person departs from an endemic area

https://emedicine.medscape.com/article/215840-print 14/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
are probably not due to dengue.

Many patients experience a prodrome of chills, erythematous mottling of the skin, and facial flushing (a sensitive and specific
indicator of dengue fever). The prodrome may last for 2-3 days. Children younger than 15 years usually have a nonspecific
febrile syndrome, which may be accompanied by a maculopapular rash. Classic dengue fever begins with sudden onset of
fever, chills, and severe (termed breakbone) aching of the head, back, and extremities, as well as other symptoms. The fever
lasts 2-7 days and may reach 41°C. Fever that lasts longer than 10 days is probably not due to dengue.

Pain and other accompanying symptoms may include any of the following:

Fever
Headache
Retro-orbital pain
General body pain (arthralgias, myalgias)
Nausea and vomiting (however, diarrhea is rare)
Rash
Weakness
Altered taste sensation
Anorexia
Sore throat
Mild hemorrhagic manifestations (eg, petechiae, bleeding gums, epistaxis, menorrhagia, hematuria)
Lymphadenopathy

Rash in dengue fever is a maculopapular or macular confluent rash over the face, thorax, and flexor surfaces, with islands of
skin sparing. The rash typically begins on day 3 and persists 2-3 days.

Fever typically abates with the cessation of viremia. Occasionally, and more commonly in children, the fever abates for a day
and then returns, a pattern that has been called saddleback fever. A second rash may occur within 1-2 days of defervescence,
lasting 1-5 days; it is morbilliform, is maculopapular, spares the palms and soles, and occasionally desquamates.

Recovery is complete but slow, with fatigue and exhaustion often persisting after the fever has subsided. The convalescent
phase may last for 2 weeks.

Patients are at risk for development of dengue hemorrhagic fever or dengue shock syndrome at approximately the time of
defervescence. Abdominal pain in conjunction with restlessness, change in mental status, hypothermia, and a drop in the
platelet count presages the development of dengue hemorrhagic fever.

Of patients with dengue hemorrhagic fever, 90% are younger than 15 years. The initial phase of dengue hemorrhagic fever is
similar to that of dengue fever and other febrile viral illnesses. Shortly after the fever breaks (or sometimes within 24 hours
before), signs of plasma leakage appear, along with the development of hemorrhagic symptoms such as bleeding from sites of
trauma, gastrointestinal bleeding, and hematuria. Patients may also present with abdominal pain, vomiting, febrile seizures (in
children), and a decreased level of consciousness.

If left untreated, dengue hemorrhagic fever most likely progresses to dengue shock syndrome. Common symptoms in
impending shock include abdominal pain, vomiting, and restlessness. Patients also may have symptoms related to circulatory
failure.

Physical Examination
Dengue fever presents in a nonspecific manner and may not be distinguishable from other viral or bacterial illness. According to
the Pan American Health Organization (PAHO), the clinical description of dengue fever is an acute febrile illness of 2-7 days
duration associated with 2 or more of the following:

Severe and generalized headache


Retro-orbital pain
Severe myalgias, especially of the lower back, arms, and legs
Arthralgias, usually of the knees and shoulders
Characteristic rash
Hemorrhagic manifestations
Leukopenia

Additional findings may include the following:

https://emedicine.medscape.com/article/215840-print 15/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
Injected conjunctivae
Facial flushing, a sensitive and specific predictor of dengue infection
Inflamed pharynx
Lymphadenopathy
Nausea and vomiting
Nonproductive cough
Tachycardia, bradycardia, and conduction defects

Up to half of patients with dengue fever develop a characteristic rash. The rash is variable and may be maculopapular or
macular. Petechiae and purpura may develop as hemorrhagic manifestations. Hemorrhagic manifestations most commonly
include petechiae and bleeding at venipuncture sites.

A tourniquet test is often positive. This test is performed by inflating a blood pressure cuff on the upper arm to midway between
diastolic and systolic blood pressures for 5 minutes. The results are considered to be positive if more than 20 petechiae per
square inch are observed on the skin in the area that was under pressure. Other hemorrhagic manifestations include nasal or
gingival bleeding, melena, hematemesis, and menorrhagia.

Neurologic manifestations such as seizures and encephalitis/encephalopathy have been reported in rare cases of dengue
infection. Some of these cases did not display other typical features of dengue infection. Other neurologic complications
associated with dengue infection include neuropathies, Guillain-Barré syndrome, and transverse myelitis.

Dengue hemorrhagic fever

Findings for dengue hemorrhagic fever are similar to those for dengue fever and include the following:

Biphasic fever curve

Hemorrhagic findings more pronounced than in dengue fever

Signs of peritoneal effusion, pleural effusion, or both

Minimal criteria for the diagnosis of dengue hemorrhagic fever, according to the World Health Organization (WHO), are as
follows[58] :

Fever

Hemorrhagic manifestations (eg, hemoconcentration, thrombocytopenia, positive tourniquet test)

Circulatory failure, such as signs of vascular permeability (eg, hypoproteinemia, effusions)

Hepatomegaly

In addition, conjunctival injection develops in approximately one third of patients with dengue hemorrhagic fever. Optic
neuropathy has been reported and occasionally results in permanent and significant visual impairment.[59] Pharyngeal injection
develops in almost 97% of patients with dengue hemorrhagic fever. Generalized lymphadenopathy is observed.

Hepatomegaly is present more often in dengue shock syndrome than in milder cases. Hepatic transaminase levels may be
mildly to moderately elevated. Encephalopathy is a rare complication that may result from a combination of cerebral edema,
intracranial hemorrhage, anoxia, hyponatremia, and hepatic injury.

Dengue shock syndrome

Findings of dengue shock syndrome include the following:

Hypotension

Bradycardia (paradoxical) or tachycardia associated with hypovolemic shock

Hepatomegaly

Hypothermia

Narrow pulse pressure (< 20 mm Hg)

Signs of decreased peripheral perfusion

https://emedicine.medscape.com/article/215840-print 16/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

DDx

Diagnostic Considerations
Studies indicate that as many as 50% of dengue cases may be misdiagnosed, as a result of inaccurate assessment of the signs
and symptoms of disease presentation. This inaccuracy can lead to increased cost of treatment, such as unneeded
hospitalizations, as well as possibly increased morbidity and mortality due to volume overload from overzealous use of
intravenous fluids.[60]

A Belgian study examined predictors of diagnosis in 1962 febrile travelers and expatriates returning from the tropics. After
malaria was ruled out, the main predictors of dengue infection included skin rash, thrombocytopenia, and leukopenia.[61]

Dengue must be carefully differentiated from preeclampsia during pregnancy. An overlap of symptoms and signs, including
thrombocytopenia, impaired liver function, capillary leak, ascites, and decreased urine output may make this clinically
challenging. Definitive diagnosis is confirmed via serology.

Rare cases of vertical dengue transmission have been reported. If the mother acquires infection in the peripartum period,
newborns should be evaluated for dengue with platelet counts and serologic studies.[62, 63]

Differential Diagnoses
Arenaviruses

Chikungunya Virus

Ebola Virus Infection

Hemorrhagic Fever Viruses

Idiopathic Thrombocytopenic Purpura (ITP) in Emergency Medicine

Influenza

Leptospirosis

Malaria

Mayaro Fever

Meningitis

Orbivirus

Rickettsial Infection

River Virus

Rocky Mountain Spotted Fever (RMSF)

Roseola Infantum

Ross River Fever

Scarlet Fever

Sinbis Virus

Typhus

Viral Hepatitis

West Nile Virus (WNV) Infection and Encephalitis (WNE)

Yellow Fever

Zika Virus

https://emedicine.medscape.com/article/215840-print 17/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Workup

Workup

Approach Considerations
Because the signs and symptoms of dengue fever are nonspecific, attempting laboratory confirmation of dengue infection is
important. Laboratory criteria for diagnosis include one or more of the following:

Demonstration of a fourfold or greater change in reciprocal immunoglobulin G (IgG) or immunoglobulin M (IgM) antibody
titers to one or more dengue virus antigens in paired serum samples
Demonstration of dengue virus antigen in autopsy tissue via immunohistochemistry or immunofluorescence or in serum
samples via enzyme immunoassay (MAC-ELISA, IgG ELISA, NSI-ELISA, EIA)
Detection of viral genomic sequences in autopsy tissue, serum, or cerebral spinal fluid (CSF) samples via reverse-
transcriptase polymerase chain reaction (RT-PCR)
Less commonly, isolation of dengue virus from serum, plasma, leukocytes, or autopsy samples

RT-PCR yields a serotype-specific diagnosis very rapidly.[64, 65] It is most useful early in the course of illness. It is not
susceptible to the cross-reactivity with other flaviviruses seen with serologic testing.

