Você está na página 1de 24

27 Herpes Simplex Virus

Infections
DAVID W. KIMBERLIN and KATHLEEN M. GUTIERREZ

CHAPTER OUTLINE Herpes Simplex Virus Treatment


Structure Background
Replication Antiviral Drugs
Latency and Reactivation Other Issues in Acute Management
Epidemiology and Transmission Long-Term Management of Infected Infants
Maternal Infection Prevention
Factors Influencing Transmission of Background
­Infection to the Fetus Management of Pregnant Women with
Incidence of Newborn Infection Known Genital Herpes
Times of Transmission of Infection Management of Infants of Mothers with
Immunologic Response Genital Herpes
Neonatal Infection Conclusion
Pathogenesis and Pathology
Clinical Manifestations
Diagnosis
Clinical Evaluation
Laboratory Assessment

Neonatal herpes simplex virus infection (HSV) was iden- development of antiviral therapy represented a significant
tified as a distinct disease in the 1930s. The first written advance in the management of infected children and has
descriptions of neonatal HSV were attributed to Hass, who substantially decreased the morbidity and mortality asso-
described the histopathologic findings of a fatal case, and ciated with neonatal HSV infections. Neonatal HSV infec-
to Batignani, who described a newborn child with HSV tion is more amenable to prevention and treatment than
keratitis,1,2 During the initial decades that followed, our many other viral and bacterial diseases affecting neonates
understanding of neonatal HSV infections was based on because it is acquired most often at birth rather than in
histopathologic descriptions of the disease, which indicated utero. Postnatal acquisition of HSV-1 has been documented
a broad spectrum of organ involvement in infants. from nonmaternal sources, and more cases of genital her-
An important scientific breakthrough occurred in the pes caused by HSV-1 have been identified more recently,
mid-1960s, when Nahmias and Dowdle3 demonstrated including HSV-1 in the maternal genital tract from changes
two antigenic types of HSV. The development of viral typing in sexual practices that result in increased likelihood of oral-
methods provided the tools required to clarify the epidemiol- genital contact. This evolving natural history of genital
ogy of these infections. HSV infection “above the belt,” pri- herpetic infection has the potential to impact the incidence
marily of the lip and oropharynx, was found in most cases and outcomes of neonatal HSV infections. Perspectives on
to be caused by HSV type 1 (HSV-1). Infections “below the the changing presentations of neonatal HSV infection, the
belt,” particularly genital infections, were usually caused by obstacles to diagnosis, and the value of antiviral therapy are
HSV type 2 (HSV-2). The finding in the 1960s and 1970s addressed in this chapter.
that genital HSV infections and neonatal HSV infections
were most often caused by HSV-2 suggested a cause and
effect relationship between these two entities. This causal
relationship was strengthened by detection of the virus in
Herpes Simplex Virus
the maternal genital tract at the time of delivery, indicating STRUCTURE
that acquisition of the virus occurs by contact with infected
genital secretions during birth. Herpes simplex virus types 1 and 2 are members of the
Knowledge of HSV structure and function, epidemiology, large family of herpesviruses.4 Other human herpesviruses
and natural history, and of the pathogenesis of neonatal include cytomegalovirus, varicella-zoster virus, Epstein-
HSV infection, has increased during the past 5 decades.4 The Barr virus, and human herpesviruses 6 (including variants
843
844 SECTION III • Viral Infections

6A and 6B), 7, and 8. Structurally, these viruses are virtu- adaptive immunity, as exemplified by an immediate-early
ally indistinguishable. The viral DNA genome is packaged protein, ICP47, that mediates the downregulation of major
inside an icosahedral capsid that is surrounded by a layer of histocompatibility complex class I molecules, which are
proteins called the tegument. A lipid envelope that contains required for recognition of HSV-infected cells by HSV-spe-
viral glycoproteins surrounds the capsid and tegument. cific CD8+ T cells.8,9 Replication of viral DNA occurs in the
These glycoproteins mediate virion attachment and entry nucleus of the cell. Assembly of the virus begins with for-
into cells. mation of nucleocapsids in the nucleus, followed by egress
The HSV-1 and HSV-2 genomes consist of approximately across the nuclear membrane and envelopment at cyto-
150,000 base pairs and encode more than 100 proteins.4 plasmic locations. Virus particles are transported to the
The viral genomes consist of two components, L (long) and plasma membrane, where progeny virions are released.
S (short), each of which contains unique sequences that HSV glycoproteins have been designated as glycoproteins
can invert, generating four isomers. Viral genomic DNA B, C, D, E, G, H, I, L, and M.4,5 Glycoproteins B, D, and H
extracted from virions or infected cells consists of four equal (gB, gD, and gH) are required for infectivity and are tar-
populations that differ in the relative orientation of these gets of neutralizing antibodies against HSV, glycoprotein
two unique components. Although the two viruses diverged C (gC) binds to the C3b component of complement, and
millions of years ago, the order of genes in the HSV-1 and the glycoprotein E (gE) and glycoprotein I (gI) complex
HSV-2 genomes follows the same linear pattern, and most binds to the Fc portion of immunoglobulin G (IgG). The
genes have counterparts in both viruses. The nucleotide amino acid sequences of glycoprotein G (gG) produced by
sequence of related genes and the amino acid residues of the HSV-1 and HSV-2 are sufficiently different to elicit anti-
proteins they encode often differ significantly, however.5,6 body responses that are specific for each virus type. The
The two viral types can be distinguished by using restric- fact that the antibody response to the two G molecules
tion enzyme analysis of genomic DNA or, more recently, exhibits minimal cross-reactivity has provided the basis for
by sequencing of selected genes or regions of the genome, type-specific serologic methods that can be used to detect
which allows precise epidemiologic investigation of virus recent or past HSV-1 and HSV-2 infections.10-13 The close
transmission. antigenic relatedness between HSV-1 and HSV-2 interferes
with the serologic diagnosis of these infections when using
standard, type-common serologic assays, which do not
REPLICATION
distinguish between individuals who have had past infec-
Herpes simplex virus replication is characterized by the tion with HSV-1 only, infection with HSV-2 only, or dual
transcription of three gene classes—α, β, and γ—that infection. Commercial type-specific tests based on gG must
encode viral proteins made at immediate-early, early, and be used for diagnosis of HSV-2 infection (Table 27-1).12,14
late times after virus entry into the cell.4 Many of these Clinicians should be knowledgeable regarding the type of
genes can be removed from the viral genome without block- testing performed by the laboratory to interpret results
ing the capacity of the virus to replicate in cultured cells, correctly.
but most have important functions during infection of the
host. The HSV genome can also be manipulated to insert LATENCY AND REACTIVATION
foreign genes without inhibiting viral replication. Muta-
tions that modify the virulence of HSV-1 and HSV-2 may A common characteristic of all members of the human her-
provide an opportunity to design genetically engineered pesvirus family is the ability to establish latency, to persist
herpesviruses for use as vaccines for genital herpes or as in this latent state for various intervals of time, and to reac-
therapeutic agents in the treatment of brain tumors.7 tivate and cause active infection (with or without disease)
HSV α genes are expressed at immediate-early times and viral transmission at mucosal or other sites. After pri-
after infection and are responsible for the initiation of rep- mary infection, the HSV genome persists in sensory gan-
lication. These genes are transcribed in infected cells in the glion neurons for the lifetime of the individual. The biologic
absence of viral protein synthesis. Products of the β genes, phenomenon of latency was first described at the beginning
or early genes, include the enzymes necessary for viral rep- of the 20th century, when Cushing15 observed in 1905
lication, such as HSV thymidine kinase, which is targeted that patients treated for trigeminal neuralgia by sectioning
by acyclovir and related antiviral drugs, and other regula- a branch of the trigeminal nerve developed herpetic lesions
tory proteins. β genes require functional α gene products in areas innervated by the sectioned branch. This specific
for expression. The onset of expression of β genes coincides association of HSV with the trigeminal ganglion was sug-
with a decline in the rate of expression of α genes and an gested by Goodpasture.16 Past observations have shown
irreversible shutoff of host cellular macromolecular pro- that microvascular surgery of the trigeminal nerve tract
tein synthesis. Structural proteins, such as proteins that to alleviate pain associated with tic douloureux resulted
form the viral capsid, are usually of the γ, or late, gene. in recurrent lesions in greater than 90% of seropositive
The γ genes are heterogeneous and are differentiated from individuals.17,18
β genes by their requirement for viral DNA synthesis for Accumulated experience in animal models and from
maximal expression. Most glycoproteins are expressed clinical observations suggests that inoculation of virus at
predominantly as late genes. In addition to its regulatory the portal of entry, usually oral or genital mucosal tissue,
and structural genes, the virus encodes genes that allow results in infection of sensory nerve endings at that site,
initial evasion of the innate host cell response, including with subsequent transport of the virus to the dorsal root
gene products that block the interferon (IFN) pathway. ganglia.19 Replication at the site of inoculation enhances
HSV-1 and HSV-2 also express proteins that interfere with access of the virus to ganglia but usually does not produce
Table 27-1 Quick Reference Guide for Blood Tests to Accurately Detect Type-Specific HSV Antibodies
Biokit HSV-2 Rapid Test Euroimmun
(Also Sold as SureVue Anti-HSV-1 and HerpeSelect HSV-1 HerpeSelect 1 and
HSV-2 Rapid Test by Anti–­HSV-2 ELISA and HerpeSe- 2 Differentiation
Fisher HealthCare) BioPlex HSV Captia ELISA ELISA lect HSV-2 ELISA Immunoblot Liaison HSV-2 AtheNA MultiLyte
Supplier Biokit USA Bio-Rad Trinity Biotech USA Euroimmun US Focus Diagnostics Focus Diagnostics DiaSorin Inverness Medical
Laboratories LLC
FDA approved 1999 2009 2004 2007 2000/2002 2000 2008 2008
Antibodies HSV-2 only HSV-1 or HSV-2 HSV-1 or HSV-2 or HSV-1 or HSV-2 HSV-1 or HSV-2 or both HSV-1 and/or HSV-1 or HSV-1 and/or HSV-2
detected or both HSV-2 HSV-2
both
Best use of POC test to screen or test Screening or Screening or testing Moderate vol- Screening or testing Low volume High volume Screening or testing
test individuals > 3 months testing (high pregnant women ume STD patients or (moderate-to-
post-exposure volume) or STD clinic pregnant women high volume)
patients (moderate (moderate volume)
volume)
Collection Finger stick, whole Blood draw (sent Blood draw (sent to Blood draw (sent Blood draw (sent to Blood draw (sent to Blood draw Blood draw (sent to
method blood or serum in to laboratory) laboratory) to laboratory) ­laboratory) laboratory) (sent to laboratory)
clinic laboratory)
Test time 10 minutes 45 minutes ≈2 hours ≈2 hours ≈2 hours ≈2 hours 35 minutes ≈2 hours
FDA approved Yes Yes Yes Yes
for use
during
pregnancy
Test avail- Limited Limited Widely available Widely available
ability

27 • Herpes Simplex Virus Infections


Website www.bioki www.bio- www.trinity www.euroi www.herpe www.herpe www.diaso www.invernessmed
tusa.com rad.com biotech.com mmunus.com select.com select.com rin.com icalpd.com
For more 800-926-3353 800-224-6723 800-325-3424 800-913-2022 800-328-5669 877-546-8633
­information

From Kimberlin DW, Baley J: Guidance on management of asymptomatic neonates born to women with active genital herpes lesions, Pediatrics 131:e635-e646, 2013.
ELISA, Enzyme-linked immunosorbent assay; HSV, herpes simplex virus; POC, point of care; STD, sexually transmitted disease.

845
846 SECTION III • Viral Infections

signs of mucocutaneous disease; only a fraction of new in acquisition of infection until later in life. Data from
infections with HSV-1 and HSV-2 cause clinically recog- the National Health and Nutrition Examination Surveys
nizable disease. When viral reactivation occurs at oral or (NHANES) recently have revealed that in 2005 to 2010
genital sites, virus then is transported back down axons to the seroprevalence of HSV-1 was 53.9%.28 From 1999 to
mucocutaneous sites, where viral replication and shedding 2004 and 2005 to 2010, HSV-1 seroprevalence declined
of infectious HSV then occurs. by nearly 7% (P < .01), with the largest decline being
Recognizing that excretion of infectious virus during observed among 14- to 19-year-olds, among whom sero-
reactivation is not usually associated with clinical signs prevalence declined by nearly 23%, from 39.0% to 30.1%
of recurrent herpes lesions is essential for understanding (P < .01). In this age group, HSV-1 seroprevalence declined
the transmission of HSV to newborns. Clinically, silent more than 29% from 1976 to 1980 and 2005 to 2010
viral reactivations are much more common than recur- (P < .01).28 As a result, an increasing number of adoles-
rent lesions that are clinically apparent. This distinction cents lack HSV-1 antibodies at sexual debut. This is occur-
between asymptomatic infection and symptomatic disease ring at a time when almost half of teenagers 15 to 19 years
underlies much of the challenge in neonatal HSV preven- of age and more than four fifths of young adults 20 to 24
tion because only those relatively fewer situations where years of age have engaged in oral sex.29 Increasing rates of
there are clinically apparent lesions can be expected to initi- genital HSV-1 infection already are being seen in several
ate a response from treating physicians. Reactivation with studies published over the past decade, with 60% to 80%
or without symptoms occurs in the healthy host in the pres- of cases of genital herpes now being attributed to this virus
ence of HSV-specific humoral and cell-mediated immunity. type.30-32
Reactivation seems to be spontaneous, although symptom- Because infection with HSV-2 is usually acquired
atic recurrences have been associated with physical or emo- through sexual contact, antibodies to this virus are rarely
tional stress, exposure to ultraviolet light, or tissue damage; found until the age of onset of sexual activity.26 A progres-
immunosuppression is associated with an increased pre- sive increase in infection rates with HSV-2 in all popula-
dilection for symptomatic HSV disease when reactivation tions begins in adolescence. In earlier studies, the precise
occurs. Persistence of viral DNA has been documented seroprevalence of antibodies to HSV-2 had been difficult
in neuronal tissue of animal models and humans.4,20-22 to determine because of cross-reactivity with HSV-1 anti-
Because the latent virus does not multiply, it is not suscep- gens. During the late 1980s, seroepidemiologic studies
tible during latency to drugs such as acyclovir, which affect performed using type-specific antigen for HSV-2 (glycopro-
DNA synthesis, and cannot be eradicated from the infected tein G-2) identified this virus in approximately 25% to 35%
host. Understanding of the mechanisms by which HSV of middle-class women in several geographic areas of the
establishes a latent state and persists in this form remains United States.11,33-35 Based on national health surveys, the
limited. seroprevalence of HSV-2 in the United States from 1988
to 1994 was 21.9% for individuals 12 years and older,
representing a 30% increase compared with data collected
Epidemiology and Transmission from 1976 to 1980.35 Among individuals with serologic
evidence of infection, less than 10% had a history of geni-
Transmission of HSV most often occurs as a consequence tal herpes symptoms. A statistically significant decline in
of intimate person-to-person contact. Virus must come in HSV-2 seroprevalence was observed from 1988 to 1994
contact with mucosal surfaces or abraded skin for infection and 1999 to 2004, but since that time findings suggest
to be initiated. seroprevalence has plateaued.36 HSV-2 seroprevalence
The usual mode of HSV-1 transmission is through direct was 15.7% in 2005 to 2010, which was not significantly
contact of a susceptible individual with infected secretions, different from the estimate in 1999 to 2004.28 Overall,
although transfer in respiratory droplets is possible. Acqui- HSV-2 seroprevalence has increased significantly since
sition often occurs during childhood. Primary HSV-1 infec- 1976 to 1980, primarily because of the large increase
tion in a young child usually is asymptomatic, but clinical from 1976-1980 to 1988-1994.35 HSV-2 seroprevalence
illness is associated with HSV gingivostomatitis. Primary is highly variable and depends on geographic region, sex,
infection in young adults has been associated with phar- age, race, and high-risk behaviors.37 The molecular epi-
yngitis only or with a mononucleosis-like syndrome. Sero- demiology of HSV infections can be determined by restric-
prevalence studies have shown that acquisition of HSV-1 tion enzyme analysis of viral DNA or polymerase chain
infection, similar to that of other herpesvirus infections, is reaction (PCR) assay and sequencing of regions of the
related to socioeconomic factors.23,24 Antibodies, indicative HSV genome obtained from infected individuals. Viruses
of past infection, are found early in life more often among have essentially identical genetic profiles when they are
individuals of lower socioeconomic groups, presumably from the same host or are epidemiologically related.38 In
reflecting the crowded living conditions that provide a a few circumstances, it has been shown, however, that
greater opportunity for direct contact with infected indi- superinfection or exogenous reinfection with a new strain
viduals. By the end of the first decade of life, 75% to 90% of of HSV is possible. Such occurrences are uncommon in a
individuals from lower socioeconomic populations develop nonimmunocompromised host with recurrent genital HSV
antibodies to HSV-1.25-27 In middle and upper-middle socio- infection.38-40 Differences in the genetic sequence of viral
economic groups, 30% to 40% of individuals are seroposi- DNAs indicating exogenous infections are more common
tive by the middle of the second decade of life. in immunocompromised individuals who are exposed to
A change in seroprevalence rates of HSV-1 has been different HSVs, such as patients with acquired immunode-
recognized in the past few decades, which reflects a delay ficiency syndrome.
27 • Herpes Simplex Virus Infections 847

