+ All Categories
Home > Documents > 1-s2.0-S0022347697700544-main

1-s2.0-S0022347697700544-main

Date post: 26-Dec-2015
Category:
Upload: hanadilah
View: 3 times
Download: 0 times
Share this document with a friend
Popular Tags:
9
ii{ii~ii!i!i~ ;¸¸~¸¸¸¸ ':: <:i~i!Jiiiiiiii~ .... virus infection: Epidemiology, pathogenesis, presentation, diagnosis, and prevention PhD, Z]/[aoc £ Robinson, MD, and Ursula Kuhnle, ~rD Dengue virus infection is now the most com- mon arthropod-borne disease worldwide with an increasing incidence in the tropical regions of Asia, Africa, and Central and South America. It presents a spectrum of disease, ranging from a harmless flulike ill- ness to a severe hemorrhagic fever with high morbidity and mortality, the latter almost entirely in children. The virus is transmitted by mosquitoes, AedeJ aegypti and Aedea albopic- taJ. Cold-resistant strains of Aedes vectors have been identified that may pose a threat to children living in temperate zones. 1-4 It is estimated that 100 million cases of infection occur worldwide per year. Between 1986 and 1990 1.5 million cases of dengue hemorrhagic fever or dengue shock syndrome with 15,940 deaths were reported to the World Health Organi- zation. Deaths occurred primarily in chil- dren between the ages of 5 and 15 years. It is clear that dengue virus infection is the most important arthropod-borne viral dis- ease in human beings both in terms of mor- bidity and mortality rates, s' 6 GLOBAL DISTRIBUTION Dengue occurs in endemic and epidem- ic forms. Recorded epidemics of a From the Depart,zwnt ofMicrobiology, FacultyofMedicine, University of Malaya, Kuala Lumpur, Malaysia; the Department of Pediatrics, Universityof Melbourne, R~al Children's Hospital, Melbourne, Australia; and the Department ofPediatrics,National UniversityofMalaysia, Kuala Lumpu~: Reprint requests: UrsulaKuhnle,MD, Universit~ts- kinderldinik, Abteilung Endokrinologie, Lindwurm- str. 4, 80337 Munich, Germany. J Pediatr 1997;131:516-24. Copyright © 1997by Mosby-YearBook, Inc. 0022-3476/97/$5.00 + 0 9/19/82209 denguelike disease date to 1779, when outbreaks were reported in Batavia (pre- sent-day Jakarta) and Cairo. Since then epidemics have been reported in Phila- delphia (1780), Zanzibar (1825 and 1870), Calcutta (1824, 1853, 1871, and 1905), the West Indies (1827), Hong Kong (1901), Greece (1927-1928), Australia (1925-1926; 1942), the United States (1922), and Japan (1942-1945). 7-9 See related article, p. 525. The earlier outbreaks were uncompli- cated cases of dengue fever characterized by high-grade fever and severe bone and back pains. Hemorrhagic complications have been increasingly documented in outbreaks since 1944. Between 1956 and 1989 more than 2.5 million cases of DHF were reported to the World Health Organization with 42,751 deaths. 5 Southeast Asia, particularly Thailand, /V[yanmar, and Vietnam, are the most af- fected areas, where large DHF epidemics occur each 3 to 4 years. 10-13 For example, between 1982 and 1987 Thailand record- ed more than 403,000 cases of DHF with approximately 2400 fatalities. Dengue he- morrhagic fever is among the 10 leading causes of illness and death in hospitalized children in these areas. 11 In recent years several countries with long-standing dengue virus activity report- ed epidemics of DHF for the first time (Fig. 1). China experienced its first epi- demic of DHF on Hainan Island in 1985-1986 with a morbidity of 1915 per 100,000 residents. 14 India (where DF has been endemic and with numerous epi- demics reported in the past 200 years) ex- perienced the first outbreak of DHF/DSS in New Delhi in 1988, with 24 patients hospitalized within 2 mouths and a case fa- tality rate of 33%. 15 In 1989 DHF/DSS was reported for the first time in Tahiti and New Caledonia. 16'17In 1990 Sri Lanka re- ported 935 cases with 54 deaths. 18 In the Americas there is an increasing incidence of DF and DHF. The first large epidemic of DHF occurred in Cuba in 1981 with 24,000 cases of DHF and 10,000 cases of DSS reported with 158 deaths during a 3-month period. 19'20 In 1986 and 1987 massive outbreaks of DF were reported in Brazil. 21'22 Subsequent serologic investigations estimated almost 4 million cases of DF compared with the clinically estimated 1 million.21 In 1988 an outbreak of DF was reported at 1700 m above sea level in Guerrero State, Mexico. 23 In 1990 almost one fourth of the 300,000 people living in Iquitos, Peru, contracted DF 24 and in the same year 3108 cases of DHF with 78 deaths were reported in Venezuela. 25 The increasingly widespread distribu- tion and the rising incidence of dengue virus infections is related to increased dis- tribution ofA. aegypti and to the increase in urban population in the "mega-cities" of Southeast Asia. Lack of effective pro- grams to contain the vector, development, and deterioration of the urban environ- ment are responsible. However, the rea- son for the change from simple DF epi- demics to a severe hemorrhagic disease,
Transcript
Page 1: 1-s2.0-S0022347697700544-main

ii{ii~ii!i!i~ ;̧ ¸~¸̧ ¸̧ ' : : <:i~i!Jiiiiiiii~ ....

virus infection: Epidemiology, pathogenesis, presentation, diagnosis, and prevention PhD, Z]/[aoc £ Robinson, MD, and Ursula Kuhnle, ~rD

Dengue virus infection is now the most com- mon arthropod-borne disease worldwide with an increasing incidence in the tropical regions of Asia, Africa, and Central and South America. It presents a spectrum of disease, ranging from a harmless flulike ill- ness to a severe hemorrhagic fever with high morbidity and mortality, the latter almost entirely in children. The virus is transmitted by mosquitoes, AedeJ aegypti and Aedea albopic-

taJ. Cold-resistant strains of Aedes vectors have been identified that may pose a threat to children living in temperate zones. 1-4

It is estimated that 100 million cases of infection occur worldwide per year. Between 1986 and 1990 1.5 million cases of dengue hemorrhagic fever or dengue shock syndrome with 15,940 deaths were reported to the World Health Organi- zation. Deaths occurred primarily in chil- dren between the ages of 5 and 15 years. It is clear that dengue virus infection is the most important arthropod-borne viral dis- ease in human beings both in terms of mor- bidity and mortality rates, s' 6

GLOBAL DISTRIBUTION

Dengue occurs in endemic and epidem- ic forms. Recorded epidemics of a

From the Depart,zwnt of Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; the Department of Pediatrics, University of Melbourne, R~al Children's Hospital, Melbourne, Australia; and the Department of Pediatrics, National University of Malaysia, Kuala Lumpu~: Reprint requests: Ursula Kuhnle, MD, Universit~ts- kinderldinik, Abteilung Endokrinologie, Lindwurm- str. 4, 80337 Munich, Germany. J Pediatr 1997;131:516-24. Copyright © 1997 by Mosby-Year Book, Inc. 0022-3476/97/$5.00 + 0 9/19/82209

denguelike disease date to 1779, when outbreaks were reported in Batavia (pre- sent-day Jakarta) and Cairo. Since then epidemics have been reported in Phila- delphia (1780), Zanzibar (1825 and 1870), Calcutta (1824, 1853, 1871, and 1905), the West Indies (1827), Hong Kong (1901), Greece (1927-1928), Australia (1925-1926; 1942), the United States (1922), and Japan (1942-1945). 7-9

See related article, p. 525.