The following laboratory tests should also be performed:

Complete blood cell (CBC) count


Metabolic panel
Serum protein and albumin levels
Liver panel
Coagulation profile and disseminated intravascular coagulation (DIC) panel

Characteristic findings in dengue fever are thrombocytopenia (platelet count < 100 x 109/L), leukopenia, and mild-to-moderate
elevation of aspartate aminotransferase and alanine aminotransferase values. Jaundice and acute liver failure are uncommon.
Peak liver enzyme levels occur later than other complications in adults studied prospectively in Vietnam. Enzyme levels begin to
rise during the early stage and peak during the second week. Clinically severe involvement was found to be idiosyncratic and
infrequent but did contribute to severe bleeding.[66]

A hematocrit level increase greater than 20% is a sign of hemoconcentration and precedes shock. The hematocrit level should
be monitored at least every 24 hours to facilitate early recognition of dengue hemorrhagic fever and every 3-4 hours in severe
cases of dengue hemorrhagic fever or dengue shock syndrome.

In patients with dengue hemorrhagic fever, the following may be present:

Increased hematocrit level secondary to plasma extravasation and/or third-space fluid loss
Hypoproteinemia
Prolonged prothrombin time
Prolonged activated partial thromboplastin time
Decreased fibrinogen
Increased amount of fibrin split products

Signs of early coagulopathy may be as subtle as a guaiac test that is positive for occult blood in the stool. Guaiac testing should
be performed on all patients in whom dengue virus infection is suspected.

Typing and crossmatching of blood should be performed in cases of severe dengue hemorrhagic fever or dengue shock
syndrome because blood products may be required.

Urinalysis identifies hematuria. Cultures of blood, urine, CSF, and other body fluids should be performed as necessary to
exclude or confirm other potential causes of the patient's condition.

Arterial blood gas should be assessed in patients with severe cases to assess pH, oxygenation, and ventilation.

Electrocardiography may demonstrate nonspecific changes as a result of fever, electrolyte disturbances, tachycardia, or
medications. The usefulness of these changes as a marker of cardiac involvement is unclear.

https://emedicine.medscape.com/article/215840-print 18/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
Biopsy of the skin lesions in patients with nonfatal, uncomplicated dengue fever reveals an abnormality of the small blood
vessels. Endothelial swelling, perivascular edema, and mononuclear cell infiltration are the primary histologic findings.

Perform chest radiography to look for pleural effusions and bronchopneumonia. Right-sided pleural effusion is typical. Bilateral
pleural effusions are common in patients with dengue shock syndrome. Head computed tomography without contrast may be
indicated in patients with altered level of consciousness, to detect intracranial bleeding or cerebral edema from dengue
hemorrhagic fever.

Since January 2010, dengue has been a reportable illness in all states in the United States. Report known or suspected cases
of dengue fever, dengue hemorrhagic fever, or dengue shock syndrome to public health authorities. Such reports should include
the following:

Patient demographics and recent travel history


Case classification
Date of onset of illness
Whether hospitalization was necessary
Outcome

When multiple patients are involved, reports should include the number of cases of dengue fever and severe dengue stratified
by age, number of confirmed cases and serotypes, and number of hospitalizations and deaths.

Complete Blood Cell Count


Leukopenia, often with lymphopenia, is observed near the end of the febrile phase of illness. Lymphocytosis, with atypical
lymphocytes, commonly develops before defervescence or shock. A systematic review found that patients with dengue had
significantly lower total WBC, neutrophil, and platelet counts than patients with other febrile illnesses in dengue-endemic
populations.[67]

A hematocrit level increase greater than 20% is a sign of hemoconcentration and precedes shock. The hematocrit level should
be monitored at least every 24 hours to facilitate early recognition of dengue hemorrhagic fever and every 3-4 hours in severe
cases of dengue hemorrhagic fever or dengue shock syndrome.

Thrombocytopenia has been demonstrated in up to 50% of dengue fever cases. Platelet counts less than 100,000 cells/μL are
seen in dengue hemorrhagic fever or dengue shock syndrome and occur before defervescence and the onset of shock. The
platelet count should be monitored at least every 24 hours to facilitate early recognition of dengue hemorrhagic fever.

Metabolic Panel and Liver Enzymes


Hyponatremia is the most common electrolyte abnormality in patients with dengue hemorrhagic fever or dengue shock
syndrome. Metabolic acidosis is observed in those with shock and must be corrected rapidly. Elevated blood urea nitrogen
(BUN) levels are observed in those with shock. Acute kidney injury is uncommon.[68, 69]

Transaminase levels may be mildly elevated into the several thousands in patients with dengue hemorrhagic fever who have
acute hepatitis. Low albumin levels are a sign of hemoconcentration.

Coagulation Studies
Coagulation studies may help to guide therapy in patients with severe hemorrhagic manifestations. Findings are as follows:

Prothrombin time is prolonged

Activated partial thromboplastin time is prolonged

https://emedicine.medscape.com/article/215840-print 19/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
Low fibrinogen and elevated fibrin degradation product levels are signs of disseminated intravascular coagulation

Serum Studies
Serum specimens should be sent to the laboratory for serodiagnosis, PCR, and viral isolation. Because the signs and symptoms
of dengue fever are nonspecific, attempting laboratory confirmation of dengue infection is important. Serodiagnosis is made
based on a rise in antibody titer in paired specimens obtained during the acute stage and during convalescence. Results vary
depending on whether the infection is primary or secondary.

Early in the illness (≤5 days after symptom onset), laboratory confirmation can be made from a single acute-phase serum
specimen by detecting dengue virus genomic sequences with RT-PCR or DENV nonstructural protein 1 (NS1) antigen via
immunoassay. Later in the illness, IgM anti-DENV can be detected with ELISA. The CDC currently recommends that, within the
first week of illness, diagnostic testing should include a test for dengue virus (RT-PCR or NS1) and IgM anti-DENV. For patients
seen more than one week after fever onset, IgM anti-DENV, such as the MAC-ELISA, is more useful, although NS1 may still be
positive up to 12 days after fever onset.

The IgM capture enzyme-linked immunosorbent assay (MAC-ELISA) has become the most widely used serologic assay for
dengue. Other tests are also used, however, including the following:

Complement fixation (CF)


Neutralization test (NT)
Hemagglutination inhibition (HI)
IgG ELISA
NS1 strip test [70]

Draw serum specimens for diagnosis as soon as possible after the onset of illness or hospitalization and at the time of death or
discharge from the hospital. Immediately place specimens on wet ice and send to the laboratory. Obtain a second (ie,
convalescent) blood sample for convalescent-phase serologic testing 7-21 days after the acute-phase serum specimen was
drawn. Ideally, draw the convalescent-phase serum specimen 10 days after the acute-phase specimen.

A European study found that if only a single serum sample is available, a single positive result on enzyme-linked ELISA (PanBio
IgM or IgG) has a high rate of false positivity and should be confirmed using a second, more specific diagnostic technique. In the
absence of further testing, platelet and white blood cell counts can be diagnostically helpful, because the combination of
thrombocytopenia and leukopenia is present in 40.4% of confirmed cases but in only 6.1% of false-positive cases.[71, 72]

Ultrasonography
Ultrasonography is a potentially timely, cost-effective, and easily used modality in the evaluation of potential dengue
hemorrhagic fever. Positive and reliable ultrasonographic findings include fluid in the chest and abdominal cavities, pericardial
effusion, and a thickened gallbladder wall. Thickening of the gallbladder wall may presage clinically significant vascular
permeability.[7, 73]

The utility of previous studies was limited because patients underwent only a single scan. However, in a study by
Srikiatkhachorn et al, daily serial ultrasonographic examinations of the thorax and abdomen proved useful in the evaluation of
patients with suspected dengue hemorrhagic fever.[73] A study conducted by Santhosh et al found similar results, with a
thickened gallbladder wall found in 66.7% of seropositive dengue cases, 64.5% having ascites, and 50% having pleural effusion.
[74]

Plasma leakage was detected in some patients within 3 days of fever onset. Pleural effusion was the most common sign. Based
on ultrasonographic findings, dengue hemorrhagic fever was predicted in 12 patients before hemoconcentration criteria had
been met.

Case Definitions
https://emedicine.medscape.com/article/215840-print 20/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Cases are classified as suspected dengue if they are compatible with the clinical description. They are classified as probable
dengue if they are compatible with the clinical definition and satisfy one or more of the following criteria:

Supportive serology (reciprocal hemagglutination-inhibition antibody titer greater than 1280, comparable IgG EIA titers, or
positive IgM antibody test in late acute or convalescent-phase serum specimen)
Occurrence at the same location and time as other confirmed cases of dengue fever

A confirmed case of dengue is one that is compatible with the clinical definition and is confirmed by the laboratory.