MATERNAL INFECTION gestation is not generally associated with premature rupture


of membranes or premature termination of pregnancy.58
Infection with HSV-2, which reactivates and is shed at Localized genital infection, whether it is associated with
genital sites, is common in pregnant women. Using assays lesions or remains asymptomatic, is the most common form
to detect type-specific antibodies to HSV-2, seroepidemio- of HSV infection during pregnancy. Overall, prospective inves-
logic investigations have shown that approximately one tigations using cytologic and virologic screening indicate that
in five pregnant women has had HSV-2 infection.24,34,41-45 genital herpes occurs with a frequency of about 1% in women
Given the capacity of HSV to establish latency, the pres- tested at any time during gestation.43,57 Most maternal genital
ence of antibodies is a marker of persistent infection of the infections are due to recurrent HSV-2 infections when charac-
host with the virus. The incidence of infection in women terized virologically and serologically.49 This potentially could
of upper socioeconomic class was 30% or greater in three change as the incidence of HSV-1 genital infection, which is
large studies.43,45,46 These investigations have shown that more likely to be primary than recurrent, increases in the pop-
most women with serologic evidence of HSV-2 infection ulation.29 Because HSV infection of the infant is usually the
have no history of symptomatic primary or recurrent dis- consequence of contact with infected maternal genital secre-
ease. New HSV-2 infections are acquired during pregnancy tions at the time of delivery, the incidence of viral excretion
with a frequency that is comparable to seroconversion rates at this time point has been of particular interest. The reported
among nonpregnant women, and these infections also usu- incidence of viral excretion at delivery is 0.01% to 0.39% for all
ally occur without clinical signs or symptoms.42-46 Even so, women, regardless of their history of genital herpes.43,46
most genital HSV shedding occurs from reactivation of viral Several prospective studies have evaluated the frequency
infection, rather than from primary HSV disease.47-49 and nature of viral shedding in pregnant women with a
HSV-1 is less likely than HSV-2 to reactivate in the genital known history of genital herpes. These women represent
tract of nonpregnant women, especially after the first year a subset of the population of women with HSV-2 infection
after infection.50 HSV-1 reactivations may occur after mid- because they had a characteristic genital lesion from which
pregnancy, however, when women are relatively immuno- virus was isolated. In a predominantly white, middle-class
suppressed.51 In contrast, genital HSV-2 continues to recur, population, symptomatic recurrent infection occurred dur-
often frequently, for many years.52 ing pregnancy in 84% of pregnant women with a history
Evaluation of pregnant women and their partners has of symptomatic disease.59 Viral shedding from the cervix
shown that women can remain susceptible to HSV-2 despite occurred in only 0.56% of symptomatic infections and
prolonged sexual contact with a partner who has known 0.66% of asymptomatic infections. These data are similar
genital herpes.44 In this study, 1 in 10 women were found to to data obtained from other populations.33 The incidence
be at unsuspected risk for acquiring HSV-2 infection during of cervical shedding in asymptomatic pregnant women has
pregnancy as a result of contact with a partner whose HSV-2 been reported to range from 0.2% to 7.4%, depending on the
infection was asymptomatic. Most maternal infections are numbers of cultures that were obtained between symptom-
clinically silent during gestation. However, infection dur- atic episodes. Overall, these data indicate that the frequency
ing gestation may manifest in several clinical syndromes, of cervical shedding is low, which may reduce the risk of
the most severe of which is widely disseminated disease. transmission of virus to the infant when the maternal infec-
As first reported by Flewett and coworkers53 in 1969 and tion is recurrent. The frequency of maternal genital shed-
by others54,55 subsequently, disseminated infection during ding does not seem to vary by trimester during gestation.
pregnancy has been documented to involve multiple visceral Most infants who develop neonatal disease are born to
sites in addition to cutaneous ones. In a few cases, dissemi- women who are completely asymptomatic for genital HSV
nation after primary oropharyngeal or genital infection has infection during the pregnancy and at the time of delivery.
led to severe manifestations of disease, including necrotizing These women usually have neither a past history of geni-
hepatitis with or without thrombocytopenia, leukopenia, tal herpes nor a sexual partner reporting a genital vesicu-
disseminated intravascular coagulopathy, and encephalitis. lar rash and account for 60% to 80% of all women whose
Although rare, the mortality rate for these pregnant women infants become infected.60, 61
is greater than 50%. Fetal deaths occur in greater than 50%
of cases, although neonatal mortality does not correlate with FACTORS INFLUENCING TRANSMISSION OF
the death of the mother. Surviving fetuses have been deliv-
INFECTION TO THE FETUS
ered by cesarean section during the acute illness or at term,
and may not have evidence of neonatal HSV infection. The development of serologic assays that distinguish
Earlier studies described an association of maternal pri- HSV-1–specific antibodies from HSV-2–specific antibodies
mary infection before 20 weeks of gestation,56 with spon- allowed an accurate analysis of risks related to perinatal
taneous abortion in some women. Although the original transmission of HSV.10-13 The category of maternal genital
incidence of spontaneous abortion after a symptomatic pri- infection at the time of delivery influences the frequency
mary infection during gestation was thought to be 25%, this of neonatal acquisition of infection. Maternal infections
estimate was not substantiated by prospective studies and are classified as caused by HSV-1 or HSV-2 and as newly
was erroneous because of the small number of women fol- acquired or recurrent.62 These categories of maternal infec-
lowed. More precise data obtained from a prospective analy- tion status are based on virologic and serologic laboratory
sis of susceptible women showed that 2% or greater acquired criteria and are independent of clinical signs.
infection, but acquisition of infection was not associated Women with recurrent infections are those who have pre-
with a risk of spontaneous abortion.57 With the exception existing antibodies to the virus type that is isolated from the
of rare case reports, primary infection that develops later in genital tract, which, until recently, has usually been HSV-2.
848 SECTION III • Viral Infections

Most women classified as having recurrent infection have cases, significant complications, such as urinary retention
no history of symptomatic genital herpes. Infections that and aseptic meningitis, occur in the mother.
are newly acquired, which have been referred to as first-­ In contrast, virus is shed for an average of only 2 to 5 days
episode infections, are categorized further as either primary or and at lower concentrations (approximately 102-103 viral
nonprimary based on type-specific serologic testing. This dif- particles/0.2 mL of inoculum) in women with symptom-
ferentiation is made whether clinical signs are present or not. atic recurrent genital infections. Asymptomatic reactiva-
First-episode primary infections are infections in which the tion is also associated with short periods of viral replication,
mother is experiencing a new infection with HSV-1 or HSV-2 often less than 24 to 48 hours. One of the most important
and has not already been infected with the other virus type. observations about HSV infections that has emerged from
These mothers are seronegative for any HSV antibodies (i.e., the evaluation of pregnant women is that new HSV-1 and
negative for both HSV-1 and HSV-2 antibodies) at the onset HSV-2 infections often occur without any of the manifesta-
of infection. First-episode nonprimary infections are infections tions that were originally described as the classic findings in
in which the mother has a new infection with one virus type, primary and recurrent genital herpes.
usually HSV-2, but has antibodies to the other virus type, In parallel with the classification of maternal infection,
usually HSV-1, because of an infection that was acquired the mother’s antibody status to HSV at delivery is an addi-
previously. tional factor that influences the likelihood of transmission
Because transmission has been studied using type-specific and probably affects the clinical course of neonatal herpes.
serologic methods, it has become apparent that attempts to Transplacental maternal neutralizing antibodies have a
distinguish primary and recurrent disease by clinical crite- protective, or at least an ameliorative, effect on acquisition
ria are unreliable. Use of both serologic and virologic data to of infection for infants inadvertently exposed to virus.65
classify the category of maternal infection is an important Maternal first-episode primary or nonprimary infection
advance because many “new” genital herpes infections in late in gestation may not result in significant passage of
pregnancy are actually recurrent infections and represent maternal antibodies across the placenta to the fetus. Based
the first symptomatic episode of a genital HSV infection on available evidence, the highest risk of transmission from
acquired at some time in the past. In one study designed mothers with newly acquired genital herpes is observed
to evaluate acyclovir therapy, pregnant women who were when the infant is born before the transfer of passive anti-
thought to have recent acquisition of HSV-2 based on symp- bodies to HSV-1 or HSV-2, when the infant is exposed at
toms all had been infected previously. These women were delivery, or within the first few days of life.57,66
experiencing genital symptoms, caused by reactivation of The duration of ruptured membranes has also been
latent virus, for the first time.55 described as an indicator of risk for acquisition of neonatal
A hierarchy of risk of transmission has emerged using infection. Observations in the early 1970s of a small cohort of
virologic and serologic laboratory tools to classify maternal women (n = 22) with symptomatic genital herpes indicated
infection. Infants born to mothers who have a first-episode that prolonged rupture of membranes (>6 hours) increased
primary infection at the time of delivery are at highest risk, the risk of acquisition of virus, perhaps as a consequence of
with transmission rates approaching 60%.49,57,60 Infants ascending infection from the cervix.56 This small case series
born to mothers with first-episode nonprimary infections resulted in the recommendation that women with active
are at lower risk, with transmission rates of approximately genital lesions at the time of onset of labor be delivered by
25%.49 The lowest risk of neonatal acquisition occurs with cesarean section.67 It was not until 2003, however, that it
recurrent infection, when the mother has active infection was proven in a large, landmark study that cesarean deliv-
caused by shedding of virus that she acquired before the ery protects against neonatal HSV infection in infants born to
pregnancy or earlier in gestation; the estimated attack rate women from whom HSV was isolated at the time of delivery
for neonatal herpes among these infants is approximately (1.2% vs. 7.7%; P = .047).49 In the single baby in this trial to
2%.49 This estimate is reliable because it is based on the develop neonatal HSV disease after cesarean delivery, mem-
cumulative experience from large, prospective studies of branes were ruptured in the mother for 19 hours. It is impor-
pregnant women in which viral shedding was evaluated at tant to note, however, that infection of the newborn with
delivery, regardless of the mother’s history of genital herpes HSV-2 has occurred despite delivery by cesarean section.61,68
or contact with a partner with suspected or documented Certain forms of medical intervention during labor and
genital herpes. delivery may increase the risk of neonatal herpes if the mother
The higher risk of transmission to the infant when the has active shedding of the virus, although in most instances,
mother has a new infection can be attributed to differences viral shedding is not suspected clinically. Fetal scalp moni-
in the quantity and duration of viral shedding in the mother tors can be a site of viral entry through disrupted skin.49,69,70
and in the transfer of passive antibodies from the mother The benefits and risks of these devices should be considered
to the infant before delivery. Primary infection is associated for women with a history of recurrent genital HSV infections.
with larger quantities of virus replication in the genital tract Because most women with genital infections caused by HSV
(>106 viral particles/0.2 mL of inoculum) and a period of are asymptomatic during labor and have no history of geni-
viral excretion that may persist for an average of 3 weeks.63 tal herpes, it is usually impossible to make this assessment.
Many women with new infections have no symptoms but
shed virus in high titers. In some mothers, these infections INCIDENCE OF NEWBORN INFECTION
cause signs of systemic illness, including fever, malaise,
myalgias, dysuria, and headache. Viremia during primary Estimates of the incidence of neonatal herpes have var-
HSV infection in women is common and is associated with ied from 1 in 3000 to 1 in 20,000 live births.71 Although
systemic maternal symptoms.64 In a small percentage of fluctuations in the incidence of neonatal HSV disease have
27 • Herpes Simplex Virus Infections 849

been observed,46,71 the current estimated rate of occur- sequelae. When using stringent diagnostic criteria, more
rence is approximately 1 in 3200 deliveries.49 Although a than 70 infants with symptomatic congenital disease have
progressive increase in the number of cases of neonatal HSV been described in the literature.75 These criteria include
infection has been noted in some areas of the country,72 identification of infected infants with lesions present at birth
neonatal HSV infections still occur far less frequently than or within the first 24 hours of life; virologic confirmation of
do genital HSV infections in the adult childbearing popu- HSV at that time; and exclusion of other infectious agents
lation. Overall, the United States, with approximately 4.0 whose pathogenesis mimics the clinical findings of HSV
million deliveries per year, has an estimated 1500 cases of infections, such as congenital cytomegalovirus infection,
neonatal HSV infection annually. rubella, syphilis, or toxoplasmosis. Virologic diagnosis is a
In studies where maternal serologic status during preg- necessary criterion because no standard method for reli-
nancy and virologic status at the time of delivery are able detection of IgM antibodies is available, and infected
evaluated prospectively, the rate of transmission leading infants often fail to produce IgM antibodies detectable by
to neonatal HSV infection ranges from 12 to 54 newborn research methods.66,79 Intrauterine HSV disease occurs in
infections per 100,000 births (1/8300 and 1/1850 births, approximately 1 in 300,000 deliveries.75 Although rare,
respectively). Higher rates of transmission are seen in in utero disease is unlikely to be missed due to the extent
infants born to seronegative mothers and mothers infected of involvement of affected babies. Infants acquiring HSV
with HSV-1.49 Based on seroprevalence studies, the high- in utero typically have a triad of clinical findings consist-
est risk of HSV transmission would be expected to occur ing of cutaneous manifestations (scarring, active lesions,
in infants born to non-Hispanic white mothers, whose hypopigmentation and hyperpigmentation, aplasia cutis,
HSV seroprevalence is the lowest.24 Some countries do and/or an erythematous macular exanthem), ophthal-
not report a significant number of cases of neonatal HSV mologic findings (microopthalmia, retinal dysplasia, optic
infection despite a similar high prevalence of antibodies to atrophy, and/or chorioretinitis), and neurologic involve-
HSV-2 in women. In the United Kingdom, genital herpes ment (microcephaly, encephalomalacia, hydranencephaly,
infection is relatively common, but very few cases of neona- and/or intracranial calcification).76,77,80,81
tal HSV infection are recognized. Neonatal HSV infection in In utero infection can result from transplacental or
the Netherlands occurs in only 2.4 of 100,000 newborns.73 ascending infection. The placenta can show evidence of
Although underreporting of cases may explain some differ- necrosis and inclusions in the trophoblasts, which suggests
ences between countries, unidentified factors may account a transplacental route of infection.82 The situation can
for these differences. The interpretation of incidence data result in an infant who has hydranencephaly at the time
must also include the potential for postnatal acquisition of of birth, or it may be associated with spontaneous abortion
HSV infection. Not all cases of neonatal infection are the and intrauterine HSV viremia. Virus has been isolated from
consequence of intrapartum contact with infected mater- the products of conception under such circumstances. His-
nal genital secretions, which alters the overall estimate of topathologic evidence of chorioamnionitis suggests ascend-
delivery-associated infection. The prevalence of neonatal ing infection as an alternative route for in utero infection.83
HSV infection relative to serious bacterial infections in hos- Risk factors associated with intrauterine transmission are
pitalized neonates was evaluated more recently in a retro- unknown. Primary and recurrent maternal infections can
spective study and found to be 0.2% compared with 0.4% result in infection of the fetus in utero. HSV DNA has been
and 4.5% for infants with bacterial meningitis and serious detected in the amniotic fluid of two women experiencing a
bacterial infections, respectively.74 first-episode nonprimary infection and in one woman dur-
ing a symptomatic recurrent infection. All three infants
were healthy at birth and showed no clinical or serologic
TIMES OF TRANSMISSION OF INFECTION
evidence of HSV infection during follow-up.84
Herpes simplex virus infection of the newborn can be The second and most common route of infection is intra-
acquired in utero, intrapartum, or postnatally. The mother partum contact of the fetus with infected maternal genital
is the source of infection for the first two of these three secretions. Intrapartum transmission can occur when a
routes of transmission of infection. With regard to post- baby passes through a birth canal when HSV is present.
natal acquisition of HSV infection, the mother can be a Approximately 85% of cases of neonatal HSV are acquired
source of infection from a nongenital site, or other contacts in the intrapartum period. Intrapartum transmission is
or environmental sources of virus can lead to infection of more likely to occur when the neonate is being delivered to a
the infant. A maternal source is suspected when maternal mother with newly acquired infection (25%-60% likelihood
herpetic lesions are discovered during or shortly after the of transmission if virus is present in the genital tract) but
birth of the infant, or when the infant’s illness is caused by can also occur with recurrent maternal infection (approxi-
HSV-2. Although intrapartum transmission accounts for mately a 2% likelihood of transmission if virus is present in
approximately 85% of cases of neonatal HSV, in utero and the genital tract).
postnatal infection must be recognized for public health and Postnatal acquisition is the third route of transmission,
prognostic purposes. accounting for approximately 10% of cases of neonatal HSV
In utero transmission is very rare, causing approximately disease. Postnatal acquisition of neonatal HSV is virtually
5% of all cases of neonatal HSV disease.75-78 Although it was always due to HSV-1 because the source of transmission is
originally presumed that in utero acquisition of infection nongenital. However, with the increasing incidence of geni-
resulted in a totally normal infant or premature termina- tal HSV-1 disease in sexually active adults, simply detecting
tion of gestation,56 it has become apparent that intrauterine HSV-1 in a neonate does not define the timing of transmis-
acquisition of infection can lead to severe clinical disease and sion as postnatal. The most recent data on the proportion of
850 SECTION III • Viral Infections