The earlier outbreaks were uncompli- cated cases of dengue fever characterized by high-grade fever and severe bone and back pains. Hemorrhagic complications have been increasingly documented in outbreaks since 1944. Between 1956 and 1989 more than 2.5 million cases of D H F were reported to the World Health Organization with 42,751 deaths. 5 Southeast Asia, part icularly Thailand, /V[yanmar, and Vietnam, are the most af- fected areas, where large D H F epidemics occur each 3 to 4 years. 10-13 For example,

between 1982 and 1987 Thailand record- ed more than 403,000 cases of D H F with approximately 2400 fatalities. Dengue he- morrhagic fever is among the 10 leading causes of illness and death in hospitalized children in these areas. 11

In recent years several countries with long-standing dengue virus activity report- ed epidemics of D H F for the first time (Fig. 1). China experienced its first epi- demic of D H F on Hainan Island in 1985-1986 with a morbidity of 1915 per 100,000 residents. 14 India (where D F has been endemic and with numerous epi- demics reported in the past 200 years) ex-

perienced the first outbreak of DHF/DSS in New Delhi in 1988, with 24 patients hospitalized within 2 mouths and a case fa- tality rate of 33%. 15 In 1989 D H F / D S S was reported for the first time in Tahiti and New Caledonia. 16'17 In 1990 Sri Lanka re- ported 935 cases with 54 deaths. 18

In the Americas there is an increasing incidence of D F and DHF. The first large epidemic of D H F occurred in Cuba in 1981 with 24,000 cases of D H F and 10,000 cases of DSS reported with 158 deaths during a 3-month period. 19'20 In

1986 and 1987 massive outbreaks of D F were reported in Brazil. 21'22 Subsequent

serologic investigations estimated almost 4 million cases of D F compared with the clinically estimated 1 million. 21 In 1988 an outbreak of D F was reported at 1700 m above sea level in Guerrero State, Mexico. 23 In 1990 almost one fourth of

the 300,000 people living in Iquitos, Peru, contracted D F 24 and in the same year 3108 cases of D H F with 78 deaths were reported in Venezuela. 25

The increasingly widespread distribu- tion and the rising incidence of dengue virus infections is related to increased dis- tribution ofA. aegypti and to the increase in urban population in the "mega-cities" of Southeast Asia. Lack of effective pro- grams to contain the vector, development, and deterioration of the urban environ- ment are responsible. However, the rea- son for the change from simple D F epi- demics to a severe hemorrhagic disease,

Page 2: 1-s2.0-S0022347697700544-main

THE JOURNAL OF PEDIATRICS Volume 13 I, N u m b e r 4

KAUTNER, ROBINSON,AND KUHNLE

often associated with shock, DHF/DSS, . . . . . is not well understood. , a ~ ~ t~ 3~ ~

Furthermore, increased air travel and c=,,, hence the spread ofthe mosquito vector al- t3o ~ t " .

most certainly facilitates the spread of ~ ~ ~ ' f f e : ~ ~ ~ ' ~ DF'26 A linlited number °f "imP°fred" a~, ~ ~ ~ ( f l cases of DF and DHF have been reported from New Zealand, 27 the Netherlands, 28 ~, 4 . . 1.4 England, 29 Germany, and Switzerland. a°'al ' ~" ,.,, 1.2 " ~ ' ~ , ) ~ Inthe United States 22 of24 confirmed ~ ~ ~ a ~ J ~r t ~BL . a f "

cases in 1990 were imported from Oceania, . . ~ 7 . ~ ~:~ " ~ ' 1 " ~ . . . ; 2 " theCarribean'Asia'lVie~de°'andPeru'32 ~ ~ J ~ j ~

T R A N S M I S S I O N 0,o0 ..... o.~

ImJ DHF/O$S

The known natural hosts for dengue viruses are man, lower primates, and mos- quitoes. The arthropod vectors are mem- bers of the genus Aedea that thrive both in urban and rural areas. 3a-as The predomi- nant species implicated in disease trans- mission are A. aegypti and A. albopictu~. 56-a8

Aedea aegypt£ considered the most effi- cient vector, originated in the forests of Africa and has now spread throughout most of the world and is found between 50 degrees north latitude and 20 degrees south latitutde. 3942 The female mosquito feeds during the daytime, with peak activ- ity in the morning and late afternoon. 43'44 After feeding on a viremic individual, the mosquito may transmit the virus directly by change of host, or after 8 to 10 days during the time the virus multiplies in the salivary glands. 45 The infected mosquito then remains capable of transmission for its entire life. 43 Transovarian transmission of dengue viruses has been document- ed, 35'46'47 anclA, aeyyptf eggs are highly re- sistant to desiccation and can survive for extended periods. 48 These facts may ex- plain the occurrence of spontaneous out- breaks of DF.

Aedes a/bapictua is indigenous to Southeast Asia, feeds during the day, and has been shown to have a higher biting frequency than A. azgypH. 49 Recently it has been introduced into Nigeria, Europe, and the United States, apparently by shipments of used automobile tires. In the United States, A. albopicl~ has spread as far north as Chicago. 50"52 With the spread of the adaptable, cold-resistant strains of A. albopict~o to Europe, it has been sug- gested that conditions might arise, which

F ~ . 1. World map demonstrating the,global distribution of the various clinical forms of dengue virus in- fections and of the various subtypes of dengue viruses, Adapted from Halstead SB.The XXth century pan- demic: need for surveillance and research.World Health Stat Q 1992;45:292-8. Reprinted with permission.

are suitable for a major European out- break of dengue virus infection .4

T H E V IRUSES

Dengue viruses are members of the family Flaviviridae, which include human pathogens, such as the Japanese en- cephalitis virus and yellow fever virus. 53 Four dengue virus serotypes and various "biotypes" can be differentiated.

All members of the family Flaviviridae share common morphologie characteris- tics, genome structure, and replication and translation strategies. 2Vrtature dengue virus particles consist of a single-stranded ribonucleic acid genome surrounded by an approximately icosahedral nucleocap- sid with a diameter of 30 nm. The nucleo- capsid is covered by a lipid envelope of 10 nm thickness derived from host cell mem- branes and contains the envelope and membrane proteins. 53

The viral genome of approximately 11 kb is infectious, has a messenger-like pos- itive polarity, and can be translated in vitro. The 5" end of the RNA has a type I cap structure but lacks a poly A tail at the 5" end. 54-56 It contains a si@e open read- ing frame of about 10,000 nucleotides en- coding three structural and seven non- structural proteins. The gene order is 5"- C-prM(M)-E-NS 1-NSYA-NSYB-

NS5-NS4A-NSdB-NS5. The proteins are synthesized as a polyprotein of about 5000 aminoacids that is processed co- translationally and posttranslationally by viral and host proteases. 57-66

The structural proteins include a capsid protein rich in arginine and lysine residues and a nonglycosylated prYi protein pro- duced from a glycosylated precursor in a late step of virus maturation. 54'55,67'68 The major structural envelope protein is in- volved in the main biologic functions of the virus particle such as cell tropism, acid- catalyzed membrane fusion, and the in- duction of hemagglutination-inhibiting, neutralizing, and protective antibodies. 69

The first nonstructural protein is NS1, a glycoprotein with a function in the virus life cycle that is unknown. 7° NS1 proteins are detected in high titers in patients with secondary dengue infections, but are rarely found in primary infectlons. 71 The NS2 region codes for two proteins (NSYA and NSYB) that are thought to be implicated in polyprotein processing; whereas NS5 is probably the viral pro- teinase that functions in the cytosol. 65'66'72 The NS4 region codes for two small hy- drophobic proteins that seem to be in- volved in the establishment of the mem- branebound RNA replication complex. The protein encoded by the NS5 gene has a molecular weight of 105,000 and is the most conserved flavivirus protein. On the

517

Page 3: 1-s2.0-S0022347697700544-main

KAUTNER, ROBINSON, AND KUHNLE THE JOURNAL OF PEDIATRICS OCTOBER 1997

F~. 2. Typical bright red rash of a dengue virus infection with clear areas scattered in between, the so- called "islands of white in a sea of red."

basis of the amino acid sequence, this pro- tein is believed to be the virus-encoded RNA-dependent RNA polymerase. 7a

CLINICAL AND LABORATORY MANIFESTATIONS

The clinical features of dengue virus in- fection vary from an asymptomatic infec- tion to a febrile "flulike" infection (DF) to the more severe DHF, which can lead to DSS. 74'76 This clinical variability is poor- ly understood and seems to be related to the age and sex of the individual but also to the immunologic and nutritional status. Dengue hemorrhagic fever is most likely to develop in immune-competent, well- nourished girls between the ages of 7 and 12 years. 76 Dengue viruses tend to cause a mild illness in the yew young infant, but DHF/DSS can develop in infants born to immune mothers as a result of waning passive maternal antibodies. 77 Dengue hemorrhagic fever is rare after the age of lb years.