Criteria for the diagnosis of severe dengue include a probable or confirmed case of dengue infection and hemorrhagic
tendencies as evidenced by one or more of the following:

A positive result from the tourniquet test


Petechiae, ecchymoses, or purpura
Bleeding from the mucosa, gastrointestinal tract, injection sites, or other sites
Hematemesis or melena and thrombocytopenia (< 100,000 cells/μL)
Evidence of plasma leakage due to increased vascular permeability

Plasma leakage may manifest as one or more of the following:

Greater than 20% rise in average hematocrit level for age and sex
Greater than 20% drop in hematocrit level following volume replacement compared with baseline
Signs of plasma leakage (eg, pleural effusion, ascites, hypoproteinemia)

Severe dengue with shock is diagnosed in cases meeting all of the above criteria plus evidence of circulatory failure, such as
the following:

Rapid, weak pulse


Narrow pulse pressure (< 20 mm Hg), with increased peripheral vascular resistance (PVR) and elevated diastolic
pressure
Hypotension
Cool, clammy skin
Altered mental status, although the patient may initially remain alert

The onset of shock may be subtle, indicated by raised diastolic pressure and increased PVR in an alert patient.

WHO classification

The accuracy of the World Health Organization (WHO) classification system for dengue has been called into question.[75] A
study in Indonesian children found that the WHO classification system was in only modest agreement with the intuitive
classification by treating physicians, whereas several modified classification systems were in good agreement.[76]

The WHO classification system was found to have a sensitivity of 86% for the detection of dengue shock syndrome.[25]
Modified systems that added the above early predictors of compensated shock and considered models using varying
combinations of evidence of hemorrhagic tendencies, thrombocytopenia, and hemoconcentration were found to yield higher
sensitivities (88-99%).

Treatment

Approach Considerations
Dengue fever is usually a self-limited illness. There is no specific antiviral treatment currently available for dengue fever. The
World Health Organization (WHO) has provided a number of free publications about dengue.

Supportive care with analgesics, fluid replacement, and bed rest is usually sufficient. Acetaminophen may be used to treat fever
and relieve other symptoms. Aspirin, nonsteroidal anti-inflammatory drugs (NSAIDs), and corticosteroids should be avoided.
Management of severe dengue requires careful attention to fluid management and proactive treatment of hemorrhage.

Single-dose methylprednisolone showed no mortality benefit in the treatment of dengue shock syndrome in a prospective,
randomized, double-blind, placebo-controlled trial.[77] The Novartis Institute for Tropical Diseases (NITD) in Singapore is
carrying out research to find inhibitors of dengue viral target proteins to reduce the viral load during active infection.[78]
https://emedicine.medscape.com/article/215840-print 21/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Suspected Dengue
Oral rehydration therapy is recommended for patients with moderate dehydration caused by high fever and vomiting. Patients
with known or suspected dengue fever should have their platelet count and hematocrit measured daily from the third day of
illness until 1-2 days after defervescence. Patients with clinical signs of dehydration and patients with a rising hematocrit level or
falling platelet count should have intravascular volume deficits replaced under close observation. Those who improve can
continue to be monitored in an outpatient setting, and those who do not improve should be admitted to the hospital for continued
hydration.

Patients who develop signs of dengue hemorrhagic fever warrant closer observation. Admission for intravenous fluid
administration is indicated for patients who develop signs of dehydration, such as the following:

Tachycardia

Prolonged capillary refill time

Cool or mottled skin

Diminished pulse amplitude

Altered mental status

Decreased urine output

Rising hematocrit

Narrowed pulse pressure

Hypotension

Severe Dengue
Successful management of severe dengue requires careful attention to fluid management and proactive treatment of
hemorrhage. Admission to an intensive care unit is indicated for patients with dengue shock syndrome.

Patients may need a central intravenous line for volume replacement and an arterial line for accurate blood pressure monitoring
and frequent blood tests. Exercise caution when placing intravascular catheters because of the increased bleeding
complications of dengue hemorrhagic fever. Urethral catheterization may be useful to strictly monitor urine output.

Intravascular volume deficits should be corrected with isotonic fluids such as Ringer lactate solution. Boluses of 10-20 mL/kg
should be given over 20 minutes and may be repeated. If this fails to correct the deficit, the hematocrit value should be
determined. If it is rising, limited clinical information suggests that a plasma expander may be administered. Starch, dextran 40,
or albumin 5% at a dose of 10-20 mL/kg may be used. One study has suggested that starch may be preferable because of
hypersensitivity reactions to dextran.[79]

If the patient does not improve after infusion of a plasma expander, blood loss should be considered. Patients with internal or
gastrointestinal bleeding may require transfusion, and patients with coagulopathy may require fresh frozen plasma.

After patients with dehydration are stabilized, they usually require intravenous fluids for no more than 24-48 hours. Intravenous
fluids should be stopped when the hematocrit falls below 40% and adequate intravascular volume is present. At this time,
patients reabsorb extravasated fluid and are at risk for volume overload if intravenous fluids are continued. Do not interpret a
falling hematocrit value in a clinically improving patient as a sign of internal bleeding.

Platelet and fresh frozen plasma transfusions may be required to control severe bleeding. A case report demonstrated good
improvement following intravenous anti-D globulin administration in 2 patients. The authors proposed that, as in immune
thrombocytopenic purpura from disorders other than dengue, intravenous anti-D produces Fcγ receptor blockade to raise
platelet counts.[80]

https://emedicine.medscape.com/article/215840-print 22/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Patients who are resuscitated from shock rapidly recover. Patients with dengue hemorrhagic fever or dengue shock syndrome
may be discharged from the hospital when they meet the following criteria:

Afebrile for 24 hours without antipyretics

Good appetite, clinically improved condition

Adequate urine output

Stable hematocrit level

At least 48 hours since recovery from shock

No respiratory distress

Platelet count greater than 50,000 cells/μL

Pregnant patients

Dengue in pregnancy must be carefully differentiated from preeclampsia. An overlap of signs and symptoms, including
thrombocytopenia, capillary leak, impaired liver function, ascites, and decreased urine output may make this clinically
challenging. Pregnant women with dengue fever respond well to the usual therapy of fluids, rest, and antipyretics. However, 3
cases of maternal death due to dengue fever in the third trimester have been reported. An awareness of the clinical and
laboratory manifestations of dengue in pregnancy should allow its early recognition and the institution of appropriate treatment.
If the mother acquires infection in the peripartum period, newborns should be evaluated for dengue with serial platelet counts
and serological studies.[81, 82]

Diet and Activity


No specific diet is necessary for patients with dengue fever. Patients who are able to tolerate oral fluids should be encouraged to
drink oral rehydration solution, fruit juice, or water to prevent dehydration from fever, lack of oral intake, or vomiting. Return of
appetite after dengue hemorrhagic fever or dengue shock syndrome is a sign of recovery.

Bed rest is recommended for patients with symptomatic dengue fever, dengue hemorrhagic fever, or dengue shock syndrome.
Permit the patient to gradually resume their previous activities, especially during the long period of convalescence.

Prevention
The only way to truly prevent dengue virus acquisition is to avoid being bitten by a vector mosquito. Although this can be
accomplished by avoiding travel to areas where dengue is endemic, that is not an ideal strategy because it would require a
person to avoid most tropical and subtropical regions of the world, many of which are popular travel and work destinations.
Other measures are as follows:

Wear N,N-diethyl-3-methylbenzamide (DEET)–containing mosquito repellant


Wear protective clothing, preferably impregnated with permethrin insecticide
Remain in well-screened or air-conditioned places
The use of mosquito netting is of limited benefit, as Aedes are day-biting mosquitoes
Eliminate the mosquito vector using indoor sprays

The most widely used mosquito-control technique, spraying cities to kill adult mosquitoes, is not effective. Efforts should target
the larval phase with larvicides and cleaning up larvae habitats. Poor sanitation and poor refuse control provide excellent
conditions for mosquito larvae to grow. Hurricanes and other natural disasters increase the habitat for mosquito growth in urban
areas by increasing rubble and garbage, which act as water reservoirs.