neonatal HSV that is caused by HSV-1 are from the 1980s,


when the National Institute of Allergy and Infectious Dis- Immunologic Response
eases (NIAID) Collaborative Antiviral Study Group (CASG)
reported that approximately 25% to 35% of cases of neona- The neonatal host response to HSV is impaired compared
tal herpes are caused by this virus type.61,85 with older children and adults.66,79,94-98 There is no evi-
The documentation of postnatal transmission of HSV-1 dence for differences in virulence of particular HSV strains.
has focused attention on nongenital sources of virus.86-90 The severity of the manifestations of HSV-1 and HSV-2
Postpartum transmission from mother to child has been infections in the newborn can be attributed to immuno-
reported as a consequence of nursing on an infected logic factors. Relevant issues are protection by transplacen-
breast.91 Transmission from fathers and grandparents has tal antibodies, the innate immune response of the exposed
also been documented.90 When the infant’s mother has not infant, and the acquisition of adaptive immunity by the
had HSV infection, the infant may be inoculated with the infected newborn.
virus from a nonmaternal contact in the absence of any Passive antibodies to HSV influence the acquisition of
possible protection from maternally derived passive anti- infection and its severity and clinical signs.45,60,66,79 Trans-
bodies. Because of the high prevalence of HSV-1 infection placentally acquired antibodies from the mother are not
in the general population, many individuals have intermit- totally protective against neonatal infection, but transpla-
tent episodes of asymptomatic excretion of the virus from centally acquired neutralizing antibodies correlate with a
the oropharynx and can provide a source of infection for lower attack rate in exposed newborns.60,65,66 Although
the newborn. The occurrence of herpes labialis, commonly the absence of any detectable antibodies has been associated
referred to as fever blisters or cold sores, has ranged from with dissemination and systemic disease in the neonate, the
16% to 46% in various groups of adults.92 presence of antibodies at the time that clinical signs appear
Population studies conducted in two hospitals indicated does not predict the subsequent outcome.61,79
that 15% to 34% of hospital personnel had a history of non- Most infected newborns eventually produce HSV IgM
genital herpetic lesions.92,93 In both hospitals surveyed, at antibodies, but the interval to detection is prolonged, requir-
least 1 in 100 individuals documented a recurrent cold sore ing at least 2 to 4 weeks.66 These antibodies increase rapidly
each week. As is true of genital herpes, many individuals during the first 2 to 3 months but may be detectable for 1
have HSV-1 infection with no clinical symptoms at the time year after infection. The quantity of neutralizing antibodies
of acquisition or during episodes of reactivation and shed- and antibodies that mediate antibody-dependent cellular
ding of infectious virus in oropharyngeal secretions. Pro- cytotoxicity in infants with disseminated disease is lower
spective virologic monitoring of hospital staff increased the than in infants with more limited disease.66,97 Humoral
frequency with which infection was detected by twofold; antibody responses to specific viral proteins, especially gly-
however, no cases of neonatal HSV infection were docu- coproteins, have been evaluated by assays for antibodies to
mented in these nurseries. gG and by immunoblot.10,79 Immunoblot studies indicate
The risk of nosocomial infection in the hospital envi- that the severity of infection correlates directly with the
ronment is a concern. Identification by restriction endo- number of antibody bands to defined polypeptides. Children
nuclease or sequence analysis of virus recovered from an with a more limited infection, such as infection of the skin,
index case and a nursery contact leaves little doubt about eye, or mouth (SEM disease), have fewer antibody bands
the possibility of spread of virus in a high-risk nursery compared with children with disseminated disease.
population.87,89 The possible vectors for nosocomial trans- A vigorous antibody response to the ICP4 α gene prod-
mission have not been defined. The risk of transmission uct, which is responsible for initiating viral replication, has
to infants by health care professionals who have herpes been correlated with poor long-term neurologic outcome,
labialis or who are asymptomatic oral shedders of virus suggesting that these antibodies reflect the extent of viral
is low. Compromising patient care by excluding health replication. A regression analysis that compared neurologic
care professionals with cold sores who are essential for the impairment with the quantity of antibodies to ICP4 identi-
operation of the hospital nursery must be weighed against fied the child at risk for severe neurologic impairment.79
the potential risk of newborn infants becoming infected. Adaptive cellular immunity is a crucial component of
Health care professionals with cold sores who have contact the host response to primary herpetic infection. Newborns
with infants should cover and not touch their lesions and with HSV infections have a delayed T-lymphocyte prolif-
should comply with hand hygiene policies. Transmission erative response compared with older individuals.66,94,95
of HSV infection from health care professionals with geni- Most infants have no detectable T-lymphocyte responses to
tal lesions is not likely as long as they comply with hand HSV when evaluated 2 to 4 weeks after the onset of clinical
hygiene policies. Health care professionals with an active symptoms.66 The delayed T-lymphocyte response to viral
herpetic whitlow should not have responsibility for direct antigens in infants whose initial disease is localized to the
care of neonates or immunocompromised patients and SEM may be an important determinant of the frequent pro-
should wear gloves and use hand hygiene during direct gression to more severe disease in infants.66,94 The impor-
care of other patients. tance of IFN-γ may be related to its effect on the induction
Because most mothers have antibodies to HSV, and these of innate immune mechanisms, such as natural killer–cell
antibodies are transferred to their infants, postnatal expo- responses.96 Other mechanisms of the innate immune
sures to the virus in the neonatal period usually do not system of the newborn that may be deficient in control-
result in neonatal disease. If the mother is seronegative, ling HSV include other nonspecific cytokine responses and
nosocomial exposure may pose a more significant risk to complement-mediated effects. T lymphocytes from infected
the neonate, however. infants have decreased IFN-γ production during the first
27 • Herpes Simplex Virus Infections 851

month of life. This defect can be predicted to limit the clonal by microscopy, there is extensive evidence of hemorrhagic
expansion of helper and cytotoxic T lymphocytes specific necrosis, clumping of nuclear chromatin, dissolution of
for herpes viral antigens, allowing more extensive and pro- the nucleolus, cell fusion with formation of multinucleate
longed viral replication. Antibody-dependent cell-mediated giant cells, and, ultimately, a lymphocytic inflammatory
cytotoxicity has been shown to be an important component response.104 Irreversible organ damage results from isch-
of adaptive immunity to viral infection.97 Lymphocytes, emia and direct viral destruction of cells. A major advance
monocytes, macrophages, or polymorphonuclear leuko- in our understanding of the pathogenesis of sequelae of
cytes and antibodies and complement lyse HSV-infected neonatal HSV CNS disease is the finding that subclinical
cells in vitro.99 However, newborns appear to have fewer reactivation of virus within the CNS occurs after successful
effector lymphocytes than older individuals do. The imma- treatment of the acute disease and that this contributes to
turity of neonatal monocytes and macrophage function the long-term neurologic sequelae of this manifestation of
against HSV infection has been demonstrated in vitro and neonatal HSV disease.105
in animal models.100,101 Additional information regarding
the immune response to HSV is provided in Chapter 4. CLINICAL MANIFESTATIONS
Pediatricians should consider the diagnosis of neonatal her-
pes in infants who have clinical signs consistent with the
Neonatal Infection disease, regardless of the maternal history of genital her-
pes. Only a minority (20%-40%) of mothers whose infants
PATHOGENESIS AND PATHOLOGY
develop neonatal herpes have had symptomatic genital
After direct exposure, replication of HSV is presumed to herpes or sexual contact with a partner who has recognized
occur at the portal of entry, which is probably the mucous HSV infection during or before the pregnancy.
membranes of the mouth or eye, or at sites where the skin The clinical presentation of infants with neonatal HSV
integrity has been compromised. Factors that determine infection depends on the initial site of infection and the
whether the infection causes symptoms at the site of inoc- extent of viral replication. In contrast to human cytomega-
ulation or disseminates to other organs are poorly under- lovirus, neonatal infections caused by HSV-1 and HSV-2 are
stood. Sites of replication during the incubation period have almost invariably symptomatic. Case reports of asymptom-
not been well defined, but the virus evades the host response atic infection in the newborn exist but are uncommon, and
during this early stage, probably by mechanisms such as long-term follow-up of these children to document absence
interfering with expression of the IFN response genes and of subtle disease or sequelae is not sufficient to determine if
blocking cell-mediated immune recognition of viral pep- neonatal infection actually occurred.
tides by preventing major histocompatibility complex class Classification of newborns with HSV infection is used for
I molecules from reaching the surface of infected cells. prognostic and therapeutic considerations.106 Historically,
Intraneuronal transmission of viral particles may pro- infants with neonatal HSV infection were classified as hav-
vide a privileged site that is relatively inaccessible to cir- ing localized or disseminated disease, with the former group
culating humoral and cell-mediated defense mechanisms, being subdivided into infants with SEM disease versus infants
facilitating the pathogenesis of encephalitis. Transplacen- with CNS infection. This classification system understated
tal maternal antibodies may be less effective under such the significant differences in outcome within each category,
circumstances. Disseminated infection seems to be the however.107 In a revised classification scheme, infants who
consequence of viremia. HSV DNA has been detected in are infected intrapartum or postnatally are divided into three
peripheral blood mononuclear cells, even in infants who groups: disease localized to the skin, eyes, or mouth (SEM dis-
have localized infection.102 Extensive cell-to-cell spread ease); encephalitis, with or without SEM involvement (CNS
could help explain primary HSV pneumonia after aspira- disease); and disseminated infection that involves multiple
tion of infected secretions. organs, including the CNS, lung, liver, adrenals, and SEM (dis-
After the virus has adsorbed to cell membranes and pen- seminated disease). This classification system is predictive of
etration has occurred, viral replication proceeds, leading to both morbidity and mortality.85,106,108-110 Patients with dis-
release of progeny virus and cell death. The synthesis of cel- seminated or SEM disease generally present to medical atten-
lular DNA and protein ceases as large quantities of HSV are tion at 10 to 12 days of life, whereas patients with CNS disease
produced. Many infants with disseminated HSV infection on average present somewhat later at 16 to 19 days of life.85
have high viral loads and higher concentrations of inflam- Knowledge of the patterns of clinical disease caused by
matory cytokines compared with infants with central HSV-1 and HSV-2 in the newborn is based on prospectively
nervous system (CNS) infection alone or SEM disease.103 acquired data obtained through the NIAID CASG. These
Uncontrolled host immune responses may contribute to analyses have used uniform case record forms from one
the development of multiorgan dysfunction. Cell death in study interval to the next. Approximately 25% of babies
critical organs of the newborn, such as the brain, results with neonatal HSV infection are classified as having dis-
in devastating consequences, as reflected by the long-term seminated disease, whereas 30% have CNS disease and
morbidity of herpes encephalitis. Cellular swelling, hemor- 45% have SEM disease.61,111
rhagic necrosis, development of intranuclear inclusions,
and cytolysis all result from the replication process and Intrauterine Infection
ensuing inflammatory response. Small, punctate, yellow- Intrauterine infection is very rare, constituting only about
to-gray areas of focal necrosis are the most prominent gross 5% of cases of neonatal HSV infection.75 When infec-
lesions in infected organs. When infected tissue is examined tion occurs in utero, severe disease follows acquisition of
852 SECTION III • Viral Infections