The incubation period of DF after the mosquito bite (as well as the febrile peri- od) averages 4 days. Dengue fever may manifest with fever and a discrete macular or maculopapular rash. In this situation

the clinical differentiation from other viral illnesses may not be possible, recovery is rapid, and need for supportive treatment is minimal) In more severe DF, the tem- perature rises rapidly (frequendy reach- ing a9 ° c or higher) and persists for 5 to 6 days. Fever is characteristically biphasic and returns to almost normal in the mid- die of the febrile period giving rise to the saddleback temperature chart. It reaches its highest level during the last 24 hours before abatement. The patient is general- ly ill. Symptoms include headache and retroorbital pain, particularly when pres- sure is applied to the eyes ("Fire is coming out of my eyes"). Arthralgia, myalgia, and a macular or maculopapular rash may ap- pear at the onset. Some patients report se- vere backache (back-break fever), sore throat, or abdominal pain, which can be severe enough to be confused with appen- dicitis. The febrile period usually lasts up to 6 days during which time the rash may become diffusely erythematous with clear areas scattered in between, the so-called "islands of white in a sea of red" (Fig. 2). These patients are lethargic with accom- panying anorexia and nausea. Hepato- megaly can be present, although spleno- megaly is uncommon. Laboratory parameters are generally normal but platelets can be decreased and serum he- patic enzyme levels (alanine aminotrans-

ferase in particular) may be moderately elevated (rarely exceeding 100 IU/L). Differentiation from viral, certain bacteri- al infections, and Kawasaki disease can be difficult. Recovery from DF is usually complete in 7 to 10 days.

The incubation period for DHF is un- known but is probably similar to that of DE Dengue hemorrhagic fever com- mences acutely with high fever and many of the symptoms of DE However, drowsi- ness and lethargy tend to be more marked. In addition, there is increased vascular permeability and abnormal he- mostasis that can lead to hypovolemia and hypotension, and in severe cases, result in hypovolemic shock often complicated by severe internal bleeding.

The hemorrhagic manifestations appear usually by the third day and consist of scattered petechiae over the trunk, limbs, and axillae. These petechiae are associat- ed with, or may be preceded by a positive tourniquet test result. Bleeding at veni- puncture sites is the rule, and there may be hemorrhage from the gastrointestinal tract, nose, and gums. After 2 to 7 days, and as the fever begins to subside, signs of circulatory insufficiency can appear; the patient becomes restless and sweaty with cold extremities. Pleural effusion, charac- teristically on the right side, and abdomi- nal ascites may be noted. These features are almost diagnostic of DHF. With ap- propriate treatment this phase usually re- solves within 24 to 48 hours. 78'79

Dengue shock syndrome results from leakage of plasma into the extravascular compartment. Rapid and poor volume pulse, hypotension, cold extremities, and restlessness occur. In addition to the plas- ma leakage, which is the result of general- ized vasculitis, disseminated intravascular coagulation is present. Dengue shock syn- drome is usually a progression of D H E but there are patients who have circulato- ry insufficiency when first seen after a febrile illness of only short duration. 80

Increasingly neurologic manifestations are recognized as a separate entity and not as sequelae of hemorrhage, acidosis, or shock. Direct isolation of virus in the cerebrospinal fluid and brain tissue ap- pears to indicate infectious encephalitis. However, further clinical, pathologic, and

518

Page 4: 1-s2.0-S0022347697700544-main

THE JOURNAl OF PEDIATRICS Volume 13 I, Number 4

KAUTNER, ROBINSON,AND KUHNLE

autopsy studies are necessary to deter- mine the clinical relevance. 81-83

Laboratory investigations in D H F con- sistently reveal thrombocytopenia that may reach levels of 20,000 platelets/mm a or less. Depending on the hemoconcen- tration and severity of shock the hemat- ocrit can be increased by 20% or more. Commonly hypoalbuminemia, hypo- volemia, and moderately elevated serum aminotransferase and blood urea nitrogen levels can be documented. Partial throm- boplastin time and the thrombin time may be prolonged. 79 Hypofibrinogenemia and complement depletion correlate with the severity of the disease.

The pathologic findings in D H F are vasculifis of small vessels in soft tissues and viscera. Lymph gland hyperplasia and areas of necrosis in spleen, liver, and bone marrow are also noted as is maturation ar- rest of megakaryocytes./Vlacroscopically there are petechial hemorrhages, ecchy- moses, and effusions into pleural cavities and into the peritoneal cavity. 84

PATHOGENESIS

The pathogenesis of D H F is not clear; D H F is rare in early infancy and uncom- mon in adulthood. Infection with one dengue serotype provides lifelong homol- ogous immunity gut limited heterologous immunity. Almost aH patients with D H F have had previous experience with at least one of the four seroWpes of dengue virus- es, leading to the hypothesis that het- erotypic antibodies from a previous dengue virus infection promote increased viral replication within mononuclear leukocytes--the phenomenon of anti- body-dependent enhancement. 85 Further- more, immunologic processes aimed at eliminating dengue virusMnfected cells can result in the release of cytokines with vasoactive or procoagulant properties, the release of interferon-7, and the activation of complement. 86-88

Other hypotheses suggest that D H F re- sults from infection by a more virulent biotype of the virus or even from unfavor- able host factors such as concomitant bac- terial infection. Thus during the last Vene- zuelan hemorrhagic epidemic a dengue 2

subtype could be genetically identified as a Southeast Asian biotype, a9

Whether certain ethnic groups are more susceptible or resistant to the hemorrhag- ic manifestations of dengue is suggested from epidemiologic studies. As shown in Fig. 1, severe hemorrhagic disease is more common in Southeast Asia compared with Africa and America. In the 1981 dengue outbreak in Cuba it was observed that black individuals are relatively resis- tant to DHF/DSS and a "resistance gene" present in the African population has been speculated. 89

Regardless of the inciting cause, in- creased vascular permeability results in hemoconcentration, reduced blood vol- ume, poor tissue perfusion, tissue hypox- ia, and shock. The cause of bleeding in D H F appears to be complex and may in- volve one or more of the following: throm- bocytopenia, microvascular inju W, platelet dysfunction, and disseminated in- travascular coagulation. The mortality rate for D H F ranges from 1% to 30% de- pending on availability of supportive care.