Breeding of vector mosquitoes can be reduced by eliminating small accumulations of stagnant water around human habitats
(eg, disposing of old tires, covering water receptacles, and changing water in birdbaths daily. Support community-based vector
control programs (including source reduction) and the use of vectoricidal agents, including predatory copepods as biological
control agents.[83, 84, 85, 86]
https://emedicine.medscape.com/article/215840-print 23/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

In 2011, an Australian research effort infected Aedes aegypti mosquitoes with the intracellular bacterium Wolbachia in the
laboratory. Maternally inherited Wolbachia prevents dengue virus from replicating within mosquitos. They then released these
mosquitos into the wild to mate with wild mosquitos and pass the Wolbachia along to their offspring. After releasing
approximately 10 mosquitos/house/week for 10 weeks, they found that greater than 80% of collected wild mosquitos had
Wolbachia infection. Infected mosquitos laid fewer eggs and exhibited shorter life spans. The research group plans further field
trials in Vietnam, Indonesia, and Brazil. If successful, this would provide a practical biological approach to dengue suppression.
[87]

Outbreaks of dengue will increasingly cross common borders of endemic and disease-free countries unless the following
measures are undertaken:

Increased health surveillance

Prompt reporting of new cases

Heightened professional awareness

Public education

Vaccine Development
One vaccine is currently approved for the prevention of dengue infection. Sanofi Pasteur registered Dengvaxia (CYD-TDV), a
live recombinant tetravalent vaccine, in several countries in late 2015-2016, with Mexico being the initial country to register the
vaccine in December 2015. The vaccine is given in 3 doses at age 0, 6, and 12 months. It underwent testing in more than
30,000 volunteers and was shown to reduce the risk of severe illness and hospitalization by as much as 30% in individuals
previously infected with one or more strains. The vaccine proved less effective in persons who were not previously exposed to
dengue and in areas with a lower burden of disease.[88] Owing to concern that the vaccine may act like an initial dengue
infection in this second group of individuals not previously infected with the virus, with additional exposure to a second serotype
placing these individuals at increased risk of severe dengue, the WHO released a position paper in July 2016, stating that
countries should consider introduction of vaccine as a part of an comprehensive dengue control strategy only where
epidemiologic data indicate a high burden of disease.[89]

In April 2018, the WHO’s Strategic Advisory Group of Experts recommended that rapid diagnostic tests to establish serostatus
be developed so that prevaccination screening might be used to assess serostatus before CYD-TDV vaccine is administered.
[90] As of late 2018, the CYD-TDV vaccine had been approved for use and was being marketed in European endemic areas to
persons aged 9-45 years with prior exposure to the disease.[91]

Dengue vaccine was approved by the FDA in 2019 for prevention of dengue disease caused by dengue virus serotypes 1, 2, 3,
and 4 in individuals aged 9-16 years with laboratory-confirmed previous dengue infection who live in endemic areas. It is
approved only in individuals previously infected by any dengue virus serotype or in whom this information is unknown. Persons
not previously infected are at an increased risk of severe dengue disease when vaccinated and subsequently infected with
dengue virus.

The approval was based on data from 2 placebo-controlled studies in patients (n > 35,000) living in dengue-endemic areas.
Patients were randomized 2:1 to receive either the vaccine or saline placebo and monitored for symptomatic virologically
confirmed dengue (VCD) starting at day 0. Vaccine efficacy was assessed beginning 28 days after the third vaccination for 12
months. The vaccine was approximately 76% effective in preventing symptomatic VCD disease among patients aged 9-16 years
who were seropositive for dengue at baseline.[92]

Immunity to a single dengue strain is the major risk factor for severe dengue; as such, a vaccine must provide high levels of
immunity to all 4 dengue strains to be clinically useful.[93] Seroconversion alone does not predict protection. Several other
immunogenic tetravalent vaccine candidates have been developed, and two are in phase 3 clinical trials.[94, 95, 96, 97, 98]

Consultations
Consultation with an infectious diseases specialist may be helpful in guiding decisions regarding diagnosis and treatment.
Consultation with a critical care medicine specialist may be helpful when treating patients with dengue hemorrhagic fever or

https://emedicine.medscape.com/article/215840-print 24/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

dengue shock syndrome and severe hemorrhagic manifestations or shock.

Telephone consultation may be obtained from the Centers for Disease Control and Surveillance (800-232-4636, 8am-8pm
ET/Monday-Friday).

Medication

Medication Summary
No specific antiviral medication is currently available to treat dengue. The treatment of dengue fever is symptomatic and
supportive in nature. Bed rest and mild analgesic-antipyretic therapy are often helpful in relieving lethargy, malaise, and fever
associated with the disease. Acetaminophen (paracetamol) is recommended for treatment of pain and fever. Aspirin, other
salicylates, and nonsteroidal anti-inflammatory drugs (NSAIDs) should be avoided.

Patients with dengue hemorrhagic fever or dengue shock syndrome may require intravenous volume replacement. Plasma
volume expanders can be used in patients who do not respond to isotonic fluids.

Dengue vaccine was approved by the FDA in 2019 for individuals aged 9-16 years with laboratory-confirmed previous dengue
infection who live in endemic areas.

Analgesics

Class Summary
These agents are used to reduce fever. They inhibit prostaglandin synthesis in the central nervous system. They also inhibit
hypothalamic heat-regulating center, which in turn promotes the return of the set-point temperature to normal.

Acetaminophen (Tylenol, Feverall, Acephen, Mapap)


Acetaminophen (paracetamol) reduces fever by acting directly on hypothalamic heat-regulating centers, which increases
dissipation of body heat via vasodilation and sweating. It is used in dengue infections to relieve pain and lower temperature
when fever is thought to contribute to patient discomfort.

Volume Expanders

Class Summary
Plasma volume expanders are used in the treatment of intravascular volume deficits or shock to restore intravascular volume,
blood pressure, and tissue perfusion.

Dextran 40 (LMD)
Dextran 40 is a polymer of glucose. When infused, it increases intravascular volume, blood pressure, and capillary perfusion. It
is used to restore intravascular volume when isotonic crystalloid administration is inadequate for that purpose.

Albumin (Albuminar-5, Buminate, Plasbumin 5)

https://emedicine.medscape.com/article/215840-print 25/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Human albumin is a sterile solution of albumin, which is the major plasma protein responsible for the colloid oncotic pressure of
blood. It is pooled from blood, serum, plasma, or placenta from healthy donors. Infusion of albumin results in a shift of fluid from
the extracellular space into the bloodstream, thereby decreasing hemoconcentration and blood viscosity.

Albumin may be administered wide open when treating shock. Patient response must be assessed before repeating the dose.

Hetastarch (Hespan, Hextend)


Hydroxyethyl starch is a sterile solution of the starch responsible for the colloid oncotic pressure of blood. Hetastarch produces
volume expansion through its highly colloidal starch structure.

Vaccines, Live, Viral

Class Summary
Elicits dengue-specific immune responses against the 4 dengue virus serotypes (ie, serotypes 1, 2, 3, and 4).

Dengue vaccine (Dengvaxia)


Indicated for prevention of dengue disease caused by dengue virus serotypes 1, 2, 3, and 4 in individuals aged 9-16 years with
laboratory-confirmed previous dengue infection who live in endemic areas. It is approved only in individuals previously infected
by any dengue virus serotype or in whom this information is unknown. Persons not previously infected are at an increased risk
of severe dengue disease when vaccinated and subsequently infected with dengue virus. Immunization is a series of 3 SC
injections administered 6 months apart.

Questions & Answers


Overview

What is dengue?

What is severe dengue (dengue hemorrhagic fever)?

What is the mortality rate for dengue?

What is the typical presentation of dengue?

What are the signs and symptoms of dengue?

What are the clinical characteristics of severe dengue (dengue hemorrhagic fever)?

What are the lab criteria for the diagnosis of dengue?

How is dengue detected in early phase of disease?

Which lab tests should be performed in the workup of possible dengue?

What are the characteristic lab findings in dengue?

What are the characteristic lab findings in severe dengue (dengue hemorrhagic fever)?

What is the role of guaiac testing in suspected dengue?

Which imaging studies are used in the evaluation of suspected dengue?

When is oral rehydration indicated for the management of dengue?

https://emedicine.medscape.com/article/215840-print 26/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

When is inpatient care indicated for dengue?

When is a transfusion of fresh frozen plasma (FFP) indicated in the treatment of dengue?

What is dengue?

How is dengue transmitted?

What is the global incidence of dengue?

What are symptoms of dengue?

What is severe dengue (dengue hemorrhagic fever)?

How is dengue transmitted?

What is characteristic of hyperendemic dengue?

Which lab techniques are used to confirm dengue infection?

What are treatment options used for dengue?

What is the earliest known documentation of dengue?

When was the first reported dengue epidemic?

How frequently do outbreaks of severe dengue (dengue hemorrhagic fever) occur?

How has dengue spread in Southeast Asia?

How was dengue prevented in the Americas in the late 20th century?

What is the global incidence of dengue outbreaks in 2014?

What are the differences between serotype 1 dengue (DENV-1) and serotype 2 dengue (DENV-2)?

Where was the first clearly identified local transmission of dengue in the US?