infection at virtually any time during gestation. In the most clinical course. Infants should be assessed for hypoxemia,
severely affected group of infants, evidence of infection is acidosis, hyponatremia, transaminitis, direct hyperbilirubi-
apparent at birth or within the first 24 hours of life and is nemia, neutropenia, thrombocytopenia, and bleeding dia-
characterized by a triad of findings: skin vesicles or scarring, thesis. Chest radiographs also should be obtained. Depending
eye damage, and severe manifestations of microcephaly or on signs and whether the infant is stable enough, abdomi-
hydranencephaly. CNS damage is caused by intrauterine nal radiography, electroencephalography, and computed
encephalitis. Infants do not have evidence of embryopa- tomography (CT) or magnetic resonance imaging (MRI)
thy, such as cardiac malformations. Often, chorioretinitis of the head should be obtained to determine further the
combined with other eye findings, such as keratoconjunc- extent of disease. The radiographic picture of HSV lung dis-
tivitis or microphthalmia, is a component of the clinical ease is characterized by a diffuse, interstitial pattern, which
presentation. progresses to a hemorrhagic pneumonitis and, rarely, a
Serial ultrasound examination of the mothers of infants significant pleural effusion.120 Frequently, necrotizing
infected in utero has demonstrated the presence of hydran- enterocolitis with pneumatosis intestinalis can be detected
encephaly, but cases are seldom diagnosed before delivery. when gastrointestinal disease is present. Meningoenceph-
Chorioretinitis alone should alert the pediatrician to the alitis seems to be a common component of disseminated
possibility of this diagnosis, although it is a sign for other, infection, occurring in about 60% to 75% of children. Usual
more commonly encountered congenital infections as well. examinations of cerebrospinal fluid (CSF), including PCR
A few infants have been described who have signs of HSV for HSV DNA and viral culture, should be performed along
infection at birth after prolonged rupture of membranes. with noninvasive neurodiagnostic tests to assess the extent
These infants may have no other findings of invasive mul- of brain disease.
tiorgan involvement—no chorioretinitis, encephalitis, or The mortality rate for disseminated HSV in the absence of
evidence of other diseased organs—and can be expected to therapy exceeds 80%, and many survivors are impaired. The
respond to antiviral therapy. Antiviral therapy cannot be most common causes of death of infants with disseminated
expected to improve long-term outcomes for infants who disease are intravascular coagulopathy or HSV pneumoni-
are born with very severe CNS involvement or hydranen- tis. Premature infants seem to be at particularly high risk
cephaly. Intrauterine HSV infection has been reported as a for disseminated disease with pneumonitis and have a high
cause of hydrops fetalis.112 mortality rate, even with appropriate antiviral therapy.121
Disseminated Disease Central Nervous System Disease
Infants with disseminated neonatal HSV disease have the Almost one third of all infants with neonatal HSV infec-
highest mortality rate of the three intrapartum and post- tion have only encephalitis as the initial manifestation of
natal disease classifications. Many of these infants are born disease.122,123 These infants have clinical manifestations
to mothers who are experiencing a first-episode primary or distinct from infants who have CNS infection associated
nonprimary HSV-1 or HSV-2 infection and may lack any with disseminated HSV, and the pathogenesis of these two
passively acquired antibodies against the infecting virus forms of brain infection is probably different. The virus is
type.10,60,113 Patients with disseminated disease gener- likely to reach brain parenchyma by a hematogenous route
ally present to medical attention at 10 to 12 days of life.85 in infants with disseminated infection, resulting in mul-
The short incubation period of disseminated herpes reflects tiple areas of cortical hemorrhagic necrosis. In contrast,
an acute viremia, which allows transport of the virus to neonates who present with only CNS disease are likely to
all organs; the principal organs involved are the adrenals develop brain infection because of retrograde axonal trans-
and the liver, resulting in fulminant hepatitis in some port of the virus to the CNS. Patients with CNS disease on
cases.106,114-117 Viremia is associated with infection of circu- average present at 16 to 19 days of life.85 Clinical mani-
lating mononuclear cells in these infants.102,114,116,118,119 festations of encephalitis include seizures (focal or gener-
Disseminated infection can affect multiple organs, alized), fever, lethargy, irritability, tremors, poor feeding,
including the CNS, larynx, trachea, lungs, esophagus, temperature instability, bulging fontanelle, and pyramidal
stomach, lower gastrointestinal tract, spleen, kidneys, pan- tract signs. Similar signs are observed when disseminated
creas, and heart. Initial signs and symptoms are irritability, herpesvirus is associated with encephalitis. Approximately
seizures, respiratory distress, jaundice, coagulopathy, and one third of infants with CNS disease do not have skin vesi-
shock. The characteristic vesicular exanthem oftentimes is cles when signs of illness begin. Some infants have a history
not present when the symptoms begin. Untreated infants or residual signs of lesions of the SEM that were not recog-
may develop cutaneous lesions resulting from viremia, but nized as herpetic.
greater than 40% of children with disseminated infection Anticipated findings on CSF examination include a
do not develop skin vesicles during the course of their ill- mononuclear cell pleocytosis, moderately low glucose
ness.61,85,114 Disseminated infections caused by HSV-1 and concentrations, and elevated protein. A few infants with
HSV-2 are indistinguishable by clinical criteria. CNS infection, proven by brain biopsy done immediately
The diagnosis of disseminated neonatal herpes is exceed- after the onset of seizures, have no abnormalities of CSF,
ingly difficult because the clinical signs are often vague and but most infants have some degree of pleocytosis and mild
nonspecific, mimicking signs of neonatal bacterial sepsis reduction of the glucose level. The hemorrhagic nature of
(e.g., group B streptococcal or Escherichia coli sepsis) or neo- the encephalitis may result in an apparent “bloody tap,”
natal enteroviral sepsis syndrome. although this is seen less frequently in the current era com-
Evaluation of the extent of dissemination is imperative to pared with the 1970s, when awareness of neonatal HSV
provide appropriate supportive interventions early in the infection was lower and earlier assessment less likely to
27 • Herpes Simplex Virus Infections 853

occur. Although initial protein concentrations may be nor- Despite the presumed differences in pathogenesis, neuro-
mal or only slightly elevated, infants with localized brain logic manifestations of disease in children with CNS disease
disease usually show progressive increases in protein. The are virtually identical to the findings for brain infection in
importance of CSF examination in all infants is underscored disseminated disease. For infants with CNS disease, approxi-
by the finding that even subtle abnormalities have been mately two thirds develop evidence of a vesicular rash char-
associated with significant developmental sequelae.106 acteristic of HSV infection. A newborn with pleocytosis and
Electroencephalography and MRI or CT can be very use- elevated protein in the CSF but without the characteristic
ful in defining the presence and extent of CNS abnormalities rash of neonatal HSV can easily be misdiagnosed as having
and should be obtained before discharge of all infants with another viral or bacterial infection unless HSV infection is
this diagnosis.124,125 Abnormalities may also be detected considered.
by ultrasound examination.125 Typical abnormalities seen
by neuroimaging include localized or multifocal areas of Skin, Eye, or Mouth Disease
abnormal parenchymal attenuation, atrophy, edema, and Infection localized to the SEM or some combination of these
hemorrhage involving the temporal, frontal, parietal, and sites seems benign at the onset but is associated with a high
subcortical regions of the brain (Fig. 27-1).126 Predominant risk of progression to serious disease. When infection is
brainstem involvement is rare but reported.127 localized to the skin, the presence of discrete vesicles remains
Localized CNS disease is fatal in approximately 50% of the hallmark of disease (Fig. 27-2). Vesicles occur in greater
infants who are not treated. With rare exceptions, survivors than 80% of children with SEM disease. The skin vesicles
who are not treated are left with neurologic impairment.106 usually erupt from an erythematous base and typically are
With parenteral antiviral therapy, mortality is decreased 1 to 2 mm in diameter. The formation of new lesions adja-
to approximately 5%,108 and 30% of survivors who also cent to the original vesicles is typical, creating a cluster that
receive 6 months of oral acyclovir suppressive therapy have may coalesce into larger, irregular vesicles. In some cases,
some degree of neurologic impairment,105 often in associa- the lesions progress to bullae larger than 1 cm in diameter.
tion with microcephaly, hydranencephaly, porencephalic Clusters of vesicles may appear initially on the presenting
cysts, spasticity, blindness, chorioretinitis, or learning dis- part of the body, presumably because of prolonged contact
abilities. Quantitative PCR methods show a greater amount with infectious secretions during birth, or at sites of trauma
of HSV-2 DNA in CSF from patients with more extensive (e.g., scalp monitor sites). However, it also is common for
neurologic impairment.128 Although many infants have the first herpetic lesions in infants with localized cutaneous
obvious severe sequelae within a few weeks after onset of disease to be on the trunk, extremities, and other sites.
HSV encephalitis,109,129 recent data document that without Patients with SEM disease generally present to medical
follow-up suppressive oral therapy progressive, subclinical attention at 10 to 12 days of life.85 Although discrete vesi-
neurologic damage occurs after parenteral treatment of the cles are usually encountered, crops and clusters of vesicles
acute infection.105 have been described, particularly before antiviral treatment
was available or when the cause of the first lesions was not
recognized. In these cases, the rash can progress to involve
other cutaneous sites, presumably by viremia and hema-
togenous spread. The scattered vesicles can resemble vari-
cella. Although progression is expected without treatment,
a few infants have had infection of the skin limited to one or
two vesicles, with no further evidence of cutaneous disease.
These infants may be identified after the newborn period
and should have a careful evaluation because many are
likely to have had neurologic disease that was not detected.
A zosteriform eruption is another manifestation of herpetic
skin disease reported in infants.130

Figure 27-1 Herpes simplex encephalitis. Computed tomography


scan of an infant with herpes simplex virus type 2 infection and severe Figure 27-2 Cutaneous herpes simplex virus infection. Initial vesicular
sequelae. lesion in a premature infant with herpes simplex type 2 infection.
854 SECTION III • Viral Infections

Table 27-2 Morbidity and Mortality Among Patients After 12 Months by Viral Type, 1981-1997
DISEASE CLASSIFICATION
SEM CNS DISSEMINATED
Outcome HSV-1 HSV-2 HSV-1 HSV-2 HSV-1 HSV-2
Normal 24 (100%) 19 (95%) 4 (57%) 7 (17.5%) 3 (23%) 14 (41%)
Mild impairment 0 (0%) 0 (0%) 0 (0%) 7 (17.5%) 0 (0%) 1 (3%)
Moderate impairment 0 (0%) 1 (5%) 1 (14%) 7 (17.5%) 0 (0%) 0 (0%)
Severe impairment 0 (0%) 0 (0%) 2 (29%) 13 (32.5%) 1 (8%) 3 (9%)
Death 0 (0%) 0 (0%) 0 (0%) 6 (15%) 9 (69%) 16 (47%)
Unknown Total of 20 Total of 16 Total of 12

Data from Douglas J, Schmidt O, Corey L: Acquisition of neonatal HSV-1 infection from a paternal source contact, J Pediatr 103:908-910, 1983.
CNS, Central nervous system; SEM, skin, eyes, or mouth.

Infections involving the eye may manifest as keratocon- not develop symptoms have been followed for the first year
junctivitis. Ocular infection may be the only site of involve- of life and did not have immunologic evidence of subclini-
ment in a newborn. When localized eye infection is observed cal infection.60 HSV-1 or HSV-2 may be recovered from the
in infants who also have microphthalmos and retinal dys- infant’s oropharyngeal secretions transiently, without repre-
plasia, intrauterine acquisition should be suspected, and senting true infection. Because of the propensity of the new-
a thorough neurologic evaluation should be done. Before born to develop severe or life-threatening disease, laboratory
antiviral therapy was available, persistent ocular disease evidence of neonatal HSV infection requires careful follow-
resulted in chorioretinitis caused by HSV-1 or HSV-2.131 up for clinical signs and administration of antiviral therapy.
Keratoconjunctivitis can progress to chorioretinitis, cata-
racts, and retinal detachment despite therapy. Cataracts
have been detected as a long-term consequence in infants
with HSV infections acquired intrapartum.
Diagnosis
Localized infection of the oropharynx involving the CLINICAL EVALUATION
mouth or tongue occurs, but newborns do not develop the
classic herpetic gingivostomatitis caused by primary HSV-1 The clinical diagnosis of neonatal HSV infection is difficult
infection in older children. Overall, approximately 10% of because the appearance of skin vesicles cannot be relied on
patients have evidence of HSV infection of the oropharynx as an initial component of disease presentation. Neonatal
by viral culture. Many of these children did not undergo enteroviral sepsis syndrome is a major differential diag-
a thorough oral examination to determine whether the nostic possibility in infants with signs suggesting neona-
detection of infectious virus in oropharyngeal secretions tal HSV. Bacterial infections of newborns, such as group
was associated with lesions. B streptococcal and E. coli sepsis, also can mimic neonatal
Long-term neurologic impairment has been encountered HSV infection. Skin lesions may resemble lesions seen with
in children whose disease seemed to be localized to the SEM bullous or crusted impetigo. Some infants infected by HSV
during the newborn period.61,106,109 Significant findings have been described who had concomitant bacterial infec-
included spastic quadriplegia, microcephaly, and blindness. tions, including group B streptococci, Staphylococcus aureus,
However, these outcomes were described before routine use Listeria monocytogenes, and gram-negative bacteria. A posi-
of CSF PCR analysis for HSV DNA and likely occurred in tive culture for one of these pathogens does not rule out
babies who were PCR positive and now would be classified HSV infection if the clinical suspicion for neonatal herpes
as having CNS disease.132 infection is present.
Most newborns who have skin lesions experience cutane- Many other disorders of the newborn can be indistin-
ous recurrences for months or years. Table 27-2 shows mor- guishable from neonatal HSV infections, including acute
bidity and mortality 12 months after infection by HSV viral respiratory distress syndrome, intraventricular hemorrhage,
type and disease classification in patients enrolled in two necrotizing enterocolitis, and various ocular or cutaneous
studies conducted by the NIAID CASG from 1981 to 1997. diseases. When vesicles are present, alternative causes of
Although HSV-2 outcomes appear to be more severe than neonatal exanthems should be excluded (Box 27-1). Cutane-
HSV-1 outcomes, the differences between viral types do not ous disorders such as erythema toxicum, neonatal melanosis,
reach statistical significance in regression analysis (P = .10).85 acrodermatitis enteropathica, and incontinentia pigmenti
often confuse physicians who suspect neonatal HSV infec-
Subclinical Infection tions. HSV lesions can be distinguished rapidly from lesions
A few cases of apparent subclinical infection with HSV caused by these diseases using direct immunofluorescence
proven by culture isolation of virus in the absence of symp- stain of lesion scrapings or other methods for rapid detection
toms have been described.133 It has been difficult to docu- of viral proteins and confirmation by viral culture.
ment such cases in the course of prospective evaluations Neonatal HSV CNS disease is a difficult clinical diagnosis
of several hundred infants from many centers around the to make, particularly because many such neonates do not
United States. Conversely, infants who were exposed to have a vesicular rash at the time of clinical presentation.
active maternal infection at the time of delivery and who did Infection of the CNS is suspected in a child who has evidence
27 • Herpes Simplex Virus Infections 855

Of the sites routinely cultured for HSV during a recent


Box 27-1 Differential Diagnosis for Vesicular
study,85 skin or eye/conjunctival cultures consistently pro-
Eruptions in the Neonate vided the greatest yields regardless of disease classification,
Infectious Etiologies with greater than or equal to 90% of cultures being positive.
Overall, 58 (94%) of 62 patients had a positive skin or eye
HSV
culture, 33 (48%) of 69 patients had a positive mouth/oro-
Staphylococcus aureus
Pseudomonas
pharyngeal culture, and 17 (40%) of 42 patients with CNS
Haemophilus influenzae type b involvement (CNS disease or disseminated disease with CNS
Treponema pallidum involvement) had a positive CSF or brain biopsy culture.85
Candida Viral DNA detection by PCR assay is the gold standard
Aspergillus diagnostic method for CNS involvement.132,134-136 PCR was
VZV used in the retrospective analysis of materials collected from
CMV 24 infants enrolled in the NIAID CASG antiviral studies.132
Listeria monocytogenes HSV was detected by PCR assay of CSF in 71% of infants
Group B streptococci before antiviral therapy was initiated. At least one speci-
Noninfectious Conditions men was positive in 76% of infants, and all samples that
Erythema toxicum
were positive by viral culture were positive by PCR. Simi-
Pustular melanosis lar findings were reported by Swedish investigators when
Miliaria stored CSF specimens obtained from infants with neonatal
Letterer-Siwe disease HSV infection were tested for HSV by PCR. HSV DNA was
Urticaria pigmentosa detected from CSF in the acute phase of illness from 78%
Bullous mastocytosis of patients with CNS disease.137 Older patients with herpes
Pemphigus vulgaris simplex encephalitis can have initial HSV PCR results that
Dermatitis herpetiformis are negative early in the course of illness; CSF obtained 4 to 7
Herpes gestationis days after the initial CSF samples was subsequently positive
Incontinentia pigmenti
for HSV DNA in a few patients.138 Most studies of PCR for the
Neonatal lupus
Epidermolysis bullosa
diagnosis of HSV CNS infections indicate the test is sensitive
Epidermolytic hyperkeratosis in approximately 75% to 100% of cases in small cohorts of
Acropustulosis infants.132,135-137 Specificity of the test ranges from 71% to
Neonatal bullous dermatitis 100%. The broad range of values for sensitivity and specific-
Langerhans cell histiocytosis ity of HSV PCR probably results from different study meth-
Bednar aphthae ods and disease classifications.111 In difficult cases in which
repeated CSF PCRs are negative, histologic examination and
Modified from Kohl S: Neonatal herpes simplex virus infection, Clin Perinatol viral culture of a brain tissue biopsy specimen is the most
24:129-150, 1997. definitive method of confirming the diagnosis of HSV CNS
disease. Viral cultures of CSF from a patient with neonatal
HSV CNS disease are positive in 25% to 40% of cases.
of acute neurologic deterioration, often but not always Application of PCR testing to blood specimens from
associated with the onset of seizure, and in the absence of infants with suspected HSV disease appears promising.*
intraventricular hemorrhage and metabolic causes. Using Data are insufficient at the current time to allow use of serial
PCR assay to detect viral DNA in CSF has become the gold blood PCR measurements to establish response to antiviral
standard diagnostic methodology for confirming neonatal therapy or guide decisions about the duration of therapy.
HSV CNS involvement, replacing the need for diagnosis by Cytologic methods, such as Papanicolaou, Giemsa, or
brain biopsy.134 Infants with localized CNS disease usually Tzanck staining, have a sensitivity of only approximately 60%
have increases in CSF cell counts and protein concentra- to 70%. A negative result must not be interpreted as excluding
tions and negative bacterial cultures of CSF. Noninvasive the diagnosis of HSV, and a positive result should not be the
neurodiagnostic studies such as MRI and CT can be used to sole diagnostic determinant of HSV infection in the newborn.
define sites of involvement. Intranuclear inclusions and multinucleated giant cells may
be consistent with, but not diagnostic of, HSV infection.
For diagnosis of neonatal HSV infection, the following
LABORATORY ASSESSMENT
specimens should be obtained141: (1) swab specimens from
The appropriate use of laboratory methods is essential if a the mouth, nasopharynx, conjunctivae, and anus (“surface
timely diagnosis of HSV infection is to be achieved. Virus cultures”) for HSV culture and, if desired, for HSV PCR assay
isolation remains the gold standard diagnostic method from (all surface swab specimens can be obtained with a single
non-CNS sites. HSV grows readily in cell culture. Special swab, ending with the anal swab, and placed in one viral
transport media are available that allow transport to local transport media tube); (2) specimens of skin vesicles and
or regional laboratories for culture. Cytopathogenic effects CSF for HSV culture and PCR assay; (3) whole-blood sample
typical of HSV infection usually are observed 1 to 3 days for HSV PCR assay; and (4) whole-blood sample for mea-
after inoculation. Methods of culture confirmation include suring alanine aminotransferase. The performance of PCR
fluorescent antibody staining, enzyme immunoassays, and assay on skin and mucosal specimens from neonates has not
monolayer culture with typing. Cultures that remain nega-
tive by day 5 likely will continue to remain negative. * References 102, 128, 135, 137, 139, 140.
856 SECTION III • Viral Infections