DIAGNOSIS

The diagnosis of dengue relies in most cases on clinical judgment because only a few major centers have the facilities and means to perform and verify the clinical impression. Diagnostic criteria for DHS based on clinical observations have been proposed by the World Health Organ- izafion and should be used to avoid over- diagnosis. 90

The c[&ical cr#eria for diagnosis are as follows: (1) fever; (2) hemorrhagic mani- festations, including at least a positive tourniquet test result and a major or minor bleeding phenomenon; (3) hepatic enlargement; (4) shock (high pulse rate and narrowing of the pulse pressure to 20 mm Hg or less, or hypotension). The/ab- oratory criteria include (5) thrombocytope- nia (_<100,000/mm3), and (6) hemoeon- centration (hematocrit increase ->20%). Thrombocytopenia with concurrent high hematocrit levels differentiates D H F from classic DE

CurrenCy routine laborato W diagnosis of dengue infections depends on virus iso-

lation or the detection of dengue virus-specific antibodies. The isolation of viruses from clinical specimens can be carried out in cultured mosquito cells, such as AP-61 or C6/56 cell cultures. 91-94 When dengue virus serotype-specifie monoclonal antibodies are used, virus identification by indirect immunofluores- cence can be achieved within 2 weeks. 95'% The development of mosquito inoculation techniques has not only improved the sen- sitivity but also reduced the time required for virus isolation and identification. Parenteral inoculation of adult A. albapic- tua yields results in 7 days. 97"98 Virus iso- lation by intracerebral inoculation of Toxorbyncbitis spZendens mosquito or its fourth instar larvae can even be achieved within 2 to 5 days. 99'1°°

The serologic identification of the vari- ous types of dengue virus infection is com- plicated by the occurrence of cross-reac- tive antibodies to antigenic determinants shared by all four dengue viruses and other members of the flavivirus family. 1°1 The commonly used serologic test is the hemagglutination inhibition test) °2 In a prima W infection dengue hemagglutina- tion inhibition antibody titer is generally less than 1:20 in a sample collected within the first 4 days after the onset of symp- toms. In the convalescent phase sample (collected 1 to 4 weeks after the onset of symptoms) a fourfold or greater rise in antibody fiter is detected, with antibody titer _<1:1280.103

A seconda W dengue infection is charac- terized by the rapid appearance of broad- ly cross-reactive antibodies. Hemag- glutinafion inhibition fiters of 1:20 in the acute-phase sample rise to _>1:2560 in the convalescent phase sample. An antibody fiter of >1:1280 in the acute-phase sample without a fourfold or greater increase in the second sample also is considered pre- sumptive of recent infection. An improved and less time-consuming method is a cap- ture enzyme-linked immunosorbent assay that can detect specific anti-dengue IgM in a single acute-phase sample. 104

Recently commercial kits for the detec- tion of specific IgG as well as IgM anti- bodies have become available. They are based on a dot enzyme assay or a nitro- cellulose membrane-based capture for-

519

Page 5: 1-s2.0-S0022347697700544-main

KAUTNER, ROBINSON,AND KUHNLE THE JOURNAL OF PEDIATRICS OCTOBER 1997

mat, respectively, and should be suitable for field research. 105-107

An alternative to virus isolation is the detection of viral RNA by reverse tran- scription polymerase chain reaction. There are various protocols available using different primers and template iso- lation. 67'108113 Reverse transcription

polymerase chain reaction coupled with hybridization with labeled serotype-spe- cific probes can detect as few as 4 plaque-forming units per 100 btl serum and gives the best results early in the acute phase of the disease when dengue antibodies are still low. 112 Less than 1 ~tl of serum can be sufficient for the detec- tion of viral RNA. 114 Reverse transcrip- tion polymerase chain reaction is a high- ly sensitive technique of particular value in the early diagnosis of dengue infec- tion, but at present is only available in re- search settings.

THERAPY AND MANAGEMENT

The management of DF is supportive with bed rest, adequate fluid intake, and control of fever and pain with antipyretics and analgesics (e.g., paracetamol). Use of aspirin is contraindicated because of the increased bleeding tendency. Patients can be treated as outpatients if close monitor- hag is possible.

Differentiation between DF and D H F is difficult in the early stages and frequent measurements of the platelet count and hematocrit are necessary. A fall in the platelet count and a rising hemotocrit level indicate development of the shock syndrome. Narrowing of the pulse pres- sure and hypotension indicate shock, which must be treated as a medical emer- gency. The major problem in DHF/DSS is fluid loss rather than blood loss, and management must be directed toward maintenance of blood volume and blood pressure. Mild cases respond after 1 or 2 days of plasma expansion with isotonic or half isotonic saline solution.

Decreased levels of coagulation factors indicate disseminated intravascular coag- ulation; infusion of heparin and replace- ment of c o @ a t i o n factors may be re-

quired. Severe hemorrhage may require blood transfusion, preferably with fresh whole blood; concentrated platelet infu- sions may be required. Regular clinical and laboratory monitoring is mandatory. Re- versal of plasma leakage can be expected within 12 to 24 hours; intravenous infu- sion must be reduced because hypervo- lemia can lead to acute heart failure, 115-118

In some centers corticosteroids (methyl- prednisoloue) or carbazochrome sodium sulfonate (acona [AC-17]) are used to sta- bilize capillary permeability and reduce plasma leakage. A relatively large, placebo- controlled; double-blind study has not shown a positive effect from steroids in DSS, whereas a somewhat smaller and less stringent study claimed a beneficial ef- fect of AC-17 in D H E Whether this dif- ference is a direct drug effect or reflection of the different patient populations studied cannot be decided with the available data. Further studies that take into account the varying degrees of severity of D H F and DSS are indicated before a fmal conclu- sion can be drawn. 119'120

PREVENTION Mosquito control measures

As long as there is no vaccine available, the spread of DF and D H F can only be curtailed by controlling vector popula- tions. This approach was successfully started in the Americas at the beginning of this century, and eradication programs were started in 1947. The mosquitoes reemerged when specific programs aimed at their eradication were given lower pri- ority. In addition, insecticide resistance has been a problem in some areas. I21

In addition to insecticide application to control adult mosquito populations, breeding sites of Aedes mosquitoes should be reduced vigorously. Stagnant water and water containers should be avoided. Education programs must be implement- ed and legislation providing fines for those allowing Aedes breeding sites might be helpful. 122'1% Insect repellents are use- ful to prevent mosquito bites. However, it must not be forgotten that programs to eradicate mosquito vectors impose a great financial burden on developing countries and are ineffective in situations of open

conflict, lawlessness, and large-scale refugee migrations.

Vaccine Development Currently no vaccine is available to pro-

tect against dengue infections. The major concern in the development of a dengue vaccine is an immunologic response in the vaccinee that could lead to antlbody-de- pendent enhancement of infection and thus produce DHF/DSS. Candidate vac- cines based on live attenuated viruses should therefore contain all four serotypes to give comprehensive protection without adverse side effects.

In 1944 a serially attenuated, dengue 1 virus candidate vaccine prepared in mouse brain was tested in human volun- teers, but abandoned because the sub- strate was thought to be unsafe. 124-127 Recently live attenuated vaccines have been developed by multiple passages in primary clog kidney cells by researchers at the Mahidol University in Bangkok and candidate monovalent, bivalent, trivalent, and tetravalent vaccines are currently being tested in phase I and II trials with favorable results in Thailand. 128"131

Efforts to create a subunit vaccine by use of recombinant D N A technology have been initiated because of the concern of possible reversion of a live attenuated vaccine to a virulent strain. Recombinant proteins have beerl expressed in Escbericbia col~ as well as vaccinia viruses and insect cells (Spodo#era frugiperda) using engineered baculoviruses. 132-I43 Both the structural and the NS1 proteins have been shown to elicit protective im- munity in monkeys and mice and thus the development of safe vaccines appears to be feasible. 133-135'157'159'141

REFERENCES 1. Halstead SB. Dengue. In Warren KS,

Mahmoud AAE editors. Tropical and geo- graphical medicine. New York: McGraw- Hill; 1990. p. 675-85.

2. Hanson SM. Field overwinter survivorship of Aedes albopictus eggs in Japan. J Am Mosq Control Assoc 1995:11:554-57.

3. Hanson SM, Craig GB. Cold acclimation, diapause and geographic origin affect cold hardiness in eggs of Aedes albofictus

520

Page 6: 1-s2.0-S0022347697700544-main

THE JOURNAL OF PEDIATRICS Volume 13 I, Number 4

KAUTNER, ROBINSON, AND KUHNLE

(Diptera: Culiddae). J Med Entomo11994; 31:192-201.

4. Grist NR, Burgess NRH. Aedes and dengue [letter]. Lancet 1994;343:477.

5. Halstead SB. The XXth century dengue pandemic: need for surveillance and re- search. Pediatr Update 1995;6:16-21.

6. Monath TE Dengue: the risk to developed and developing countries. Proc Nail Acad Sci U S A 1994;91:2395-400.

7. Schlesinger RW. Dengue viruses. New York: Springer Verlag; 1977.

8. Siler dE Hall MW, Hitchens AP. Dengue: its history, epldemiology, mechanism of transmission, etiology, clinical manifesta- tions, immunity and prevention. Philip J Sci 1926;29:1-304.