What is characteristic of the dengue outbreaks in Hawaii in the 21st century?

Which vectors for dengue are present in the US and what is their geographic range?

What is the mortality rate of dengue in the US?

What is the prevalence of dengue in Europe?

Which factors are responsible for the increased spread of dengue?

What are the benefits of travel surveillance for dengue?

What is the pathophysiology for dengue?

How are dengue viruses transmitted?

What are characteristics of the Aedes mosquito species that transmit dengue?

What are the primary hosts for the transmission of dengue?

How does the transmission of dengue occur?

What is the duration of classic dengue fever?

What is the disease progression of dengue?

Which age group is at highest risk for dengue (dengue hemorrhagic fever)?

What is the disease progression of severe dengue (dengue hemorrhagic fever)?

What is the role of plasma leakage in the pathophysiology of severe dengue (dengue hemorrhagic fever)?

https://emedicine.medscape.com/article/215840-print 27/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

What is severe dengue shock?

What is the immunopathology of severe dengue (dengue hemorrhagic fever)?

Which strain of dengue is most virulent?

What are the causes of thrombocytopenia in dengue?

What causes dengue infection?

What are the serotypes of dengue?

What are risk factors for dengue infection?

What is the largest known epidemic of dengue?

What is the incidence of dengue in the US?

What is the principal mosquito vector for dengue in the US?

Where in the US do most dengue cases occur?

What is the global incidence of dengue?

In the 21st century, which countries previously without dengue have reported outbreaks?

What are the morbidity and mortality of severe dengue (dengue hemorrhagic fever) in Southeast Asia?

What is the economic burden of dengue infection in Southeast Asia?

How does the incidence and mortality of dengue vary by age in in Southeast Asia?

What is the epidemiology of dengue in Africa?

Which areas of South America are at highest risk for dengue?

How does the incidence of dengue vary by race?

How does the incidence of dengue vary by sex?

How does the incidence of dengue vary by age?

What is the prognosis of dengue fever?

What are the risk factors for death by dengue fever?

Which factors affect the severity of dengue?

What are the complications and sequelae of dengue virus infections?

What is dengue shock syndrome?

What is the economic impact of dengue?

What information about dengue should patients be given?

Where can information about reducing the risks of contracting dengue fever during travel be found?

Presentation

What should be the focus of medical history in suspected dengue?

What are the typical symptoms of dengue?

What are the signs and symptoms of dengue?

How is rash characterized in dengue?

How long is the recovery process for patients with dengue?

https://emedicine.medscape.com/article/215840-print 28/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

When in the disease course of dengue is the risk of developing severe dengue (dengue hemorrhagic fever) the highest?

What are the signs and symptoms of severe dengue (dengue hemorrhagic fever)?

What happens if severe dengue (dengue hemorrhagic fever) is left untreated?

What are the Pan American Health Organization (PAHO) diagnostic criteria for dengue?

Which physical findings are characteristic of dengue fever?

What is the prevalence of rash due to dengue infection and how is it characterized?

How is a tourniquet test performed in the evaluation of dengue?

What are neurologic manifestations of dengue?

Which physical findings are characteristic of severe dengue?

What are the WHO criteria for the diagnosis of severe dengue (dengue hemorrhagic fever)?

Which physical findings are characteristic of severe dengue (dengue hemorrhagic fever)?

Which physical findings are characteristic of dengue shock syndrome?

DDX

How frequently is dengue misdiagnosed?

What are the main indicators of dengue infection?

How is dengue differentiated from preeclampsia?

What are the diagnostic considerations for vertical dengue transmission?

What are the differential diagnoses for Dengue?

Workup

What are the lab criteria for the diagnosis of dengue?

Which lab tests should be performed for the diagnosis of dengue?

What are the characteristic lab findings in dengue fever?

What is the role of hematocrit level measurement in the diagnosis of dengue?

Which lab findings suggest severe dengue (dengue hemorrhagic fever)?

When is guaiac testing indicated in the diagnosis of suspected dengue?

When is typing and crossmatching of blood indicated for the diagnosis of dengue?

What is the role of urinalysis in the diagnosis of dengue?

What is the role of arterial blood gas assessment in the diagnosis of dengue?

What is the role of electrocardiography in the diagnosis of dengue?

What are the primary histologic findings of dengue?

What is the role of chest radiography in the diagnosis of dengue?

What should be included in a public health report of dengue?

What is the significance of a finding of leukopenia in dengue?

What is the role of hematocrit level monitoring in the workup of suspected dengue?

What is the prevalence of thrombocytopenia in dengue?

https://emedicine.medscape.com/article/215840-print 29/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

What is the most common electrolyte abnormality in patients with severe dengue (dengue hemorrhagic fever)?

What is the role of liver enzyme testing in the diagnosis of dengue?

What is the role of coagulation studies in the management of severe dengue (dengue hemorrhagic fever)?

What is the role of serum studies in the diagnosis of dengue?

Which serum tests are used for the diagnosis of dengue?

How is a serum study performed in the diagnosis of dengue?

What is the role of ultrasonography in the diagnose dengue?

What are the criteria used to classify a case as suspected or probable dengue?

What is required to confirm a case of dengue?

What are the criteria required to diagnose a case of severe dengue (dengue hemorrhagic fever)?

What are the manifestations of plasma leakage in dengue?

How is circulatory failure characterized in the diagnostic criteria for severe dengue shock?

How does the WHO classify dengue?

Treatment

What is the antiviral treatment for dengue?

What are the treatment approaches for dengue?

What is the role of steroids in the treatment of dengue?

What is the role of oral rehydration therapy in the treatment of dengue?

When is IV fluid administration indicated in the treatment of dengue?

What is required for the successful management of severe dengue (dengue hemorrhagic fever)?

How are fluid volume and blood pressure managed in severe dengue (dengue hemorrhagic fever)?

When are patients with severe dengue (dengue hemorrhagic fever) discharged from the hospital?

How is dengue diagnosed and treated during pregnancy?

Which dietary modifications are beneficial during the treatment of dengue?

Which activity modifications are beneficial during treatment of dengue?

How is dengue prevented?

What is the most widely used mosquito-control technique for the prevention of dengue?

How is the breeding of vector mosquitos reduced for the prevention of dengue?

What is a biological approach to reducing vector mosquitos and preventing dengue?

Which public health measures need to be taken to prevent dengue outbreaks?

What is the role of vaccination in the prevention of dengue?

What are the limitations of vaccine in the prevention of dengue?

Which specialists should be consulted for the management of dengue?

Medications

Which medications are used for the treatment of dengue?

https://emedicine.medscape.com/article/215840-print 30/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

Which medications in the drug class Vaccines, Live, Viral are used in the treatment of Dengue?

Which medications in the drug class Volume Expanders are used in the treatment of Dengue?

Which medications in the drug class Analgesics are used in the treatment of Dengue?

Which medications in the drug class are used in the treatment of Dengue?

Contributor Information and Disclosures

Author

Darvin Scott Smith, MD, MSc, DTM&H Adjunct Associate Clinical Professor, Department of Microbiology and Immunology,
Stanford University School of Medicine; Chief of Infectious Diseases and Geographic Medicine, Department of Internal
Medicine, Kaiser Permanente Medical Group

Darvin Scott Smith, MD, MSc, DTM&H is a member of the following medical societies: American Medical Association, American
Society of Tropical Medicine and Hygiene, Infectious Diseases Society of America, International Society of Travel Medicine

Disclosure: Nothing to disclose.

Coauthor(s)

David J Mariano Stanford University

Disclosure: Nothing to disclose.

Micah Lynne Trautwein Stanford University

Disclosure: Nothing to disclose.

Chief Editor

Michael Stuart Bronze, MD David Ross Boyd Professor and Chairman, Department of Medicine, Stewart G Wolf Endowed
Chair in Internal Medicine, Department of Medicine, University of Oklahoma Health Science Center; Master of the American
College of Physicians; Fellow, Infectious Diseases Society of America; Fellow of the Royal College of Physicians, London

Michael Stuart Bronze, MD is a member of the following medical societies: Alpha Omega Alpha, American College of
Physicians, American Medical Association, Association of Professors of Medicine, Infectious Diseases Society of America,
Oklahoma State Medical Association, Southern Society for Clinical Investigation

Disclosure: Nothing to disclose.

Additional Contributors

Suzanne Moore Shepherd, MD, MS, DTM&H, FACEP, FAAEM Professor of Emergency Medicine, Education Officer,
Department of Emergency Medicine, Hospital of the University of Pennsylvania; Director of Education and Research, PENN
Travel Medicine; Medical Director, Fast Track, Department of Emergency Medicine

Suzanne Moore Shepherd, MD, MS, DTM&H, FACEP, FAAEM is a member of the following medical societies: Alpha Omega
Alpha, American Academy of Emergency Medicine, American Society of Tropical Medicine and Hygiene, International Society of
Travel Medicine, Society for Academic Emergency Medicine, Wilderness Medical Society

Disclosure: Nothing to disclose.