been studied; if used, surface PCR assay should be performed diagnosis, when possible. Without treatment, approxi-
in addition to, and not instead of, the gold standard surface mately 70% of infants presenting with disease localized to
culture. Positive cultures obtained from any of the surface the SEM develop involvement of the CNS or disseminated
sites more than 12 to 24 hours after birth indicate viral repli- infection. Treatment initiated after disease progression is
cation and therefore are suggestive of infant infection rather not optimal because many of these children die or are left
than merely contamination after intrapartum exposure. As with significant neurologic impairment.
with any PCR assay, false-negative and false-positive results
can occur. Whole-blood PCR assay may be of benefit in diag- ANTIVIRAL DRUGS
nosis of neonatal HSV disease, but its use should not sup-
plant the standard workup of such patients, which includes Historically, four nucleoside analogues have been used
surface cultures and CSF PCR assay; no data exist to support to treat neonatal herpes: idoxuridine, cytosine arabino-
use of serial blood PCR assay to monitor response to ther- side, vidarabine, and acyclovir. Of these, the first three are
apy. Rapid diagnostic techniques also are available, such nonspecific inhibitors of cellular and viral replication. The
as direct fluorescent antibody staining of vesicle scrapings fourth, acyclovir, is monophosphorylated by HSV-specific
or enzyme immunoassay detection of HSV antigens. These thymidine kinases and then converted to its diphosphate
techniques are as specific but slightly less sensitive than cul- and triphosphate forms by cellular enzymes. Acyclovir acts
ture. Typing HSV strains differentiates between HSV-1 and as a competitive inhibitor of HSV DNA polymerase and ter-
HSV-2 isolates. Histologic examination of lesions for pres- minates DNA chain elongation.142 Idoxuridine and cyto-
ence of multinucleated giant cells and eosinophilic intranu- sine arabinoside have no value as systemic therapy for any
clear inclusions typical of HSV (e.g., with Tzanck test) has viral infection because of toxicity and equivocal efficacy.
low sensitivity and should not be performed. Vidarabine was the first drug shown to be efficacious, with
Interpretation of negative or positive HSV cultures and decreased mortality and improved morbidity in neonatal
PCR results must depend on clinical findings. A negative HSV infections.110
PCR result for HSV in CSF in the setting of clinical, labora- In the preantiviral era, 85% of patients with disseminated
tory, or radiologic findings consistent with CNS infection neonatal HSV disease died by 1 year of age, as did 50% of
does not rule out HSV infection. It is important to continue patients with CNS neonatal HSV disease (Table 27-3).110
to use standard clinical and laboratory diagnostic methods Evaluations of two different doses of vidarabine and of a
for the evaluation of infants with possible neonatal HSV. lower dose of acyclovir (30 mg/kg/day for 10 days) docu-
In contrast to some other neonatal infections, serologic mented that both of these antiviral drugs reduce mortality
diagnosis of HSV infection has little clinical value. The inter- to comparable degrees,109,110,143 with mortality rates at 1
pretation of serologic assays is complicated by the fact that year from disseminated disease decreasing to 54% and from
transplacentally acquired maternal IgG cannot be differenti- CNS disease decreasing to 14% (see Table 27-3).109 Despite
ated from endogenously produced antibodies, making it dif- its lack of therapeutic superiority, the lower dose of acyclo-
ficult to assess the neonate’s antibody status during acute vir quickly supplanted vidarabine as the treatment of choice
infection. Serial type-specific antibody testing may be useful for neonatal HSV disease because of its favorable safety pro-
for retrospective diagnosis if a mother without a prior history file and its ease of administration. Unlike acyclovir, vidara-
of HSV infection has a primary infection late in gestation and bine had to be administered over prolonged infusion times
transfers little or no antibody to the fetus. However, thera- and in large volumes of fluid. With use of a higher dose of
peutic decisions cannot await a diagnostic approach based acyclovir (60 mg/kg/day for 21 days), 12-month mortality
on comparing acute-phase and convalescent-phase antibody is further reduced to 29% for disseminated neonatal HSV
titers. IgM production is delayed or does not occur in infected disease and to 4% for CNS HSV disease (Fig. 27-3A and
infants because of inherent immaturity in the immune B, respectively).108 Differences in mortality at 24 months
response to systemic viral infections in the newborn, and among patients treated with the higher dose of acyclovir
commercially available assays for IgM antibodies to HSV have and the lower dose of acyclovir are statistically significant
limited reliability. The results of specific laboratory tests for after stratification for disease category (CNS vs. dissemi-
HSV should be used in conjunction with clinical findings and nated) (P = .0035; odds ratio, 3.3 with 95% confidence
general laboratory tests, such as platelet counts, CSF analysis, interval of 1.5 to 7.3).108 Lethargy and severe hepatitis are
and liver function tests, to establish a disease classification. associated with mortality among patients with dissemi-
nated disease, as are prematurity and seizures in patients
with CNS disease.85
Improvements in morbidity rates with antiviral therapies
Treatment have not been as dramatic as with mortality. In the prean-
tiviral era, 50% of survivors of disseminated neonatal HSV
BACKGROUND
infections were developing normally at 12 months of age
The cumulative experience of the past 4 decades shows (see Table 27-3).110 With use of the higher dose of acyclo-
that intrapartum-acquired HSV infections are amenable to vir for 21 days, this percentage has increased to 83% (Fig.
treatment with antiviral agents, with parenteral acyclovir 27-4).108 In the case of CNS neonatal HSV disease, 33% of
being the standard of care.122 Because most infants acquire patients in the preantiviral era were developing normally at
infection at the time of delivery or shortly thereafter, anti- 12 months of age (see Table 27-3), whereas 31% of higher-
viral therapy has the potential to decrease mortality and dose acyclovir recipients develop normally at 12 months
improve long-term outcome. The benefits that antiviral today (see Fig. 27-4).108,110 Seizures at or before the time of
therapy can provide are influenced substantially by earlier initiation of antiviral therapy are associated with increased
27 • Herpes Simplex Virus Infections 857

Table 27-3 Mortality and Morbidity Outcomes Among 295 Infants With Neonatal HSV Infection, Evaluated by the National
Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group Between 1974 and 1997
EXTENT OF DISEASE TREATMENT
Placebo110 Vidarabine109 Acyclovir109 30 mg/kg/day Acyclovir108 60 mg/kg/day
DISSEMINATED DISEASE n = 13 n = 28 n = 18 n = 34
Dead 11 (85%) 14 (50%) 11 (61%) 10 (29%)
Alive 2 (15%) 14 (50%) 7 (39%) 24 (71%)
   Normal 1 (50%) 7 (50%) 3 (43%) 15 (63%)
   Abnormal 1 (50%) 5 (36%) 2 (29%) 3 (13%)
   Unknown 0 (0%) 2 (14%) 2 (29%) 6 (25%)
CENTRAL NERVOUS SYSTEM INFECTION n=6 n = 36 n = 35 n = 23
Dead 3 (50%) 5 (14%) 5 (14%) 1 (4%)
Alive 3 (50%) 31 (86%) 30 (86%) 22 (96%)
   Normal 1 (33%) 13 (42%) 8 (27%) 4 (18%)
   Abnormal 2 (67%) 17 (55%) 20 (67%) 9 (41%)
   Unknown 0 (0%) 1 (3%) 2 (7%) 9 (41%)
SKIN, EYE, OR MOUTH INFECTION n=8 n = 31 n = 54 n=9
Dead 0 (0%) 0 (0%) 0 (0%) 0 (0%)
Alive 8 (100%) 31 (100%) 54 (100%) 9 (100%)
   Normal 5 (62%) 22 (71%) 45 (83%) 2 (22%)
   Abnormal 3 (38%) 3 (10%) 1 (2%) 0 (0%)
   Unknown 0 (0%) 6 (19%) 8 (15%) 7 (78%)

Modified from Kimberlin DW: Advances in the treatment of neonatal herpes simplex infections, Rev Med Virol 11:157-163, 2001.
HSV, Herpes simplex virus.

risk of morbidity, both in patients with CNS disease and in at least twice weekly throughout the course of intrave-
patients with disseminated infection.85 Unlike disseminated nous acyclovir therapy, with consideration being given to
or CNS neonatal HSV disease, morbidity after SEM disease decreasing the dose of acyclovir or administering granulo-
has dramatically improved during the antiviral era. Before cyte colony-stimulating factor (G-CSF) if the ANC remains
use of antiviral therapies, 38% of SEM patients experienced less than 500/μL for a prolonged period of time.108
developmental difficulties at 12 months of age (see Table Determining which infants admitted to the hospital with
27-3).110 With vidarabine and lower-dose acyclovir, these presumed sepsis should be treated empirically with acyclovir
percentages were reduced to 12% and 2%, respectively.109 remains a topic of debate.146,147 In all cases of presumptive
In the high-dose acyclovir study, no SEM patients developed therapy, specimens should be obtained before initiation of
neurologic sequelae at 12 months of life (see Fig. 27-4).108 antiviral treatment for laboratory testing to guide the deci-
The improvements in mortality and morbidity achieved sion of whether to continue treatment. During the course of
with use of higher-dose acyclovir support use of acyclovir therapy, careful monitoring is important to assess the ther-
at 60 mg/kg/day delivered intravenously in three divided apeutic response. Even in the absence of clinical evidence
daily doses, as is currently recommended.108,141 The dosing of encephalitis, evaluation of the CNS should be done for
interval of intravenous acyclovir may need to be increased prognostic purposes when starting antiviral therapy. Serial
in premature infants, based upon their creatinine clear- evaluations of hepatic and hematologic parameters may
ance.144 Duration of therapy is 21 days for patients with indicate changes caused by the viral infection or by drug
disseminated or CNS neonatal HSV disease and 14 days toxicity. Intravenous acyclovir is tolerated well by infants.
for patients with HSV infection limited to the SEM.145 All Adequate hydration is necessary to minimize the risk of
patients with CNS HSV involvement should have a repeat nephrotoxicity, and dosage adjustments are necessary if
lumbar puncture at the end of intravenous acyclovir ther- renal clearance is impaired. As for all drugs, the possibility
apy to determine that the specimen is PCR negative in a of acute toxicity should be considered in any child receiving
reliable laboratory and to document the end-of-therapy CSF parenteral antiviral therapy and should be assessed by seri-
indices.85 Those persons who remain PCR positive should ally evaluating bone marrow, renal, and hepatic functions.
continue to receive intravenous antiviral therapy until PCR Acyclovir resistance has been reported in neonatal HSV
negativity is achieved.85,132 disease, but only rarely and in the form of case reports.
The primary apparent toxicity associated with the use One infant with acute HSV infection of the larynx in the
of intravenous acyclovir administered at 60 mg/kg/day is newborn period developed antiviral resistance during the
neutropenia, with approximately one fifth of patients devel- course of therapy; in this case, the initial isolate was not
oping an absolute neutrophil count (ANC) of less than or inhibited by acyclovir, although the source of this infection
equal to 1000/μL.108 Although the neutropenia resolves could not be explained.148 Acyclovir resistance has also
either during continuation of intravenous acyclovir or after been reported in a premature infant with cutaneous and
its cessation, it is prudent to monitor neutrophil counts CNS disease caused by an initially acyclovir-susceptible
858 SECTION III • Viral Infections

1.0
0.9
0.8

Proportion surviving
0.7
0.6
0.5
0.4
0.3 30 mg/kg/day (n = 18)*
45 mg/kg/day (n = 7)
0.2
60 mg/kg/day (n = 34)
0.1
*Historical cohort
0
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24
A Months

1.0
0.9
0.8
Proportion surviving

0.7
0.6
0.5
0.4
0.3 30 mg/kg/day (n = 35)*
45 mg/kg/day (n = 5)
0.2
Figure 27-3 Mortality rates for patients with dis- 60 mg/kg/day (n = 23)
seminated disease (A) and central nervous system 0.1
disease (B) depending on dose of acyclovir. (Data *Historical cohort
from Kimberlin DW, Lin CY, Jacobs RF, et al: Safety 0
and efficacy of high-dose intravenous acyclovir in 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24
the management of neonatal herpes simplex virus
infections, Pediatrics 108:230-238, 2001.) B Months

Severe Moderate Mild Normal primary HSV-2 infection developed an acyclovir-resistant


N=6 N=2 N = 28 N = 13 N = 5 N = 18 mutant during acyclovir therapy for disseminated HSV
100 infection and eventually died. The use of steroids to treat
90 blood pressure instability may have hampered this infant’s
80 immune response to infection further.150 Emergence of
70
Percentage