9. Sabin AB. Dengue. In: Rivers T, Horsfall E editors. Viral and rickettsial infections of man. Philadelphia: JB Lippineott; 1959. p. 361-73.

10. Gratz NG. Emergency control ofAedes ae- gypti as a disease vector in urban areas. J Am Mosq Control Assoc 1991;7:353-65.

11. Maung LW, 'Tin E Thein M, Wm S. De- scilpdve epidemiology of dengue haemor- rhagic fever in Yanguon division, Myanmar. Dengue Newsletter 1992; 17:9-19.

12. Halstead SB. Dengue and haemorrhagic fevers of South and Southeast Asia: with discussion by Max Theiler. Yale d Biol Med 1965;37:434-54.

13. World Health Organization. DHF situation and activities in the WHO South-East Asia region in 1988. Weekly Epidemiol Rec 1988;64:175-6.

14. Qiu FX, Chen QQ, Ho QY, Chen WZ, Zhao ZG, Zhao BW The first epidemic of dengue hemorrhagic fever in the Peoples Republic of China. Am J Trop Med Hyg 1991;44:364-70.

15. Srivastava VK, Suri S, Bhasin A, Srivastava L, Bharadwaj M. An epidemic of dengue haemorrhagic fever and dengue shock syndrome in Delhi: a clinical study. Ann Trop Paediatr 1990;10:329-34.

16. Laille M, Huerre M, Flye Saints Marie E An epidemic of dengue in 1989 in New Caledonia. Proceedings of the International Symposium of Dengue and Dengue Haem- orrhagic Fever; Oct t-3, 1990; Bangkok, Thailand.

17. Chungue E, Speigel A, Philippon G, Landon E Cardines E, Roux J. Dengue and dengue haemorrhagic fever in French Polynesia: a comparative study of two suc- cessive epidemics. Proceedings of the International Symposium on Dengue and Dengue Haemorrhagic Fever; Oct 1-3, 1990; Bangkok, Thailand.

18. Vitarana T, JayasekasaN. Dengue haemor- rhagic fever outbreak in Sri Lanka. Southeast Asian J Trop Med Public Health 1990;21:682-4.

19. Guzman MG, Kouri GP, Bravo J, Soler M, Vasquez S, Santos M, et al. Dengue haem-

orrhagic fever in Cuba. II. Clinical investi- gations. Trans R Soc Trop Med Hyg 1984;78:239-41.

20. Kouri G, Mas P, Guzman MG, Soler M, Goyenechea A, Moiler L. Dengue haemor- rhagic fever in Cuba, 1981: rapid diagnosis of the etiologic agent. Bull Pan Am Health Organ 1983;17:126-32.

21. Fiqueiredo LTM, Calvacante SMB, Simoes MC. Dengue serologic survey of school children in Rio de Janeiro, Brazil in 1986 and 1987. Bull Pan Am Health Organ 1990;24:217-25.

22. Pontes R J, Dat Fabro AL, Roeha G, Santiago RC, Fiquelredo LT, Castro e Silva de MAA, et al. Dengue epidemic in Ribeirao Pretu, SP, Brazil: a preliminary note. Rev Saude Publica 1991;25:315-7.

23. Herrera-Basto E, Prevots DR, Zarate NIL, Silva JL, Sepulveda-Amor J. First report- ed outbreak of Classical dengue fever at 1,700 meters above sea level in Guerrero State, Mexico, June, 1988. Am J Trop Med Hyg 1992;46:649-53.

24. Colan E. Dengue epidemic Peru, 1990. MMWR Morb Mortal Wkly Pep 1991; 40:145-7.

25. Anonymous. Dengue haemorrhagic fever in Venenzuela. Epidemiol Bull Am Health Organ 1990;1l:7.

26. Lange WR, Beall B, Denny SC. Dengue fever: a resurgent risk for the international traveler. Am Faro Physician 1992;45:1161-8.

27. Mills GD, Jones PD. Clinical spec~um of dengue fever in travelers. N Z Med J 1991;104:228-50.

28. Ligtenberg J J, Hospers GA, Sprenger HGi Welts J. Haemorrhagic fever caused by dengue in two tourists. Ned Tijdschr Geneeskd 199l;155:2594-7.

29. Cunningham R, Mutton K. Dengue haem- orrhagic fever. Br Med d 1991;302:1083-4.

30. Krippner R, Hanisch G, Kretschmer H. Denguefieber mit haemorrhagischen Mani- festationen nach Thailandanfenthalt. Dtsch Med Wochenschr 1990;115:858-62.

31. Hasler C, Schnorf H, Enderlin N, Gyr K. Importiertes Dengue-Fieber nach einem Tropenaufenthalt. Schweiz Med Wochen- schr 1993; 125:120-4.

32. Anonymous. Imported dengue~United States. MMWR Morb Mortal Wkly Rep 1991;40:519-20.

33. Rosen L, Rozeboom LE, Sweet BH, Sabin AB. The transmission of dengue by Ae. po[y- neslensis. Am J Trop Med Hyg 1954; 3:878-82.

34. Rosen L. The global importance and epi- demiology of dengue infection and disease. In: Pang T, Pathmanathan R, editors. Proceedings of the International Con- ference on Dengue/DHE Sept 1-3, 1982. Kuala Lumpur: University of Malaya; 1983. p. 519.

35. Nasidi A, Monath TP, De Cock K, Tomori O, Cordelier R, Olaleye OO, et al. Urban

yellow fever epidemic in Western Nigeria, 1987. Trans Soc Trop Med Hyg 1989; 83:401-6.

36. Simmons aS, St Johns JH, Reynolds FHK. Experimental studies of dengue. Philip J Sci 1931;44:1-251.

37. Hammon WM, Rudnick A, Slather GE. V'truses associated with epidemic haemor- rhagic fevers of the Phillipines and Thailand. Science 1960;151:1102-3.

38. Gould DJ, Yuill TM, Moussa MA, Simasathien P, Rutledge LC. An insular outbreak of dengue haemorrhagic fever. IIL Identification of vectors and observations on vector ecology. Am J Trop Med Hyg 1968;17:609-18.

39. Rico-Hesse R. Molecular evolution and dis- tribution of dengue viruses type 1 and 2 in nature. V'wology 1990; 174:479-93.

40. Laird AM, editor. Commerce and spread of pests and disease vectors. New York: Praeger; 1984.

41. Halstead SB. The pathogenesis of dengue. Molecular epidemiology in infectious dis- ease [Alexander D. Langmuir lecture]. Am d Epidemiol 1984; 114:632-48.

42. Service MW Review: importance of ecolo- gy in Aedes aegypti control. Southeast Asian J Trop Med Public Health 1992;25:681-90.

43. Halstead SB. Selective primary health care: Strategies for control of disease in the de- veloping world. XI. Dengue. Rev Infect Dis 1984;6:251-64.

44. Fox I, Specht E Evaluating ultra-low vol- ume ground applications of malathion against Aedes aegyptl using landing counts in Puerto Rico, 1980-1986. J Am Mosq Control Assoc 1988;4:165-7.

45. Gubler D J, Rosen L. A simple technique for demonstrating transmission of dengue virus by mosquitoes without the use of ver- tebrate hosts. Am J Trop Med Hyg 1976; 25:146-50.

46. Khin MM, Than KA. Transovarian trans- mission of dengue 2 viruses by Aedes aegypti in nature. Am d Trop Med Hyg 1985; 32:590-4.

47. Hull B, Tikasingh E, De SouzaM, Maz6nez R. Natural transovarial transmission of dengue 4 virus in Aedes aegypti in Trinidad. Am J Trop Med Hyg 1984;33:1248-5.