Patrick B Hinfey, MD Emergency Medicine Residency Director, Department of Emergency Medicine, Newark Beth Israel
Medical Center; Clinical Assistant Professor of Emergency Medicine, New York College of Osteopathic Medicine

Patrick B Hinfey, MD is a member of the following medical societies: American Academy of Emergency Medicine, Wilderness
Medical Society, American College of Emergency Physicians, Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

https://emedicine.medscape.com/article/215840-print 31/36
8/26/2019 https://emedicine.medscape.com/article/215840-print

William H Shoff, MD, DTM&H Former Director, PENN Travel Medicine; Former Associate Professor, Department of Emergency
Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania School of Medicine

William H Shoff, MD, DTM&H is a member of the following medical societies: American College of Physicians, American Society
of Tropical Medicine and Hygiene, International Society of Travel Medicine, Society for Academic Emergency Medicine,
Wilderness Medical Society

Disclosure: Nothing to disclose.

Acknowledgements

Joseph Domachowske, MD Professor of Pediatrics, Microbiology and Immunology, Department of Pediatrics, Division of
Infectious Diseases, State University of New York Upstate Medical University

Joseph Domachowske, MD is a member of the following medical societies: Alpha Omega Alpha, American Academy of
Pediatrics, American Society for Microbiology, Infectious Diseases Society of America, Pediatric Infectious Diseases Society,
and Phi Beta Kappa

Disclosure: Nothing to disclose.

Hagop A Isnar, MD, FACEP Department of Emergency Medicine, Crouse Hospital

Hagop A Isnar, MD, FACEP is a member of the following medical societies: American College of Emergency Physicians,
American Medical Association, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Thomas M Kerkering, MD Chief of Infectious Diseases, Virginia Tech, Carilion School of Medicine, Roanoke, Virginia

Thomas M Kerkering, MD is a member of the following medical societies: Alpha Omega Alpha, American College of Physicians,
American Public Health Association, American Society for Microbiology, American Society of Tropical Medicine and Hygiene,
Infectious Diseases Society of America, Medical Society of Virginia, and Wilderness Medical Society

Disclosure: Nothing to disclose.

Deborah Sentochnik, MD Consulting Staff, Department of Internal Medicine, Division of Infectious Disease, The Mary Imogene
Bassett Hospital

Deborah Sentochnik, MD is a member of the following medical societies: American College of Physicians, Infectious Diseases
Society of America, and Medical Society of the State of New York

Disclosure: Nothing to disclose.

Russell W Steele, MD Head, Division of Pediatric Infectious Diseases, Ochsner Children's Health Center; Clinical Professor,
Department of Pediatrics, Tulane University School of Medicine

Russell W Steele, MD is a member of the following medical societies: American Academy of Pediatrics, American Association of
Immunologists, American Pediatric Society, American Society for Microbiology, Infectious Diseases Society of America,
Louisiana State Medical Society, Pediatric Infectious Diseases Society, Society for Pediatric Research, and Southern Medical
Association

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy;
Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Reference Salary Employment

Mary L Windle, PharmD, Adjunct Associate Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-
Chief, Medscape Drug Reference

Disclosure: Nothing to disclose.

References

1. World Health Organization. Dengue and severe dengue fact sheet. WHO. Available at
http://www.who.int/mediacentre/factsheets/fs117/en/. April 2017; Accessed: September 28, 2017.

https://emedicine.medscape.com/article/215840-print 32/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
2. Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, et al. The global distribution and burden of dengue. Nature.
2013 Apr 25. 496 (7446):504-7. [Medline].

3. Brady OJ, Gething PW, Bhatt S, Messina JP, Brownstein JS, Hoen AG, et al. Refining the global spatial limits of dengue virus
transmission by evidence-based consensus. PLoS Negl Trop Dis. 2012. 6 (8):e1760. [Medline].

4. Wilson ME, Chen LH. Dengue: update on epidemiology. Curr Infect Dis Rep. 2015 Jan. 17 (1):457. [Medline].

5. Ten threats to global health in 2019. World Health Organization. Available at https://www.who.int/emergencies/ten-threats-to-global-
health-in-2019. 2019; Accessed: Feb 2, 2019.

6. Kyle JL, Harris E. Global spread and persistence of dengue. Annu Rev Microbiol. 2008. 62:71-92. [Medline].

7. Statler J, Mammen M, Lyons A, Sun W. Sonographic findings of healthy volunteers infected with dengue virus. J Clin Ultrasound.
2008 Sep. 36(7):413-7. [Medline].

8. Gubler DJ. Cities spawn epidemic dengue viruses. Nat Med. 2004 Feb. 10(2):129-30. [Medline].

9. Wilder-Smith A, Gubler DJ. Geographic expansion of dengue: the impact of international travel. Med Clin North Am. 2008 Nov.
92(6):1377-90, x. [Medline].

10. Messina JP, Brady OJ, Scott TW, Zou C, Pigott DM, Duda KA, et al. Global spread of dengue virus types: mapping the 70 year
history. Trends Microbiol. 2014 Mar. 22 (3):138-46. [Medline]. [Full Text].

11. Halstead SB. Dengue. Lancet. 2007 Nov 10. 370(9599):1644-52. [Medline].

12. Chowell G, Torre CA, Munayco-Escate C, Suárez-Ognio L, López-Cruz R, Hyman JM. Spatial and temporal dynamics of dengue
fever in Peru: 1994-2006. Epidemiol Infect. 2008 Dec. 136(12):1667-77. [Medline].

13. Osterwell N. Dengue 'Under-recognized' as Source of Febrile Illness in US. Medscape Medical News. Jan 23 2014. Available at
http://www.medscape.com/viewarticle/819656. Accessed: January 25, 2014.

14. Sharp TM, Gaul L, Muehlenbachs A, Hunsperger E, Bhatnagar J, Lueptow R, et al. Fatal hemophagocytic lymphohistiocytosis
associated with locally acquired dengue virus infection - new Mexico and Texas, 2012. MMWR Morb Mortal Wkly Rep. 2014 Jan 24.
63(3):49-54. [Medline].

15. Freedman DO, Weld LH, Kozarsky PE, Fisk T, Robins R, von Sonnenburg F. Spectrum of disease and relation to place of exposure
among ill returned travelers. N Engl J Med. 2006 Jan 12. 354(2):119-30. [Medline].

16. Liu-Helmersson J, Quam M, Wilder-Smith A, Stenlund H, Ebi K, Massad E, et al. Climate Change and Aedes Vectors: 21st Century
Projections for Dengue Transmission in Europe. EBioMedicine. 2016 May. 7:267-77. [Medline].

17. CDC. Imported dengue--United States, 1997 and 1998. MMWR Morb Mortal Wkly Rep. 2000 Mar 31. 49(12):248-53. [Medline]. [Full
Text].

18. Engelthaler DM, Fink TM, Levy CE, Leslie MJ. The reemergence of Aedes aegypti in Arizona. Emerg Infect Dis. 1997 Apr-Jun.
3(2):241-2. [Medline]. [Full Text].

19. Chye JK, Lim CT, Ng KB, et al. Vertical transmission of dengue. Clin Infect Dis. 1997 Dec. 25(6):1374-7. [Medline].

20. Wagner D, de With K, Huzly D, Hufert F, Weidmann M, Breisinger S, et al. Nosocomial acquisition of dengue. Emerg Infect Dis. 2004
Oct. 10(10):1872-3. [Medline].

21. Dejnirattisai W, Duangchinda T, Lin CL, Vasanawathana S, Jones M, Jacobs M, et al. A complex interplay among virus, dendritic
cells, T cells, and cytokines in dengue virus infections. J Immunol. 2008 Nov 1. 181(9):5865-74. [Medline].

22. Halstead SB, Heinz FX, Barrett AD, Roehrig JT. Dengue virus: molecular basis of cell entry and pathogenesis, 25-27 June 2003,
Vienna, Austria. Vaccine. 2005 Jan 4. 23(7):849-56. [Medline].

23. Limjindaporn T, Wongwiwat W, Noisakran S, Srisawat C, Netsawang J, Puttikhunt C, et al. Interaction of dengue virus envelope
protein with endoplasmic reticulum-resident chaperones facilitates dengue virus production. Biochem Biophys Res Commun. 2009
Feb 6. 379(2):196-200. [Medline].