60 viral resistance to acyclovir also has been described in


50 patients requiring prolonged or repeated treatment with
40 this drug. One infant who was given long-term oral acy-
30 clovir for suppression of recurrences during the first 6
20 months of life had a resistant HSV isolated from a lesion
10
0 after therapy was discontinued, but subsequent isolates
30 60 30 60 30 60 were susceptible.151 However, isolates of HSV recovered
SEM CNS Disseminated from infants who received intravenous acyclovir for cuta-
Figure 27-4 Morbidity among patients with known outcomes after 12 neous disease in the newborn period and had subsequent
months of life. CNS, Central nervous system; SEM, skin, eyes, or mouth. recurrent cutaneous lesions typically remain sensitive to
(Modified from Kimberlin DW, Lin CY, Jacobs RF, et al: Safety and efficacy acyclovir,152 and antiviral resistance generally does not
of high-dose intravenous acyclovir in the management of neonatal herpes explain the failure of infants with the disseminated or CNS
simplex virus infections, Pediatrics 108:230-238, 2001.)
form of the disease to respond well to antiviral therapy.
Rather, clinical deterioration despite appropriate therapy
HSV. The infant developed recurrent disseminated HSV and supportive care can almost always be attributed to
infection 8 days after a 21-day course of acyclovir. The virus-induced destruction of cells compromising infected
virus isolated at the onset of recurrent symptoms was organs, such as liver or brain, or irreversible changes,
found to lack thymidine kinase activity on the basis of a such as disseminated intravascular coagulopathy.
frameshift mutation in the thymidine kinase gene.149 The NIAID CASG recently completed a multicenter, ran-
Another infant born to a mother with severe systemic domized, placebo-controlled, double-blind study of 6 months
27 • Herpes Simplex Virus Infections 859

of oral acyclovir suppressive therapy after parenteral treat- No other forms of adjunctive therapy are useful for
ment of acute disease.105 Subjects were stratified by CNS treating neonatal HSV infections. Various experimental
versus SEM disease and were randomized to either oral acy- modalities, including interferon, immunomodulators, and
clovir at 300 mg/m2/dose administered three times per day immunization, have been attempted, but none has pro-
or to a matching placebo. If a subject had two cutaneous duced demonstrable effects.
recurrences, the blinded suppression drug was stopped and
open-label suppression was provided. Subjects randomized LONG-TERM MANAGEMENT OF INFECTED
to receive acyclovir had significantly higher (better) mean INFANTS
Bayley Mental Scores at 1 year (88.24 vs. 68.12; P = .046).
Distribution of neurologic outcomes was: 69% normal, 6% With the advent of antiviral therapy, an increasing num-
mild impairment, 6% moderate impairment, and 19% severe ber of newborns with HSV infection are surviving and
impairment for subjects randomized to acyclovir suppression require careful long-term follow-up. The most common
versus 33%, 8%, 25%, and 33%, respectively, for subjects complications of neonatal HSV infection include neuro-
randomized to placebo. Of note, this distribution of outcomes logic and ocular sequelae that may be detected only on
was virtually identical to that from a small uncontrolled case long-term follow-up. It is necessary that these children
series (N = 16) of oral acyclovir suppression after neonatal receive serial long-term evaluation from qualified pediatric
HSV CNS disease in which patients received five times the specialists in these areas, which should include neurode-
dose for four times the duration (69% having normal out- velopmental, ophthalmologic, and hearing assessments.
comes in both studies).153 In the CASG study, the 12-month Recurrent skin vesicles are present in many children, includ-
Bayley Mental Scores were incrementally higher because ing children who did not have obvious mucocutaneous dis-
subjects were on active suppression for longer periods of ease during the acute phase of the clinical illness. Skin vesicles
time. For subjects in the SEM study, no differences in Bayley provide a potential source for transmission of infection to other
Mental Scores were noted. Combining the 74 subjects with children or adults who have direct contact with these infants.
either CNS or SEM disease, time to a second cutaneous HSV The increasing use of daycare for children, including children
recurrence was statistically significantly longer for the group surviving neonatal HSV infections, stimulates many ques-
randomized to acyclovir suppression. Time to first ANC less tions from daycare providers about these children. There is
than or equal to 500 cells/mm3 was not statistically different some risk that children with recurrent HSV skin lesions would
for those subjects randomized to acyclovir versus placebo, transmit the virus to other children in this environment. The
although a trend for neutropenia in those who received acy- most reasonable recommendation in this situation is to cover
clovir was noted (P = .09). In total, 25% and 20% of subjects the lesions to prevent direct contact. HSV-1 is much more
on the CNS study and SEM study, respectively, who were likely to be present in the daycare environment in the form of
randomized to acyclovir developed an ANC of less than or asymptomatic infection or gingivostomatitis. In both cases,
equal to 500 cells/mm3, compared with 5% and 7% of pla- virus is present in the mouth and pharynx, and the frequent
cebo recipients in the respective trials. Neutropenia resolved exchange of saliva and other respiratory droplets that occurs
in all affected subjects both with and without cessation of among children in this setting makes this route of transmis-
suppressive therapy, and none had associated complica- sion more likely. Education of daycare workers and the gen-
tions.105 These results have led to routine use of oral acyclovir eral public about herpesvirus infections, their implications,
suppressive therapy for the 6 months after treatment of acute and the frequency with which they occur in the population as
neonatal HSV disease to improve neurodevelopmental a whole can calm fears and correct common misconceptions.
outcomes in infants with HSV CNS disease and to prevent Parents of children with neonatal HSV infection often
skin recurrences in infants with any disease classification of have significant feelings of guilt. Parents often require
neonatal HSV.141 support from psychologists, psychiatrists, or counselors.
The family physician or pediatrician can provide a valu-
able supportive role to the family in this situation. Most
OTHER ISSUES IN ACUTE MANAGEMENT
parents and many physicians are unaware of the high
Neonates with HSV infection should be hospitalized and man- prevalence of HSV-2 infection and the increasing inci-
aged with contact precautions if mucocutaneous lesions are dence of HSV-1 genital infection in the United States and
present. Many infants with this infection have life-threatening of the lifelong persistence and subclinical nature of these
problems, including disseminated intravascular coagulation, infections. Concern about the risk of fetal and neonatal
shock, and respiratory failure, and they require supportive infection during subsequent pregnancies is often a major
care that is available only at tertiary medical centers. issue that can be addressed effectively based on the low
There is no indication that administration of immu- risks as proven from large, prospective studies.
noglobulin or hyperimmunoglobulin is of value for the
treatment of neonatal HSV infection. Although a series of
studies have suggested that the quantity of transplacen-
tal neutralizing antibodies affects the attack rate among
Prevention
exposed infants and may influence the initial disease BACKGROUND
manifestations, the presence of antibodies may or may not
influence the subsequent course of infection.60,61,65,66,97 Despite the progress that has been made in antiviral treat-
The administration of standard preparations of intrave- ment of neonatal HSV infection, the ideal approach is to pre-
nous immunoglobulin does not enhance the titers of func- vent the exposure of infants to active maternal infection at
tional antibodies against HSV in infants with low birth the time of delivery. Genital infections caused by HSV often
weight.154 are clinically silent when they are acquired as new infections
860 SECTION III • Viral Infections

Table 27-4 Projected Risk of Transmission of Herpes infection, it is appropriate to deliver infants of women who
Simplex Virus Type 2 (HSV-2) From Mothers to Infants at have a history of recurrent genital herpes but who have no
Delivery in a Cohort of 100,000 Pregnant Women active clinical disease at delivery by the vaginal route.67 An
analysis of the occurrence of HSV infections in infants in
25% with past HSV-2 infection 75% susceptible to HSV-2 infection California showed no change from 1985 to 1995, despite a
25,000 women 75,000 women documented decrease in deliveries by cesarean section and an
1.5% reactivation at delivery 0.02% seroconversion/week increase in the proportion of women with a previous diagnosis
375 women with reactivation 30 women with infection <2 weeks of genital herpes whose infants were delivered vaginally.155
before delivery A culture for HSV obtained at the time of delivery may be
<5% risk of transmission 50% risk of transmission useful in establishing whether the virus was present at deliv-
19 infected infants 15 infected infants ery to facilitate recognition of neonatal infection if it occurs.
The value of this approach has not been established, however.
Data from Arvin AM: The epidemiology of perinatal herpes simplex infec-
tions. In Stanberry L, editor: Genital and neonatal herpes, New York, 1996,
Alternative diagnostic approaches, such as those based on PCR
John Wiley & Sons, pp 179-192. assay to detect virus, may ultimately expedite identification of
women at risk for delivering infected infants.156,157 Evidence
indicates that detection of viral presence in genital samples by
and when the virus reactivates. With the high prevalence of PCR assay is more sensitive than culture methods. The signifi-
HSV-2 infection in the U.S. population and the increasing cance of a positive PCR result in predicting risk of transmission
practice of oral-genital sexual contact, women are at risk of HSV to the infant is unknown, however.158 Viral DNA can
for acquiring new genital infections during pregnancy. The persist for a longer interval than infectious virus.
problem of asymptomatic genital HSV infection means that The utility of suppressive therapy of genital herpes in
the transmission of HSV from mothers to infants cannot be women with a known history of recurrent infection remains
eliminated even with the best obstetric management. a question for clinical investigation because of risk-benefit
Sequential genital cultures during the last weeks of ges- considerations. Some studies indicate that prophylactic
tation in women with a history of genital herpes do not acyclovir (400 mg three times daily) reduces the number of
predict the infant’s risk of exposure at delivery59 because genital lesions from HSV. Despite prophylaxis, a few women
of the usually brief duration of asymptomatic shedding continue to have virus detectable by PCR assay.158 Use of
and the time required for the culture to become positive. prophylactic valacyclovir also reduced clinical HSV recur-
Because of the attention of the lay press to the devastating rences but did not decrease shedding of HSV within 7 days
outcome of neonatal HSV infections, many women who of delivery compared with placebo.159 Even with suppres-
know that they have genital herpes experience severe anx- sive therapy, the potential for neonatal HSV infection is not
iety about the potential risks to the fetus and the newborn. eliminated entirely, and cases of neonatal HSV after delivery
As a consequence, these women may have an unneces- to women on antiviral suppression have been reported.160
sarily high frequency of cesarean deliveries. The risk of Based on limited scientific evidence, the American College of
neonatal HSV infection in the newborn is approximately Obstetricians and Gynecologists recommends that women
equivalent for women who have no prior history of genital with active recurrent genital herpes should be offered sup-
herpes or a partner with known infection (Table 27-4). pressive viral therapy at or beyond 36 weeks of gestation
Although development of a vaccine has received sub- until delivery.67 Universal antenatal type-specific HSV
stantial industry and government support, a recent large screening to prevent neonatal HSV is not recommended.161
clinical trial of the vaccine product that was farthest along
in development failed to show protection.31 Although MANAGEMENT OF INFANTS OF MOTHERS WITH
development efforts continue, it will be many years, if GENITAL HERPES
ever, before a vaccine for genital herpes is available.
Infants of mothers with histories of genital herpes delivered
MANAGEMENT OF PREGNANT WOMEN WITH vaginally or by cesarean section and whose mothers have
no evidence of active genital herpetic infection are at low
KNOWN GENITAL HERPES
risk for acquiring neonatal HSV infection. These infants
Women who have a history of recurrent genital herpes need no special evaluation during the newborn period.
should be reassured that the risk of fetal or neonatal infec- Infants delivered to women with clinically apparent
tion is very low. Intrauterine HSV infections are very rare, genital herpetic lesions are at known risk for acquisition
with an estimated overall risk of 1 in 300,000 pregnancies.77 of neonatal HSV. However, the magnitude of the risk is
Information about the risk of exposure to asymptomatic viral influenced by the category of maternal infection, with
reactivation at delivery derived from six large-scale prospec- women with first episode primary or nonprimary genital
tive studies is sufficient to conclude that the incidence of infection being at 30 to 50 times the likelihood of having
asymptomatic reactivation in these women is about 2% and their babies acquire neonatal HSV than women with recur-
that the attack rate for their exposed infants is approximately rent genital infection. The American Academy of Pedi-
2% or less. Because laboratory methods cannot be used to atrics (AAP) recently released a clinical report that seeks
detect asymptomatic infection in a timely manner, the cur- to provide guidance on the management of these babies,
rent approach to management is to perform a careful vaginal using serologic and virologic testing to ascertain the level
examination at presentation and to elect cesarean delivery if of risk and then providing recommendations on manage-
the mother has signs or symptoms of recurrent genital herpes ment of the infants accordingly.14 The algorithms from this
at the onset of labor. Given the low probability of neonatal clinical report are reproduced in Figures 27-5 and 27-6.
27 • Herpes Simplex Virus Infections 861

Asymptomatic neonate after vaginal or cesarean delivery to mother with visible genital
lesions that are characteristic of HSV

Obstetric provider obtains swab of lesion for HSV PCR and culture
Type all positives

Maternal history of genital HSV preceding pregnancy?

No Yes

Send maternal type specific serology for At ~24 hours of age* obtain from the neonate:
HSV-1 and HSV-2 antibodies, if test assays • HSV surface† cultures (and PCRs if desired)
are available at the delivery hospital • HSV blood PCR‡
If infant remains asymptomatic, do not start acyclovir

At ~24 hours of age* obtain from the neonate:


• HSV surface† cultures (and PCRs if desired)
• HSV blood PCR‡
• CSF cell count, chemistries, and HSV PCR Neonatal surface Neonatal surface
• Serum ALT cultures negative, AND cultures positive, OR
Start IV acyclovir at 60 mg/kg/day in three divided doses blood and surface blood or surface PCRs
PCRs negative positive

Determine maternal HSV infection classification


Educate family on signs Obtain CSF for cell
and symptoms of count, chemistries, and
neonatal HSV disease HSV PCR. Send serum
and follow closely§ ALT. Start IV acyclovir
at 60 mg/kg/day in three
First episode primary or divided doses
Recurrent infection
First episode nonprimary

Go to Figure 27-6.

Neonatal virology studies


negative (PCRs negative; Neonatal PCRs or viral
viral cultures negative at cultures positive
48-72 hours)

Go to Figure 27-6.
Stop acyclovir. Educate Go to Figure 27-6.
family for signs and
symptoms of neonatal
HSV disease and follow
closely§

This algorithm should be applied only in facilities where access to PCR and type-specific serologic testing is readily available and turnaround time for test
results is appropriately short. In situations where this is not possible, the approach detailed in the algorithm will have limited, and perhaps no, applicability.
*Evaluation and treatment is indicated before 24 hours of age if the infant develops signs and symptoms of neonatal HSV disease. In addition, immediate
evaluation and treatment may be considered if:
• There is prolonged rupture of membranes (>4-6 hours)
• The infant is premature (<37 weeks' gestation)
†Conjunctivae, mouth, nasopharynx, and rectum, and scalp electrode site, if present
‡HSV blood PCR is not used for assignment of disease classification
§Discharge after 48 hours of negative HSV cultures (and negative PCRs) is acceptable if other discharge criteria have been met, there is ready access to
medical care, and a person who is able to comply fully with instructions for home observation will be present. If any of these conditions is not met, the
infant should be observed in the hospital until HSV cultures are finalized as negative or are negative for 96 hours after being set up in cell culture,
whichever is shorter.

Figure 27-5 Algorithm for the evaluation of asymptomatic neonates after vaginal or cesarean delivery to women with active genital herpes lesions. ALT,
Alanine aminotransferase; CSF, cerebrospinal fluid; HSV, herpes simplex virus; IV, intravenous; PCR, polymerase chain reaction. (Modified from Kimberlin
DW, Baley J: Guidance on management of asymptomatic neonates born to women with active genital herpes lesions, Pediatrics 131:e635-e646, 2013.)
862 SECTION III • Viral Infections

Patient remains asymptomatic, CSF indices not indicative of infection,


CSF and blood PCR negative, and normal serum ALT*

No Yes

Treatment of infection and proven disease Preemptive therapy of infection but no


Treat with intravenous acyclovir at 60 mg/kg/day proven disease
in three divided daily doses for 14 days (SEM) or Treat with intravenous acyclovir at 60 mg/kg/day
21 days (CNS or disseminated) in three divided daily doses for 10 days
Additional evaluation may be indicated

*Serum ALT values in neonates may be elevated due to noninfectious


Repeat CSF HSV PCR near end of 21-day
causes (delivery-related perfusion, etc.). For this algorithm, ALT values
course of treatment† more than two times the upper limit of normal may be considered
suggestive of neonatal disseminated HSV disease for HSV-exposed
neonates
†If evidence of CNS disease at beginning of therapy

Positive Negative

Continue intravenous D/C intravenous


acyclovir for 7 days acyclovir after 21-day
more treatment course

Figure 27-6 Algorithm for the treatment of asymptomatic neonates after vaginal or cesarean delivery to women with active genital herpes lesions.
ALT, Alanine aminotransferase; CSF, cerebrospinal fluid; D/C, discontinue; HSV, herpes simplex virus; PCR, polymerase chain reaction; SEM, skin, eyes,
or mouth. (Modified from Kimberlin DW, Baley J: Guidance on management of asymptomatic neonates born to women with active genital herpes lesions,
Pediatrics 131:e635-e646, 2013.)