48. Cheong LL. The vectors of dengue and dengue haemorrhagic fever in Malaysia. In: Rudnick A, Lin TW,, editors. Dengue fever studies in Malaysia, bulletin 23. Kuala Lumpur, Malaysia: Institute of Medical Research; 1986.

49. Lira JL. Oviposition periodicitis, landing and biting frequencies and the use of ovi- traps to controlAedes albopictus (Skuse) [dis- sertation]. Kuala Lumpur, Malaysia: Insti- tute of Medical Research; 1979.

50. Anonymous. New mosquito strain for Nigeria [letter]. Lancet 1990;339:1048.

51. Gilst NR.Aedes a[bopictus: the tyre-travelling tiger. J Infect 1993;27:1-4.

521

Page 7: 1-s2.0-S0022347697700544-main

KAUTNER, ROBINSON,AND KUHNLE THE JOURNAL OF PEDIATRICS OCTOBER 1997

52. Ward/VIA, Burgess NRH. Aedea albopictus; a new disease vector in Europe? J R Army Med Corps 1993;139:109-11.

53. Westaway EG, Brinton MA, Gaimamovich S, Horzink MC, Igarashi A, Kaariainen L, et al. Flaviridiae. Intervirology 1985;24: 183-92.

54. Rice CM, Lenches EM, Eddy SR, Shin S J, Sheets RL, Strauss JH. Nucleotide se- quence of yellow fever virus: implications for flavivirus gene expression and evolu- tion. Science 1985;229:726-33.

55. Hahn YS, Galler R, Hunkapillar T, Dalrymple JM, Strauss JH, Strauss EG. Nucleotide sequence of dengue-2 RNA and comparison of the encoded proteins with those of other flaviviruses. ~trology 1988; 162:167-80.

56. Irie K, Mohan PM, Sasaguri Y, Putnak R, Padmanabhan R. Sequence analysis of cloned dengue virus type-2 genome (New Guinea-C strain). Gene 1989;74:197-211.

57. Biedrzycka A, Canchi MR, Bartholomensz A, Gorman dd, Wright PJ. Character- ization of protease cleavage sites involved in the formation of the envelope glycoprotein and three nonstructural proteins of dengue type-2, New Guinea C strain. J Gen Virol 1987;68:1317-26.

58. Mackow E, Makino Y, Zhoa B, Zhang YM, Markoff L, Buckler-White A, et al. The nu- deotide sequence of dengue type 4 virus: analysis of the genes encoding for nonstruc- tural proteins NSYA, NSYB, NS3, NS4B and NS5 of the flavivirus Kunjin and their cleavage sites. J Gen Virol 1987;69:23-4.

59. Speight G, Cola G, Parker MD, Westaway EG. Gene mapping and positive identifica- tion of nonstruetural proteins NSYA, NSYB, NS3, NS4B and NS5 of the fla- vivirus Kunjin and their cleavage sites. J Gen Virol 1988;69:23-34.

60. Chambers T J, Mccourt DW, Rice CM. Yellow fever virus proteins NSYA, NSYB and NS4B: identification and partial N- ter- minal amino acid sequence analysis. V'tro- logy 1989;169:100-9.

61. Markoff L. In vitro processing of dengue virus structural proteins. J Virol 1989;63: 3345-52.

62. Preugschat E Yao CW, Strauss JH. In vitro processing of dengue virus type 2 nonstruc- rural proteins NSYA, NSYB and NS3. J Virol 1990;64:4364-74.

63. Falgout B, Pethel M, Zhang YM, Lal CO. Both nonstructural proteins NSYB and NS3 are required for the proteolytic pro- cessing of dengue virus nonstructural pro- teins. J Virol 1991;65:2467-75.

64. Preugschat E Lenches EM, Strauss JH. Flavivirus enzyme-substrate interactions studied with chimeric proteinases: identifi- cation of an intragcnic locus important for substrate recognition. J Virol 1991;65: 4749-58.

65. Preugschat E Strauss JH: Processing of

nonstructural proteins NS4A and NS4B of dengue 2 viruses in vitro and in vivo. Viro- logy 1991; 185:689-97.

66. Cahour A, Falgout B, Lai CO. Cleavage of the dengue virus polyprotein at the NS3/ NS4A and NS4B/NS5 junctions is mediat- ed by viral protease NSYB-NS3, whereas NS4A/NS4B may be processed by a cellu- lar protease. J Virol 1992;66:1535-42.

67. Deubel V, Laille M, Hugnot J, Chungue E, Guesdon J, Drouet MT, et al. Identification of dengue sequences by genomic amplifica- tion: rapid diagnosis of dengue virus serotypes in peripheral blood. J Virol Methods 1990;30:41-54.

68. Randolph VB, Winkler G, Stollar V. Acidotrophic amines inhibit proteolytic pro- cessing of flavivirus prM protein. Virology 1990; 174:450-8.

69. Depr~s E Frenkiel MP, Deubel V. Differ- ences between cell membrane fusion activi- ties of two dengue type-1 isolates reflect modifications of viral structure. Virology 1993;196:209-19.

70. Sehlesinger JJ, Brandiss MW, Walsh EE. Protection against 17D yellow fever en- cephalitis in mice by passive transfer of MeAbs to the nonstructural glycoprotein gp48 and by active immunization with gp48. J Immunol 1985;135:2805-9.

71. Kuno G, Vorndam AV, Gubler DJ, Gomez I. Study of anti-dengue NS1 antibody by Western Blot. J Med Virol 1990;32:102-8.

72. Falgout B, Chanock R, Lai CO. Proper pro- eessing of dengue virus nonstructural glyco- protein NS1 requires the N-terminal hydro- phobic signal sequence and the downstream nonstructural protein NSYa. J Virol 1989;63:1852-60.

73. Rice CM, Strauss EG, Strauss JH. Structure of the flavivirus genome. In: Schlesinger S, Schlesinger M, editors. The togaviridae and flaviviridae. New York: Plenum; 1986.

74. Halstead SB. Dengue haemorrhagic fever: a public health problem and a field for re- search. Bull World Health Organ 1980; 58:1-21.

75. Hayes EB, Gubler DJ. Dengue and dengue hemorrhagic fever. Pediatr Infect Dis J 1992;11:311-7.

76. Halstead SB, Nimmannitya S, Cohen SN. Observations related to pathogenesis of dengue hemorrhagic fever, IV, relation of disease severity to antibody response and virus recovered. Yale J Biol Med 1979;42: 311-28.

77. Kliks SC, Nimmanitya S, Nisalak A, Burke DS. Evidence that maternal dengue anti- bodies are important in the development of dengue hemorrhagic fever in infants. Am J Trop Med Hyg 1988;38:411-9.

78. Hathirat P, Isarangkura P, Sriehaikul T, Suvatte V, Mitrakul C. Abnormal hemostasis in dengue hemorrhagic fever. Southeast Asian J Trop Ivied Public Health 1993;24:80-4.

79. Mary N, Molanida CR, Guzman M, Laberuza E Prothrombin time and partial thromboplastin time as a predictor of bleed- ing in patients with dengue hemorrhagic fever. Southeast Asian J Trop Med Public Health 1993;24:141-31

80. Lain SK. Dengue and dengue hemorrhagic fever. Southeast Asian J Trop Med Public Health 1990;21:520-1.

81. Sumarmo, Wulur H, Jahja E, Gubler D J, Sutomenggolo TS, Saroso JS. Encephalop- athy associated with dengue infection. Lancet 1978;1:449-50.

82. Lam SK. Dengue infections with central nervous system manifestations. Neurol J Southeast Asia 1996; 1:3-5.

83. Renganathan A, Ng WK, Tan CT. Trans- verse myelitis in association with dengue infection. Neurol J Southeast Asia 1996; 1:61-3.

84. Lum LC, Lam SK, George R, Devi S. Fulminant hepatitis in dengue infection. Southeast Asian J Trop Med Public Health 1993;24:467-71.