24. Zhang JL, Wang JL, Gao N, Chen ZT, Tian YP, An J. Up-regulated expression of beta3 integrin induced by dengue virus serotype 2
infection associated with virus entry into human dermal microvascular endothelial cells. Biochem Biophys Res Commun. 2007 May
11. 356(3):763-8. [Medline].

25. Rothman AL, Ennis FA. Immunopathogenesis of Dengue hemorrhagic fever. Virology. 1999 Apr 25. 257(1):1-6. [Medline].

26. Chen LC, Lei HY, Liu CC, Shiesh SC, Chen SH, Liu HS. Correlation of serum levels of macrophage migration inhibitory factor with
disease severity and clinical outcome in dengue patients. Am J Trop Med Hyg. 2006 Jan. 74(1):142-7. [Medline].

https://emedicine.medscape.com/article/215840-print 33/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
27. Green S, Rothman A. Immunopathological mechanisms in dengue and dengue hemorrhagic fever. Curr Opin Infect Dis. 2006 Oct.
19(5):429-36. [Medline].

28. Guzman MG, Alvarez M, Rodriguez-Roche R, Bernardo L, Montes T, Vazquez S. Neutralizing antibodies after infection with dengue
1 virus. Emerg Infect Dis. 2007 Feb. 13(2):282-6. [Medline].

29. Restrepo BN, Ramirez RE, Arboleda M, Alvarez G, Ospina M, Diaz FJ. Serum levels of cytokines in two ethnic groups with dengue
virus infection. Am J Trop Med Hyg. 2008 Nov. 79(5):673-7. [Medline].

30. Rothman AL. Dengue: defining protective versus pathologic immunity. J Clin Invest. 2004 Apr. 113(7):946-51. [Medline].

31. de Macedo FC, Nicol AF, Cooper LD, Yearsley M, Pires AR, Nuovo GJ. Histologic, viral, and molecular correlates of dengue fever
infection of the liver using highly sensitive immunohistochemistry. Diagn Mol Pathol. 2006 Dec. 15(4):223-8. [Medline].

32. Shah I. Dengue and liver disease. Scand J Infect Dis. 2008. 40(11-12):993-4. [Medline].

33. Dejnirattisai W, Jumnainsong A, Onsirisakul N, et al. Cross-reacting antibodies enhance dengue virus infection in humans. Science.
2010 May 7. 328(5979):745-8. [Medline].

34. Schmidt AC. Response to dengue fever--the good, the bad, and the ugly?. N Engl J Med. 2010 Jul 29. 363(5):484-7. [Medline].

35. Kurane I, Innis BL, Nimmannitya S, Nisalak A, Meager A, Ennis FA. High levels of interferon alpha in the sera of children with
dengue virus infection. Am J Trop Med Hyg. 1993 Feb. 48(2):222-9. [Medline].

36. Ojha A, Nandi D, Batra H, Singhal R, Annarapu GK, Bhattacharyya S, et al. Platelet activation determines the severity of
thrombocytopenia in dengue infection. Sci Rep. 2017 Jan 31. 7:41697. [Medline].

37. Hottz ED, Oliveira MF, Nunes PC, Nogueira RM, Valls-de-Souza R, Da Poian AT, et al. Dengue induces platelet activation,
mitochondrial dysfunction and cell death through mechanisms that involve DC-SIGN and caspases. J Thromb Haemost. 2013 May.
11 (5):951-62. [Medline].

38. Wang E, Ni H, Xu R, Barrett AD, Watowich SJ, Gubler DJ. Evolutionary relationships of endemic/epidemic and sylvatic dengue
viruses. J Virol. 2000 Apr. 74(7):3227-34. [Medline].

39. Centers for Disease Control and Prevention Web site. CDC traveler's health page. Dengue. Available at
http://www.cdc.gov/Dengue/travelOutbreaks/. Accessed: October 20, 2011.

40. Chen WS, Wong CH, Cillekens L. Dengue antibodies in a suburban community in Malaysia. Med J Malaysia. 2003 Mar. 58(1):142-3.
[Medline].

41. Istúriz RE, Gubler DJ, Brea del Castillo J. Dengue and dengue hemorrhagic fever in Latin America and the Caribbean. Infect Dis Clin
North Am. 2000 Mar. 14(1):121-40, ix. [Medline].

42. Hotez PJ, Bottazzi ME, Franco-Paredes C, Ault SK, Periago MR. The neglected tropical diseases of Latin America and the
Caribbean: a review of disease burden and distribution and a roadmap for control and elimination. PLoS Negl Trop Dis. 2008 Sep
24. 2(9):e300. [Medline]. [Full Text].

43. Centers for Disease Control and Prevention (CDC). Travel-associated Dengue surveillance - United States, 2006-2008. MMWR
Morb Mortal Wkly Rep. 2010 Jun 18. 59(23):715-9. [Medline]. [Full Text].

44. Centers for Disease Control and Prevention (CDC). Locally acquired Dengue--Key West, Florida, 2009-2010. MMWR Morb Mortal
Wkly Rep. 2010 May 21. 59(19):577-81. [Medline].

45. Rey JR. Dengue in Florida (USA). Insects. 2014 Dec 16. 5 (4):991-1000. [Medline].

46. Malavige GN, Fernando S, Fernando DJ, Seneviratne SL. Dengue viral infections. Postgrad Med J. 2004 Oct. 80(948):588-601.
[Medline]. [Full Text].

47. Stephenson JR. Understanding dengue pathogenesis: implications for vaccine design. Bull World Health Organ. 2005 Apr.
83(4):308-14. [Medline]. [Full Text].

48. World Health Organization. Impact of Dengue. Available at http://www.who.int/csr/disease/dengue/impact/en/index.html. Accessed:


October 14, 2011.

49. Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, et al. The global distribution and burden of dengue. Nature.
2013 Apr 25. 496 (7446):504-7. [Medline].

50. Brady OJ, Gething PW, Bhatt S, Messina JP, Brownstein JS, Hoen AG, et al. Refining the global spatial limits of dengue virus
transmission by evidence-based consensus. PLoS Negl Trop Dis. 2012. 6 (8):e1760. [Medline].

51. Lin CC, Huang YH, Shu PY, et al. Characteristic of dengue disease in Taiwan: 2002-2007. Am J Trop Med Hyg. 2010 Apr. 82(4):731-
9. [Medline]. [Full Text].

https://emedicine.medscape.com/article/215840-print 34/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
52. Anderson KB, Chunsuttiwat S, Nisalak A, Mammen MP, Libraty DH, Rothman AL. Burden of symptomatic dengue infection in
children at primary school in Thailand: a prospective study. Lancet. 2007 Apr 28. 369(9571):1452-9. [Medline].

53. Anker M, Arima Y. Male-female differences in the number of reported incident dengue fever cases in six Asian countries. Western
Pac Surveill Response J. 2011 Apr. 2 (2):17-23. [Medline].

54. Lahiri M, Fisher D, Tambyah PA. Dengue mortality: reassessing the risks in transition countries. Trans R Soc Trop Med Hyg. 2008
Oct. 102(10):1011-6. [Medline].

55. Beatty ME, Beutels P, Meltzer MI, et al. Health economics of dengue: a systematic literature review and expert panel's assessment.
Am J Trop Med Hyg. 2011 Mar. 84(3):473-88. [Medline]. [Full Text].

56. Shepard DS, Coudeville L, Halasa YA, Zambrano B, Dayan GH. Economic impact of dengue illness in the Americas. Am J Trop Med
Hyg. 2011 Feb. 84(2):200-7. [Medline]. [Full Text].

57. Suaya JA, Shepard DS, Siqueira JB, et al. Cost of dengue cases in eight countries in the Americas and Asia: a prospective study.
Am J Trop Med Hyg. 2009 May. 80(5):846-55. [Medline].

58. WHO. Dengue haemorrhagic fever: diagnosis, treatment, prevention and control. World Health Organization. 1997. Available at
http://www.who.int/topics/dengue/en/.

59. Sanjay S, Wagle AM, Au Eong KG. Dengue optic neuropathy. Ophthalmology. 2009 Jan. 116(1):170; author reply 170. [Medline].

60. Teves Maria A. Wrong treatment most common cause of dengue fatality. ABS/CBN News. Available at http://www.abs-
cbnnews.com/nation/09/03/09/wrong-treatment-most-common-cause-dengue-fatality. Accessed: September 3, 2010.

61. Bottieau E, Clerinx J, Van den Enden E, Van Esbroeck M, Colebunders R, Van Gompel A. Fever after a stay in the tropics:
diagnostic predictors of the leading tropical conditions. Medicine (Baltimore). 2007 Jan. 86(1):18-25. [Medline].

62. Malhotra N, Chanana C, Kumar S. Dengue infection in pregnancy. Int J Gynaecol Obstet. 2006 Aug. 94(2):131-2. [Medline].