This report seeks to move beyond the recommendations Conclusion


that preceded its publication in 2013 that “some experts”
do one thing and “other experts” do something else. Parts Neonatal HSV infection is a life-threatening disease in the
of the report use expert consensus in some of its recom- newborn. With an increasing prevalence of HSV-1 geni-
mendations, such as preemptive therapy with parenteral tal herpes, a continuing high prevalence of HSV-2 genital
acyclovir in neonates delivered to women with first-epi- herpes, and the recognition that many infections are com-
sode primary and nonprimary infections because of their pletely asymptomatic in the mother, pediatricians, neo-
substantial risk of neonatal HSV disease, even though natologists, obstetricians, and family practitioners must
such an approach has not been, and never will be, clini- continue to remain vigilant to infants whose symptoms
cally investigated. However, these expert consensus rec- may be compatible with HSV infections.
ommendations are grounded in the risk assessments
derived from categorizing the mother’s infection as first- Acknowledgments
episode primary, first-episode nonprimary, and recurrent. The databases on clinical presentations, diagnosis, and anti-
The clinical report was written by the AAP Committee on viral treatment of neonatal HSV infections have been gener-
Infectious Diseases and Committee on Fetus and Newborn ated through the efforts of the NIAID Collaborative Antiviral
and was supported by the American College of Obstetri- Study Group for more than 35 years, with support from the
cians and Gynecologists. National Institute of Allergy and Infectious Diseases. The
An issue of frequent concern is whether the mother Institute also has supported prognostic studies of HSV infec-
with an active genital HSV infection at delivery should tion in pregnancy and newborns in the United States.
be isolated from her infant after delivery. Women with
recurrent orolabial HSV infection and cutaneous HSV References
infections at other sites (e.g., breast lesions) are at simi­lar 1. Hass M: Hepatoadrenal necrosis with intranuclear inclusion bodies:
risk for transmission of virus to their newborns. Because report of a case, Am J Pathol 11:127, 1935.
2. Batignani A: Conjunctivite da virus erpetico in neonato, Boll Ocul
transmission occurs by direct contact with the virus, 13:1217, 1934.
appropriate precautions by the mother, including careful 3. Nahmias AJ, Dowdle WR: Antigenic and biologic differences in her-
hand washing before touching the infant, should prevent pesvirus hominis, Prog Med Virol 10:110-159, 1968.
any need to separate mother and child. In some cases, 4. Roizman B, Knipe DM, Whitley RJ: Herpes simplex viruses. In Knipe
it is possible to have the exposed infant room-in with DM, Howley PM, editors: Fields virology, ed 5, vol 2, Philadelphia,
2007, Lippincott Williams & Wilkins, pp 2501-2601.
the mother as a means of isolating the infant from other 5. Spear PG: Membrane fusion induced by herpes simplex virus. In
newborns. Breastfeeding is contraindicated if the mother Bentz J, editor: Viral fusion mechanisms. Boca Raton, Fla, 1993, CRC
has vesicular lesions involving the breast. Press, pp 201-232.
27 • Herpes Simplex Virus Infections 863

6. Roizman B, Norrild B, Chan C, et al: Identification and preliminary 32. Bernstein DI, Bellamy AR, Hook EW 3rd, et al: Epidemiology, clinical
mapping with monoclonal antibodies of a herpes simplex virus 2 presentation, and antibody response to primary infection with her-
glycoprotein lacking a known type 1 counterpart, Virology 133: pes simplex virus type 1 and type 2 in young women, Clin Infect Dis
242-247, 1984. 56:344-351, 2013.
7. Markert JM, Gillespie GY, Weichselbaum RR, et al: Genetically 33. Wald A, Zeh J, Selke S, et al: Virologic characteristics of subclini-
engineered HSV in the treatment of glioma: a review, Rev Med Virol cal and symptomatic genital herpes infections, N Engl J Med 333:
10:17-30, 2000. 770-775, 1995.
8. York IA, Roop C, Andrews DW, et al: A cytosolic herpes simplex 34. Frenkel LM, Garratty EM, Shen JP, et al: Clinical reactivation of herpes
virus protein inhibits antigen presentation to CD8+ T lymphocytes, simplex virus type 2 infection in seropositive pregnant women with no
Cell 77:525-535, 1994. history of genital herpes, Ann Intern Med 118:414-418, 1993.
9. Jugovic P, Hill AM, Tomazin R, et al: Inhibition of major histo- 35. Fleming DT, McQuillan GM, Johnson RE, et al: Herpes simplex
compatibility complex class I antigen presentation in pig and pri- virus type 2 in the United States, 1976 to 1994, N Engl J Med 337:
mate cells by herpes simplex virus type 1 and 2 ICP47, J Virol 72: 1105-1111, 1997.
5076-5084, 1998. 36. Xu F, Sternberg MR, Kottiri BJ, et al: Trends in herpes simplex
10. Sullender WM, Yasukawa LL, Schwartz M, et al: Type-specific anti- virus type 1 and type 2 seroprevalence in the United States, JAMA
bodies to herpes simplex virus type 2 (HSV-2) glycoprotein G in preg- 296:964-973, 2006.
nant women, infants exposed to maternal HSV-2 infection at delivery, 37. Smith JS, Robinson NJ: Age-specific prevalence of infection with
and infants with neonatal herpes, J Infect Dis 157:164-171, 1988. herpes simplex virus types 2 and 1: a global review, J Infect Dis 186:
11. Coleman RM, Pereira L, Bailey PD, et al: Determination of herpes S3-S28, 2002.
simplex virus type-specific antibodies by enzyme-linked immunosor- 38. Buchman TG, Roizman B, Nahmias AJ: Demonstration of exogenous
bent assay, J Clin Microbiol 18:287-291, 1983. genital reinfection with herpes simplex virus type 2 by restriction
12. Wald A, Ashley-Morrow R: Serological testing for herpes simplex virus endonuclease fingerprinting of viral DNA, J Infect Dis 140:295-304,
(HSV)-1 and HSV-2 infection, Clin Infect Dis 35:S173-S182, 2002. 1979.
13. Ashley RL, Wu L, Pickering JW, et al: Premarket evaluation of a 39. Schmidt OW, Fife KH, Corey L: Reinfection is an uncommon occur-
commercial glycoprotein G-based enzyme immunoassay for herpes rence in patients with symptomatic recurrent genital herpes, J Infect
simplex virus type-specific antibodies, J Clin Microbiol 36:294-295, Dis 149:645-646, 1984.
1998. 40. Lakeman AD, Nahmias AJ, Whitley RJ: Analysis of DNA from recur-
14. Kimberlin DW, Baley J: Guidance on management of asymptomatic rent genital herpes simplex virus isolates by restriction endonuclease
neonates born to women with active genital herpes lesions, Pediat- digestion, Sex Transm Dis 13:61-66, 1986.
rics 131:e635-e646, 2013. 41. Boucher FD, Yasukawa LL, Bronzan RN, et al: A prospective eval-
15. Cushing H: Surgical aspects of major neuralgia of trigeminal nerve: uation of primary genital herpes simplex virus type 2 infections
report of 20 cases of operation upon the gasserian ganglion with acquired during pregnancy, Pediatr Infect Dis J 9:499-504, 1990.
anatomic and physiologic notes on the consequence of its removal, 42. Brown ZA, Vontver LA, Benedetti J, et al: Genital herpes in preg-
JAMA 44:1002-1009, 1905. nancy: risk factors associated with recurrences and asymptomatic
16. Goodpasture EW: Herpetic infection, with especial reference to viral shedding, Am J Obstet Gynecol 153:24-30, 1985.
involvement of the nervous system 1929, Medicine (Baltimore) 43. Prober CG, Hensleigh PA, Boucher FD, et al: Use of routine viral cul-
72:125-132, 1993; discussion 133–135. tures at delivery to identify neonates exposed to herpes simplex virus,
17. Carton CA, Kilbourne ED: Activation of latent herpes simplex by N Engl J Med 318:887-891, 1988.
trigeminal sensory-root section, N Engl J Med 246:172-176, 1952. 44. Kulhanjian JA, Soroush V, Au DS, et al: Identification of women at
18. Pazin GJ, Armstrong JA, Lam MT, et al: Prevention of reactivated unsuspected risk of primary infection with herpes simplex virus type
herpes simplex infection by human leukocyte interferon after opera- 2 during pregnancy, N Engl J Med 326:916-920, 1992.
tion on the trigeminal root, N Engl J Med 301:225-230, 1979. 45. Brown ZA, Vontver LA, Benedetti J, et al: Effects on infants of a first epi-
19. Cunningham AL, Diefenbach RJ, Miranda-Saksena M, et al: The sode of genital herpes during pregnancy, N Engl J Med 317:1246-1251,
cycle of human herpes simplex virus infection: virus transport and 1987.
immune control, J Infect Dis 194(Suppl 1):S11-S18, 2006. 46. Brown ZA, Benedetti J, Ashley R, et al: Neonatal herpes simplex virus
20. Stevens JG, Cook ML: Latent herpes simplex virus in spinal ganglia of infection in relation to asymptomatic maternal infection at the time
mice, Science 173:843-845, 1971. of labor, N Engl J Med 324:1247-1252, 1991.
21. Rock DL, Fraser NW: Detection of HSV-1 genome in central nervous 47. Wald A, Zeh J, Selke S, et al: Reactivation of genital herpes simplex
system of latently infected mice, Nature 302:523-525, 1983. virus type 2 infection in asymptomatic seropositive persons, N Engl J
22. Baringer JR: Recovery of herpes simplex virus from human sacral Med 342:844-850, 2000.
ganglions, N Engl J Med 291:828-830, 1974. 48. Tronstein E, Johnston C, Huang ML, et al: Genital shedding of herpes
23. Xu F, Lee FK, Morrow RA, et al: Seroprevalence of herpes simplex simplex virus among symptomatic and asymptomatic persons with
virus type 1 in children in the United States, J Pediatr 151:374-377, HSV-2 infection, JAMA 305:1441-1449, 2011.
2007. 49. Brown ZA, Wald A, Morrow RA, et al: Effect of serologic status and
24. Xu F, Markowitz LE, Gottlieb SL, et al: Seroprevalence of herpes sim- cesarean delivery on transmission rates of herpes simplex virus from
plex virus types 1 and 2 in pregnant women in the United States, Am mother to infant, JAMA 289:203-209, 2003.
J Obstet Gynecol 196:43.e1-43.e6, 2007. 50. Engelberg R, Carrell D, Krantz E, et al: Natural history of genital
25. Wentworth BB, Alexander ER: Seroepidemiology of infections due herpes simplex virus type 1 infection, Sex Transm Dis 30:174-177,
to members of the herpesvirus group, Am J Epidemiol 94:496-507, 2003.
1971. 51. Gardella C, Brown ZA: Managing genital herpes infections in preg-
26. Arvin AM: The epidemiology of perinatal herpes simplex infections. nancy, Cleve Clin J Med 74:217-224, 2007.
In Stanberry L, editor: Genital and neonatal herpes, New York, 1996, 52. Wald A, Corey L, Cone R, et al: Frequent genital herpes simplex virus
John Wiley and Sons, pp 179-192. 2 shedding in immunocompetent women. Effect of acyclovir treat-
27. Nahmias AJ, Josey WE, Naib ZM, et al: Antibodies to Herpesvirus ment, J Clin Invest 99:1092-1097, 1997.
hominis types 1 and 2 in humans. I. Patients with genital herpetic 53. Flewett TH, Parker RG, Philip WM: Acute hepatitis due to herpes
infections, Am J Epidemiol 91:539-546, 1970. simplex virus in an adult, J Clin Pathol 22:60-66, 1969.
28. Bradley H, Markowitz LE, Gibson T, et al: Seroprevalence of herpes 54. Young EJ, Killam AP, Greene JF Jr: Disseminated herpesvirus infec-
simplex virus types 1 and 2—United States, 1999-2010, J Infect Dis tion. Association with primary genital herpes in pregnancy, JAMA
209:325-333, 2013. 235:2731-2733, 1976.
29. Kimberlin DW: The scarlet H, J Infect Dis 209:315-317, 2013. 55. Hensleigh PA, Andrews WW, Brown Z, et al: Genital herpes during
30. Roberts CM, Pfister JR, Spear SJ: Increasing proportion of herpes pregnancy: inability to distinguish primary and recurrent infections
simplex virus type 1 as a cause of genital herpes infection in college clinically, Obstet Gynecol 89:891-895, 1997.
students, Sex Transm Dis 30:797-800, 2003. 56. Nahmias AJ, Josey WE, Naib ZM, et al: Perinatal risk associated with
31. Belshe RB, Leone PA, Bernstein DI, et al: Efficacy results of a trial of a maternal genital herpes simplex virus infection, Am J Obstet Gynecol
herpes simplex vaccine, N Engl J Med 366:34-43, 2012. 110:825-837, 1971.
864 SECTION III • Viral Infections