85. Halstead SB. Pathogenesis of dengue: chal- lenge to molecular biology. Science 1988; 239:476-81.

86. Cobra C, Rigau-Perez JG, Kuno G, Vorndam V. Symptoms of dengue fever in relation to host immunologic response and virus serotype, Puerto Rico, 1990-1991. Am J Epidemiol 1995;142;1204-11.

87. Wang S, He R, Patarapotikul J, Innis BL, Anderson I~ Antibody-enhanced binding of dengue-2 virus to human platelets. V'trology 1995;213:254-7.

88. Mukerjee R, Chamrvedi UC, Dhawan R. Dengue virus induced human cytotoxie fac- tor: production by peripheral blood leuko- cytes in vitro. Clin Exp Immunol 1995;102: 262-7.

89. Guzman MG. Dengue haemorrhagie fever in Cuba, 1981: a retrospective seroepidemi- ologic study. Am 3 Trop Med Hygiene 1981;42:179-84.

90. World Health Organization. Dengue haem- orrhagic fever: diagnosis, treatment and control. Geneva: WHO; 1986.

91. Varma MGR, Pudney M, Leake CJ. Cell lines from larvae of Aedes (Stegomyia) malayensis (Colless) and Aeries (S.) pseudos- cuMlaris (Theobald) and their infection with some arboviruses. Trans R Soc Trop Med Hyg 1974;68:374-82.

92. Igarashi A. Isolation of a Singh's Aedes al- bapictus cell clone sensitive to dengue and ehikungunya viruses, d Gen Virol 1978;40: 531-44.

93. Race MW,, Agostini CFN, Fortune RAJ, Varma MGR. Isolation of dengue viruses in mosquito cell cultures under field condi- tions. Lancet 1978;1:48-9.

94. Tesh RB. A method for the isolation and identification of dengue viruses using mos- quito cell cultures. Am J Trop Med Hyg 1979;28:1053-9.

522

Page 8: 1-s2.0-S0022347697700544-main

THE JOURNAL OF PEDIATRICS Volume 13 I, Number 4

KAUTNER, ROBINSON,AND KUHNLE

95. Henchal EA, Gentry MK, McCown JM, Brandt WE. Dengue virus-specific and fla- vivirus group determinants identified with monoclonal antibodies by indirect immuno- fluorescence. Am J Trop Med Hyg 1982; 31:830-6.

96. Henchal EA, McCown JM, Seguin MC, Gentry MK, Bran& WE. Rapid identifica- tion of dengue virus isolates by using mon- oclonal antibodies in an indirect immuno- fluorescence assay. Am J Trop Med Hyg 1983;32:164-9.

97. Rosen L, Gubler E The use of mosquitoes to detect and propagate dengue viruses. Am J Trop Med Hyg 1974;23:1153-60.

98. Thet-Win. Detection of dengue virus by im- munofluorescence after intracerebral inocu- lation of mosquitoes. Lancet 1982;1:53-4.

99. Pang T, Lain SK, Chew CB, Poon GK, Ramalingam S. Detection of dengue virus by immunofluorescence following inocula- tion of mosquito larvae. Lancet 1983; I:1271.

100. Lain SK, Chew CB, Poon GK, Rama- lingam S, Scow SC, Pang T. Isolation of dengue viruses by intracerebral inoculation of mosquito larvae. J Viroi Methods 1986; 14:133-40.

101. Innis BL, Nisalak A, Nimmannitya S, Kusalerdchariya S, Chongwasdi V, Suntayakorn S, et al. An enzyme-linked im- munosorbent assay to characterize dengue infections where dengue and Japanese en- cephalitis co-circulate. Am J Trop Med Hyg 1989;40:418-27.

102. Clarke DH, Casals J. Techniques for hemagglutinafion and hemagglutination-in- hibition with arthropod-borne viruses. Am J Trop Med Hyg 1958;7:561-73.

103. Gubler DJ. Dengue, In: Monath TP, editor. The arboviruses: epidemiolos;y and ecology vol 2. Boca Raton (FL): CRC Press; 1984. p. 223-60.

104. Lain SK, Devi S, Pang T. Detection of spe- cific IgM in dengue infection. Southeast Asian J Trop Med Public Health 1987;18: 532-8.

105. Cardosa M J, Tio PH, Noorsham ??. Development of a clot enzyme assay for dengue 3: a sensitive method for the detec- tion of anti-dengue antibodies. J Virol Methods 1988;22:81-8.

106. Cardosa M J, Noorsham S, Tin PH, Lira SS. A dot enzyme immunoassay for dengue 3 virus: comparison with haemagglutination inhibition test. Southeast Asian J Trop Med Public Health 1988;19:591-4.

107. Cardosa M J, Baharudin E Hamid S, Tio PH, Nimmanitya S. A nitrocellulose mem- brane based IgM capture enzyme im- munoassay for etiological diagnosis of dengue virus infections. Clin Diagu Virol 1995;3:343-50.

108. Henchal EA, Polo SL, Vorndam V, Yaemsiri C, Innis BL, Hoke CH. Sensitivity and specificity of a specific primer set for

the rapid diagnosis of dengue virus infec- tions by polymerase chain reaction and nu- cleic acid hybridization. Am J Trop ivied Hyg 1991;45:418-28.

109. Morita K, Tanaka M, Igarahi A. Rapid identification of dengue virus serotypes by using polymerase chain reaction, d Clin Microbinl 1991;29:2107-10.

110. Morita K, Maemotot T, Honda S, Onishi K, Murata iV[, Tanaka M, et al. Rapid detec- tion of virus genome from imported dengue fever and dengue hemorrhagic fever pa- tients by direct polyrnerase chain reaction. J Med Virol 1994;44:54-8.

111. Lanciotti RS, Calisher CH, Gubler D J, Chang G J, Vorndam AV. Rapid detection and typing of dengue viruses from clinical samples by using reverse transcriptase- polymerase chain reaction. J Clin Micro- biol 1992;30:545-51.

112. Chan SY, Kautner IM, Lam SK. Detection and serotyping of dengue viruses by PCR: a simple and rapid method for the isolation of viral RNA from infected mosquito larvae. Southeast Asian J Trop Med Public Health 1994;25:258-61.

113. Seah CL, Chow VT, Tan HC, Chan YC. Rapid, single-step RT-PCR typing of dengue viruses using five NS3 gene primers. J Virol Methods 1995;51:193-200.

114. Chan SY, Kautner IM, Lain SK. The influ- ence of antibody levels in dengue diagnosis by poIymerase chain reaction. J Virol Methods 1994;49:315-22.

115. Nimmafiya S. Clinical spectrum and man- agement of dengue hemorrhagic fever. Southeast Asian J Trop Med Public Health 1987;24:398-406.

116. Butthep P, Bunyaratvej A, Bhamara- pratvati N. Dengue virus and endothelial cell: a related phenomena to thrombocy- topenia and granulocytopenia in dengue he- morrhagic fever. Southeast Asian J Trop Med Public Health 1993;24:245-8.

117. Isarankura P, Tuchinda S. The behaviour of transfused platelets in dengue hemorrhagic fever. Southeast Asian J Trop Med Public Health 1993;24:222-4.

118. Kurane I, Rothman AL, Livingstone PG, Green S, Gagnon S J, Janus J, et al. Immunopathologie mechanisms of dengue hemorrhagic fever and dengue shock syn- drome. Arch Virol 1994;suppl 9:59-64.

119. Tassniyom S, Vasanawathana S, Chirawat- kui A, Rojanasuphot S. Failure of high-dose methylprednisolone in established dengue shock syndrome: a placebo-controlled, dou- ble-blind study. Pediatrics 1993;92:111-5.

120. Funahara Y, Shirahata A, Harun SR, Setiabudy-Dharma R, Nathin MA, Karjomanggolo TW, et al. Protection against marked plasma leakage in dengue haemorrhagic fever by infusion of car- bazochrome sodium sulfonate (AC-17). Southeast Asian J Trop Med Public Health 1987; 18:356-9.