63. Singh N, Sharma KA, Dadhwal V, Mittal S, Selvi AS. A successful management of dengue fever in pregnancy: report of two cases.
Indian J Med Microbiol. 2008 Oct-Dec. 26(4):377-80. [Medline].

64. Warrilow D, Northill JA, Pyke A, Smith GA. Single rapid TaqMan fluorogenic probe based PCR assay that detects all four dengue
serotypes. J Med Virol. 2002 Apr. 66(4):524-8. [Medline].

65. Kong YY, Thay CH, Tin TC, Devi S. Rapid detection, serotyping and quantitation of dengue viruses by TaqMan real-time one-step
RT-PCR. J Virol Methods. 2006 Dec. 138(1-2):123-30. [Medline].

66. Trung DT, Thao le TT, Hien TT, et al. Liver involvement associated with dengue infection in adults in Vietnam. Am J Trop Med Hyg.
2010 Oct. 83(4):774-80. [Medline]. [Full Text].

67. Potts JA, Rothman AL. Clinical and laboratory features that distinguish dengue from other febrile illnesses in endemic populations.
Trop Med Int Health. 2008 Nov. 13(11):1328-40. [Medline]. [Full Text].

68. Lima EQ, Nogueira ML. Viral hemorrhagic fever-induced acute kidney injury. Semin Nephrol. 2008 Jul. 28(4):409-15. [Medline].

69. Lombardi R, Yu L, Younes-Ibrahim M, Schor N, Burdmann EA. Epidemiology of acute kidney injury in Latin America. Semin Nephrol.
2008 Jul. 28(4):320-9. [Medline].

70. Chaterji S, Allen JC Jr, Chow A, Leo YS, Ooi EE. Evaluation of the NS1 rapid test and the WHO dengue classification schemes for
use as bedside diagnosis of acute dengue fever in adults. Am J Trop Med Hyg. 2011 Feb. 84(2):224-8. [Medline]. [Full Text].

71. Wichmann O, Stark K, Shu PY, Niedrig M, Frank C, Huang JH. Clinical features and pitfalls in the laboratory diagnosis of dengue in
travellers. BMC Infect Dis. 2006. 6:120. [Medline].

72. Domingo C, de Ory F, Sanz JC, Reyes N, Gascón J, Wichmann O, et al. Molecular and serologic markers of acute dengue infection
in naive and flavivirus-vaccinated travelers. Diagn Microbiol Infect Dis. 2009 Sep. 65(1):42-8. [Medline].

73. Srikiatkhachorn A, Krautrachue A, Ratanaprakarn W, Wongtapradit L, Nithipanya N, Kalayanarooj S. Natural history of plasma
leakage in dengue hemorrhagic fever: a serial ultrasonographic study. Pediatr Infect Dis J. 2007 Apr. 26(4):283-90; discussion 291-
2. [Medline].

74. Santhosh VR, Patil PG, Srinath MG, Kumar A, Jain A, Archana M. Sonography in the diagnosis and assessment of dengue Fever. J
Clin Imaging Sci. 2014. 4:14. [Medline]. [Full Text].

75. Srikiatkhachorn A, Gibbons RV, Green S, et al. Dengue hemorrhagic fever: the sensitivity and specificity of the world health
organization definition for identification of severe cases of dengue in Thailand, 1994-2005. Clin Infect Dis. 2010 Apr 15. 50(8):1135-
43. [Medline]. [Full Text].

76. Setiati TE, Mairuhu AT, Koraka P, Supriatna M, Mac Gillavry MR, Brandjes DP, et al. Dengue disease severity in Indonesian children:
an evaluation of the World Health Organization classification system. BMC Infect Dis. 2007 Mar 26. 7:22. [Medline]. [Full Text].
https://emedicine.medscape.com/article/215840-print 35/36
8/26/2019 https://emedicine.medscape.com/article/215840-print
77. Tassniyom S, Vasanawathana S, Chirawatkul A, Rojanasuphot S. Failure of high-dose methylprednisolone in established dengue
shock syndrome: a placebo-controlled, double-blind study. Pediatrics. 1993 Jul. 92(1):111-5. [Medline].

78. WHO. Dengue. Available at http://www.who.int/topics/dengue/en/. Accessed: October 20, 2011.

79. 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 Sep 1. 353(9):877-89. [Medline].

80. Yadav SP, Sachdeva A, Gupta D, Sharma SD, Kharya G. Control of massive bleeding in dengue hemorrhagic fever with severe
thrombocytopenia by use of intravenous anti-D globulin. Pediatr Blood Cancer. 2008 Dec. 51(6):812-3. [Medline].

81. Waduge R, Malavige GN, Pradeepan M, Wijeyaratne CN, Fernando S, Seneviratne SL. Dengue infections during pregnancy: a case
series from Sri Lanka and review of the literature. J Clin Virol. 2006 Sep. 37(1):27-33. [Medline].

82. Ismail NA, Kampan N, Mahdy ZA, Jamil MA, Razi ZR. Dengue in pregnancy. Southeast Asian J Trop Med Public Health. 2006 Jul.
37(4):681-3. [Medline].

83. Billingsley PF, Foy B, Rasgon JL. Mosquitocidal vaccines: a neglected addition to malaria and dengue control strategies. Trends
Parasitol. 2008 Sep. 24(9):396-400. [Medline].

84. Erlanger TE, Keiser J, Utzinger J. Effect of dengue vector control interventions on entomological parameters in developing countries:
a systematic review and meta-analysis. Med Vet Entomol. 2008 Sep. 22(3):203-21. [Medline].

85. Kay B, Vu SN. New strategy against Aedes aegypti in Vietnam. Lancet. 2005 Feb 12-18. 365(9459):613-7. [Medline].

86. Hanh TT, Hill PS, Kay BH, Quy TM. Development of a framework for evaluating the sustainability of community-based dengue
control projects. Am J Trop Med Hyg. 2009 Feb. 80(2):312-8. [Medline].

87. 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 Aug 24. 476 (7361):454-7. [Medline].

88. Lang J. Recent progress on sanofi pasteur's dengue vaccine candidate. J Clin Virol. 2009 Oct. 46 Suppl 2:S20-4. [Medline].

89. World Health Organization. Dengue vaccine: WHO position paper – July 2016. Wkly Epidemiol Rec. 2016 Jul 29. 91 (30):349-64.
[Medline].

90. Aguiar M, Stollenwerk N, Halstead SB. The risks behind Dengvaxia recommendation. Lancet Infect Dis. 2016 Aug. 16 (8):882-3.
[Medline]. [Full Text].

91. Dengvaxia. European Medicines Agency. Available at https://www.ema.europa.eu/en/medicines/human/EPAR/dengvaxia. 2018 Dec


18; Accessed: Feb 15, 2019.

92. Dengvaxia (dengue vaccine) [package insert]. Swiftwater, PA: Sanofi Pasteur, Inc. May 2019. Available at [Full Text].

93. Monath TP. Dengue and yellow fever--challenges for the development and use of vaccines. N Engl J Med. 2007 Nov 29.
357(22):2222-5. [Medline].

94. McArthur JH, Durbin AP, Marron JA, Wanionek KA, Thumar B, Pierro DJ, et al. Phase I clinical evaluation of rDEN4Delta30-200,201:
a live attenuated dengue 4 vaccine candidate designed for decreased hepatotoxicity. Am J Trop Med Hyg. 2008 Nov. 79(5):678-84.
[Medline]. [Full Text].

95. O'Brien J. 12th Annual Conference on Vaccine Research. Expert Rev Vaccines. 2009 Sep. 8(9):1139-42. [Medline].

96. Edelman R. Dengue vaccines approach the finish line. Clin Infect Dis. 2007 Jul 15. 45 Suppl 1:S56-60. [Medline].

97. Blaney JE Jr, Durbin AP, Murphy BR, Whitehead SS. Development of a live attenuated dengue virus vaccine using reverse genetics.
Viral Immunol. 2006 Spring. 19(1):10-32. [Medline].

98. Questions and Answers on Dengue Vaccines. World Health Organization. Available at
https://www.who.int/immunization/research/development/dengue_q_and_a/en/. 2018 April 20; Accessed: February 10, 2019.

99. Larsen CP, Whitehead SS, Durbin AP. Dengue human infection models to advance dengue vaccine development. Vaccine. 2015
Sep 28. [Medline].

100. Anker M, Arima Y. Male-female differences in the number of reported incident dengue fever cases in six Asian countries. Western
Pac Surveill Response J. 2011 Apr. 2 (2):17-23. [Medline].

https://emedicine.medscape.com/article/215840-print 36/36

You might also like