121. Gubler DJ. Aedes aegypti and Aedes aegypti- borne disease control in the 1990's: top clown or bottom up. Am J Trop IVied Hyg 1989;40:57i-8.

122. Lira SE. National control programme for malaria and dengue in Malaysia./Vial d Lab Sci 1992;9:34-41.

123. Anonymous. Vgorld malaria situation in 1989, part 1. Wkly Epidemiol Rec 1991; 66:157-63.

124. Sabin AB, Sehlesinger RW. Production of immunity to dengue with virus modified by propagation in mice. Science 1945; I01:640-2.

125. Schlesinger RW. Propagation in chick em- bryos of the Hawaiian strain of dengue virus, I, sustained serial passage in eggs after one hundred intraeerebral passages in mice. Am d Hyg 1950;51:248-52.

126. Schlesinger RW. Propagation in chick em- bryos of dengue virus, Hawaiian strain, II, findings in infected eggs. Proe Soe Exp Biol Med 1951;76:817-23.

127. Wisseman CL, Sweet BH, Rosenzweig EC, Eylar OR. Attenuated living type 1 dengue vaccines. Am J Trop Med Hyg 1963;12: 620-3.

128. Angsubhakorn S, Yoksan S, Pradermwong A, Nitatpattana N, Sahaphong S, Bhamarapravati N. Dengue-3 (16562) PGMK 33 vaccine: neurovirulence, viremia and immune response in zYIacacafascicu/,aris. Southeast Asian J Trop Med Pub Hlth 1994;25:554-9.

129. Ansubhakorn S, Yoksan S, Bhamarapravati N, Moe JB, Marchette N J, Praderrnwong A, et al. Dengue-4 vaccine: neurovirulence, viremia and immune responses in rhesus and cynomolgus monkeys. Trans R Soc Trop Med Hyg 1988;82:746-9.

130. Bhamarapravati N, Yoksan S, Chayaniya- yothin T, Angsubphakorn S, Bunyaratvej A. Immunization with a live attenuated dengue-2 virus candidate vaccine (16681- PDK 53): clinical, immunological and bio- logical responses in adult volunteers. Bull World Health Organ 1987;65:189-95.

131. Bhamarapravati N, Yoksan S. Study of bi- valent dengue vaccine in volunteers. Lancet 1989;1:1077.

132. Eckels KH, Dubois DR, Summers PL, Schlesinger J J, Shelley M, Cohen S, et al. Immunization of monkeys with baculovlnas dengue type-4 recombinants containing en- velope ancl nonstructural proteins: evidence of priming and partial protection. Am J Trop Med Hyg 1994;50:472-8.

133. Bray M, Lai C,,J. Dengue virus premem- brahe and membrane proteins elicit a pro- tective immune response. Virology 1991; 185:505-8.

134. Delenda C, Frenkiei ME Deubel V. Protective efficacy in mice of a secreted form of recombinant dengue-2 virus enve- lope protein produced in baeulovirus infect- ed cells. Arch V~roI 1994; 139:197-207.

523

Page 9: 1-s2.0-S0022347697700544-main

KAUTNER, ROBINSON, AND KUHNLE THE JOURNAL OF PEDIATRICS OCTOBER 1997

135. Men RH, Bray M, Lai C,J. Carboxy-terml- nally mmcated dengue virus envelope glyco- proteins expressed on the cell surface and secreted extracellularly exhibit increased im- munogenicityin mice. J Vtroi 1991;65:1400-7.

136. Putnak JR, Charles PC, Padmanabhan R, Irie K, Hoke CH, Burke DS. Functional and antigenic domains of dengue 2 non- structural glyeoprotein NS 1. Virolos;y 1988; 163:93-103.

137. Bray M, Zhao BT, Markoff L, Eckels KH, Chanock RM, Lai CJ. Mice immunized with recombinant vaccinia virus expressing dengue 4 virus structural proteins with or without nonstmctural protein NS 1 are pro- tected against fatal virus encephalitis, d Virol 1989;63:2853-6.

138. Zhao BT, Prince G, Horswood R, Eckels K, Summers PL, Chanock RM, et al. Expres- sion of dengue virus structural proteins and nonstructural protein NS1 by a recombi- nant vaceinia virus. J Viro11987; 1:4019-22.

139. Zhang YM, Hayes EP, McCarty TC, Dubois DR, Summers PL, Eckels KH, et al. Immunization of mice with dengue struc- tural proteins and nonstructural proteins NS 1 expressed by baculovirus recombinant induce resistance to dengue virus encephali- tis. J V'wol 1988;62:3027-31.

140. Mason PW, Zugel MU, Semproni AR, Fournier M J, Mason TL. The antigenic structure of dengue type 1 virus envelope and NS1 proteins expressed in E. coli. J Gen Vn'ol 1990;71:2107-114.

141. Putnak R, Feighny R, Burros J, Cochran M, Hackett C, Smith G, et al. Dengue-1 virus envelope glycoprotein gene expres- sion in recombinant baculovirus elicits virus neutralizing antibody in mice and protects them from virus challenge. Am J Trop Med Hyg 1991;45:159-67.

142. Qu FX, Chen QO~ Ho QY, Clien WZ, Zhao ZG, Zhao BW. The first epidemic of dengue haemorrhagic fever in the People's Republic of China. Am J Trop Hyg 1991; 44:364-70.

143. HahnYS, Lenches EM, Galler R, Rice CM, Dalrymple JM, Strauss JH. Expression of the structural proteins of dengue 2 virus and yellow fever virus by recombinant vaccinia

• viruses. Arch Vlrol 1990;115:167-80.

50 Years Ago in The Journal of Pediatrics RHEUMATIC FEVER RECURRENCE IN CHILDREN WITHOUT SULFONAMIDE PROPHYLAXIS: AN EVALUATION OF ENVIRONMENTAL FACTORS

Jackson RL, Kelly HG, Ro/~ret CH, Duane JM. J Pediatr 1947,'31:390-402

Fifty years ago in rural Iowa, just as in urban American settings, rheumatic fever (RF) was a threat for indivt~- ual patients and a major public health problem for schoolchildren. Hospitals were fdled with patients with RF and rheumatic heart disease, and there were numerous reports describing the high incidence of RF and prevalence of rheumatic heart disease. In contrast, half a century later, the reduction in RF incidence in industrialized countries is striking. Yet in most of the developing world, the disease remains at a level comparable to that alluded to by Jackson and colleagues. The perceptive article by Jackson et al. described the relationship between recurrences of RF and suboptimal social, economic, and nutritional status. The decline in incidence of RF in industrialized countries began before the clinical introduction of antibiotics; this reduction has historically been associated with improving social and economic factors. Yet during the past decade, a "resurgence" of RF has been described in North American middle class suburban children with ready access to medical care and in military recruits. These

published observations suggest that factors other than social and economic status are equally important. "Rheumatogenic" potential and the virulence of specific strains of group A streptococci cannot be ignored. It was not possible to study such variables in the laboratory 50 years ago. Thus the relationship between social, econom- ic, and nutritional status and microbiologic and immunologic factors could not be compared at the time when the disease was so common. Maybe the fact that the pathogenesis of this unique disease remains incompletely defined

is a consequence. Antibiotics and better living conditions are clearly not the complete solution to controlling group A streptococ-

cal infections and their complications. One attractive theoretical approach to conquering this major cardiovascu- lar disease would be development of a cost-effective vaccine. Perhaps this can be realized in the not too distant fu- ture. Then it is possible that the incidence of RF and the prevalence of rheumatic heart disease, sometimes referred to as the most preventable of all cardiovascular diseases, could be significantly reduced. Despite a need to improve social and economic status of at-risk pediatric populations for many health-related reasons, time-tested techniques of accurate diagnosis and appropriate antibiotic therapy for the preceding streptococcal infection will continue to be the most practical and effective methods of control.

Ed,~ard L. Kaplan, ~ID, and James H. Ztloller, ~/ID Department of Pediatrics University of Minnesota

Minneapolis, M N 55455

524


Recommended