57. Brown ZA, Selke S, Zeh J, et al: The acquisition of herpes simplex 83. Arvin AM: Fetal and neonatal infections. In Nathanson N, Murphy
virus during pregnancy, N Engl J Med 337:509-515, 1997. F, editors: Viral pathogenesis, New York, 1996, Lippincott-Raven,
58. Dietrich YM, Napolitano PG: Acyclovir treatment of primary herpes pp 801-814.
in pregnancy complicated by second trimester preterm premature 84. Alanen A, Hukkanen V: Herpes simplex virus DNA in amniotic fluid
rupture of membranes with term delivery: case report, Am J Perinatol without neonatal infection, Clin Infect Dis 30:363-367, 2000.
19:235-238, 2002. 85. Kimberlin DW, Lin CY, Jacobs RF, et al: Natural history of neona-
59. Arvin AM, Hensleigh PA, Prober CG, et al: Failure of antepartum tal herpes simplex virus infections in the acyclovir era, Pediatrics
maternal cultures to predict the infant’s risk of exposure to herpes 108:223-229, 2001.
simplex virus at delivery, N Engl J Med 315:796-800, 1986. 86. Douglas J, Schmidt O, Corey L: Acquisition of neonatal HSV-1 infec-
60. Prober CG, Sullender WM, Yasukawa LL, et al: Low risk of herpes tion from a paternal source contact, J Pediatr 103:908-910, 1983.
simplex virus infections in neonates exposed to the virus at the time 87. Hammerberg O, Watts J, Chernesky M, et al: An outbreak of herpes
of vaginal delivery to mothers with recurrent genital herpes simplex simplex virus type 1 in an intensive care nursery, Pediatr Infect Dis
virus infections, N Engl J Med 316:240-244, 1987. 2:290-294, 1983.
61. Whitley RJ, Corey L, Arvin A, et al: Changing presentation of her- 88. Light IJ: Postnatal acquisition of herpes simplex virus by the new-
pes simplex virus infection in neonates, J Infect Dis 158:109-116, born infant: a review of the literature, Pediatrics 63:480-482, 1979.
1988. 89. Linnemann CC Jr, Buchman TG, Light IJ, et al: Transmission of her-
62. Kimberlin DW, Rouse DJ: Genital herpes, N Engl J Med 350:1970- pes-simplex virus type 1 in a nursery for the newborn. Identification
1977, 2004. of viral isolates by D.N.A. “fingerprinting”, Lancet 1:964-966, 1978.
63. Corey L, Adams HG, Brown ZA, et al: Genital herpes simplex virus 90. Yeager AS, Ashley RL, Corey L: Transmission of herpes simplex virus
infections: clinical manifestations, course, and complications, Ann from father to neonate, J Pediatr 103:905-907, 1983.
Internl Med 98:958-972, 1983. 91. Sullivan-Bolyai JZ, Fife KH, Jacobs RF, et al: Disseminated neonatal
64. Johnston C, Magaret A, Selke S, et al: Herpes simplex virus viremia herpes simplex virus type 1 from a maternal breast lesion, Pediatrics
during primary genital infection, J Infect Dis 198:31-34, 2008. 71:455-457, 1983.
65. Yeager AS, Arvin AM, Urbani LJ, et al: Relationship of antibody to 92. Hatherley LI, Hayes K, Jack I: Herpes virus in an obstetric hospital.
outcome in neonatal herpes simplex virus infections, Infect Immun II: Asymptomatic virus excretion in staff members, Med J Aust 2:
29:532-538, 1980. 273-275, 1980.
66. Sullender WM, Miller JL, Yasukawa LL, et al: Humoral and cell- 93. Hatherley LI, Hayes K, Jack I: Herpes virus in an obstetric hospi-
mediated immunity in neonates with herpes simplex virus infection, tal. III: Prevalence of antibodies in patients and staff, Med J Aust 2:
J Infect Dis 155:28-37, 1987. 325-329, 1980.
67. ACOG Committee on Practice Bulletins: ACOG Practice Bulletin: 94. Burchett SK, Corey L, Mohan KM, et al: Diminished interferon-gamma
Management of herpes in pregnancy. No. 82 June 2007. Clinical and lymphocyte proliferation in neonatal and postpartum primary
management guidelines for obstetrician-gynecologists, Obstet Gyne- herpes simplex virus infection, J Infect Dis 165:813-818, 1992.
col 109:1489-1498, 2007. 95. Chilmonczyk BA, Levin MJ, McDuffy R, et al: Characterization of
68. Stone KM, Brooks CA, Guinan ME, et al: National surveillance the human newborn response to herpesvirus antigen, J Immunol
for neonatal herpes simplex virus infections, Sex Transm Dis 16: 134:4184-4188, 1985.
152-156, 1989. 96. Kohl S, Harmon MW: Human neonatal leukocyte interferon pro-
69. Kaye EM, Dooling EC: Neonatal herpes simplex meningoencepha- duction and natural killer cytotoxicity in response to herpes simplex
litis associated with fetal monitor scalp electrodes, Neurology 31: virus, J Interferon Res 3:461-463, 1983.
1045-1047, 1981. 97. Kohl S, West MS, Prober CG, et al: Neonatal antibody-dependent
70. Parvey LS, Ch’ien LT: Neonatal herpes simplex virus infection intro- cellular cytotoxic antibody levels are associated with the clinical
duced by fetal-monitor scalp electrodes, Pediatrics 65:1150-1153, presentation of neonatal herpes simplex virus infection, J Infect Dis
1980. 160:770-776, 1989.
71. Nahmias AJ, Keyserling HL, Kerrick GM: Herpes simplex. In 98. Pass RF, Dworsky ME, Whitley RJ, et al: Specific lymphocyte blas-
­Remington JS, Klein JO, editors: Infectious diseases of the fetus and new- togenic responses in children with cytomegalovirus and herpes
born infants, ed 2, Philadelphia, 1983, Saunders, pp 636-678. simplex virus infections acquired early in infancy, Infect Immun
72. Sullivan-Bolyai J, Hull HF, Wilson C, et al: Neonatal herpes sim- 34:166-170, 1981.
plex virus infection in King County, Washington. Increasing 99. Kohl S: Neonatal herpes simplex virus infection, Clin Perinatol
incidence and epidemiologic correlates, JAMA 250:3059-3062, 24:129-150, 1997.
1983. 100. Mintz L, Drew WL, Hoo R, et al: Age-dependent resistance of human
73. Gaytant MA, Steegers EA, van Cromvoirt PL, et al: Incidence of alveolar macrophages to herpes simplex virus, Infect Immun 28:
herpes neonatorum in Netherlands, Ned Tijdschr Geneeskd 144: 417-420, 1980.
1832-1836, 2000. 101. Hirsch MS, Zisman B, Allison AC: Macrophages and age-dependent
74. Caviness AC, Demmler GJ, Almendarez Y, et al: The prevalence resistance to Herpes simplex virus in mice, J Immunol 104:1160-1165,
of neonatal herpes simplex virus infection compared with serious 1970.
bacterial illness in hospitalized neonates, J Pediatr 153:164-169, 102. Diamond C, Mohan K, Hobson A, et al: Viremia in neonatal herpes
2008. simplex virus infections, Pediatr Infect Dis J 18:487-489, 1999.
75. Baldwin S, Whitley RJ: Intrauterine herpes simplex virus infection, 103. Kawada J, Kimura H, Ito Y, et al: Evaluation of systemic inflam-
Teratology 39:1-10, 1989. matory responses in neonates with herpes simplex virus infection,
76. Florman AL, Gershon AA, Blackett PR, et al: Intrauterine infection J Infect Dis 190:494-498, 2004.
with herpes simplex virus. Resultant congenital malformations, 104. Singer DB: Pathology of neonatal herpes simplex virus infection, Per-
JAMA 225:129-132, 1973. spect Pediatr Pathol 6:243-278, 1981.
77. Hutto C, Arvin A, Jacobs R, et al: Intrauterine herpes simplex virus 105. Kimberlin DW, Whitley RJ, Wan W, et al: Oral acyclovir suppres-
infections, J Pediatr 110:97-101, 1987. sion and neurodevelopment after neonatal herpes, N Engl J Med
78. South MA, Tompkins WA, Morris CR, et al: Congenital malformation 365:1284-1292, 2011.
of the central nervous system associated with genital type (type 2) 106. Whitley R, Arvin A, Prober C, et al: Predictors of morbidity and mor-
herpesvirus, J Pediatr 75:13-18, 1969. tality in neonates with herpes simplex virus infections, N Engl J Med
79. Kahlon J, Whitley RJ: Antibody response of the newborn after herpes 324:450-454, 1991.
simplex virus infection, J Infect Dis 158:925-933, 1988. 107. Nahmias AJ, Alford CA, Korones SB: Infection of the newborn with
80. Karesh JW, Kapur S, MacDonald M: Herpes simplex virus and con- herpesvirus hominis, Adv Pediatr 17:185-226, 1970.
genital malformations, South Med J 76:1561-1563, 1983. 108. Kimberlin DW, Lin CY, Jacobs RF, et al: Safety and efficacy of high-
81. Monif GR, Kellner KR, Donnelly WH Jr: Congenital herpes simplex dose intravenous acyclovir in the management of neonatal herpes
type II infection, Am J Obstet Gynecol 152:1000-1002, 1985. simplex virus infections, Pediatrics 108:230-238, 2001.
82. Garcia AG: Maternal herpes-simplex infection causing abortion. His- 109. Whitley R, Arvin A, Prober C, et al: A controlled trial comparing
topathologic study of the placenta, Hospital (Rio J) 78:1267-1274, vidarabine with acyclovir in neonatal herpes simplex virus infection,
1970. N Engl J Med 324:444-449, 1991.
27 • Herpes Simplex Virus Infections 865

110. Whitley RJ, Nahmias AJ, Soong SJ, et al: Vidarabine therapy of neo- 138. Weil AA, Glaser CA, Amad Z, et al: Patients with suspected herpes
natal herpes simplex virus infection, Pediatrics 66:495-501, 1980. simplex encephalitis: rethinking an initial negative polymerase
111. Kimberlin DW: Neonatal herpes simplex infection, Clin Microbiol Rev chain reaction result, Clin Infect Dis 34:1154-1157, 2002.
17:1-13, 2004. 139. Barbi M, Binda S, Primache V, et al: Use of Guthrie cards for the early
112. Anderson MS, Abzug MJ: Hydrops fetalis: an unusual presentation diagnosis of neonatal herpes simplex virus disease, Pediatr Infect Dis J
of intrauterine herpes simplex virus infection, Pediatr Infect Dis J 17:251-252, 1998.
18:837-839, 1999. 140. Lewensohn-Fuchs I, Osterwall P, Forsgren M, et al: Detection of her-
113. Malm G, Berg U, Forsgren M: Neonatal herpes simplex: clinical find- pes simplex virus DNA in dried blood spots making a retrospective
ings and outcome in relation to type of maternal infection, Acta Pae- diagnosis possible, J Clin Virol 26:39-48, 2003.
diatr 84:256-260, 1995. 141. American Academy of Pediatrics: Herpes simplex. In Pickering LK,
114. Arvin AM, Yeager AS, Bruhn FW, et al: Neonatal herpes sim- Baker CJ, Kimberlin DW, Long SS, editors: Red book: 2012 report
plex infection in the absence of mucocutaneous lesions, J Pediatr of the Committee on Infectious Diseases, ed 29, Elk Grove Village, Ill,
100:715-721, 1982. 2012, American Academy of Pediatrics, pp 398-408.
115. Greenes DS, Rowitch D, Thorne GM, et al: Neonatal herpes simplex 142. Elion GB, Furman PA, Fyfe JA, et al: Selectivity of action of an anti-
virus infection presenting as fulminant liver failure, Pediatr Infect Dis J herpetic agent, 9-(2-hydroxyethoxymethyl) guanine, Proc Natl Acad
14:242-244, 1995. Sci U S A 74:5716-5720, 1977.
116. Lee WS, Kelly DA, Tanner MS, et al: Neonatal liver transplantation 143. Whitley RJ, Yeager A, Kartus P, et al: Neonatal herpes simplex virus
for fulminant hepatitis caused by herpes simplex virus type 2, J Pedi- infection: follow-up evaluation of vidarabine therapy, Pediatrics
atr Gastroenterol Nutr 35:220-223, 2002. 72:778-785, 1983.
117. Ford A, Swing DC Jr, Tobin JR, et al: A 10-day-old neonate with ful- 144. Englund JA, Fletcher CV, Balfour HH Jr: Acyclovir therapy in neo-
minant hepatitis, J Paediatr Child Health 44:471-472, 2008. nates, J Pediatr 119:129-135, 1991.
118. Golden SE: Neonatal herpes simplex viremia, Pediatr Infect Dis J 145. American Academy of Pediatrics: Herpes simplex. In Pickering LK,
7:425-427, 1988. editor: Red book: 2000 report of the Committee on Infectious Diseases, ed
119. Gressens P, Martin JR: HSV-2 DNA persistence in astrocytes of the 25, Elk Grove Village, Ill, 2000, American Academy of Pediatrics, pp
trigeminal root entry zone: double labeling by in situ PCR and immu- 309-318.
nohistochemistry, J Neuropathol Exp Neurol 53:127-135, 1994. 146. Long SS: In defense of empiric acyclovir therapy in certain neonates,
120. Langlet C, Gaugler C, Castaing M, et al: An uncommon case of dissem- J Pediatr 153:157-158, 2008.
inated neonatal herpes simplex infection presenting with pneumonia 147. Kimberlin DW: When should you initiate acyclovir therapy in a
and pleural effusions, Eur J Pediatr 162:532-533, 2003. neonate? J Pediatr 153:155-156, 2008.
121. O’Riordan DP, Golden WC, Aucott SW: Herpes simplex virus infec- 148. Nyquist AC, Rotbart HA, Cotton M, et al: Acyclovir-resistant neo-
tions in preterm infants, Pediatrics 118:e1612-e1620, 2006. natal herpes simplex virus infection of the larynx, J Pediatr 124:
122. Whitley RJ, Kimberlin DW: Treatment of viral infections during preg- 967-971, 1994.
nancy and the neonatal period, Clin Perinatol 24:267-283, 1997. 149. Oram RJ, Marcellino D, Strauss D, et al: Characterization of an
123. Kimberlin DW: Herpes simplex virus infections of the central nervous acyclovir-resistant herpes simplex virus type 2 strain isolated from
system, Semin Pediatr Infect Dis 14:83-89, 2003. a premature neonate, J Infect Dis 181:1458-1461, 2000.
124. Mizrahi EM, Tharp BR: A characteristic EEG pattern in neonatal herpes 150. Levin MJ, Weinberg A, Leary JJ, et al: Development of acyclovir-
simplex encephalitis, Neurology 32:1215-1220, 1982. resistant herpes simplex virus early during the treatment of herpes
125. O’Reilly MA, O’Reilly PM, de Bruyn R: Neonatal herpes simplex type 2 neonatorum, Pediatr Infect Dis J 20:1094-1097, 2001.
encephalitis: its appearances on ultrasound and CT, Pediatr Radiol 151. Kimberlin D, Powell D, Gruber W, et al: Administration of oral
25:68-69, 1995. acyclovir suppressive therapy after neonatal herpes simplex virus
126. Toth C, Harder S, Yager J: Neonatal herpes encephalitis: a case disease limited to the skin, eyes and mouth: results of a phase I/II
series and review of clinical presentation, Can J Neurol Sci 30:36-40, trial, Pediatr Infect Dis J 15:247-254, 1996.
2003. 152. Rabalais GP, Nusinoff-Lehrman S, Arvin AM, et al: Antiviral suscep-
127. Pelligra G, Lynch N, Miller SP, et al: Brainstem involvement in tibilities of herpes simplex virus isolates from infants with recurrent
neonatal herpes simplex virus type 2 encephalitis, Pediatrics 120: mucocutaneous lesions after neonatal infection, Pediatr Infect Dis J
e442-e446, 2007. 8:221-223, 1989.
128. Kimura H, Ito Y, Futamura M, et al: Quantitation of viral load in neo- 153. Tiffany KF, Benjamin DK Jr, Palasanthiran P, et al: Improved
natal herpes simplex virus infection and comparison between type 1 neurodevelopmental outcomes following long-term high-dose
and type 2, J Med Virol 67:349-353, 2002. oral acyclovir therapy in infants with central nervous system
129. Gutman LT, Wilfert CM, Eppes S: Herpes simplex virus encephalitis and disseminated herpes simplex disease, J Perinatol 25:156-
in children: analysis of cerebrospinal fluid and progressive neurode- 161, 2005.
velopmental deterioration, J Infect Dis 154:415-421, 1986. 154. Kohl S, Loo LS, Rench MA, et al: Effect of intravenously administered
130. Music SI, Fine EM, Togo Y: Zoster-like disease in the newborn due to immune globulin on functional antibody to herpes simplex virus in
herpes-simplex virus, N Engl J Med 284:24-26, 1971. low birth weight neonates, J Pediatr 115:135-139, 1989.
131. Nahmias AJ, Hagler WS: Ocular manifestation of herpes simplex 155. Gutierrez KM, Falkovitz Halpern MS, Maldonado Y, et al: The epi-
in the newborn (neonatal ocular herpes), Int Ophthalmol Clin 12: demiology of neonatal herpes simplex virus infections in California
191-213, 1972. from 1985 to 1995, J Infect Dis 180:199-202, 1999.
132. Kimberlin DW, Lakeman FD, Arvin AM, et al: Application of the poly- 156. Cone RW, Hobson AC, Brown Z, et al: Frequent detection of geni-
merase chain reaction to the diagnosis and management of neonatal tal herpes simplex virus DNA by polymerase chain reaction among
herpes simplex virus disease, J Infect Dis 174:1162-1167, 1996. pregnant women, JAMA 272:792-796, 1994.
133. Cherry JD, Soriano F, Jahn CL: Search for perinatal viral infection. 157. Hardy DA, Arvin AM, Yasukawa LL, et al: Use of polymerase chain
A prospective, clinical, virologic, and serologic study, Am J Dis Child reaction for successful identification of asymptomatic genital infec-
116:245-250, 1968. tion with herpes simplex virus in pregnant women at delivery,
134. Whitley RJ, Lakeman F: Herpes simplex virus infections of the cen- J Infect Dis 162:1031-1035, 1990.
tral nervous system: therapeutic and diagnostic considerations, Clin 158. Watts DH, Brown ZA, Money D, et al: A double-blind, randomized,
Infect Dis 20:414-420, 1995. placebo-controlled trial of acyclovir in late pregnancy for the reduc-
135. Kimura H, Futamura M, Kito H, et al: Detection of viral DNA in neo- tion of herpes simplex virus shedding and cesarean delivery, Am J
natal herpes simplex virus infections: frequent and prolonged pres- Obstet Gynecol 188:836-843, 2003.
ence in serum and cerebrospinal fluid, J Infect Dis 164:289-293, 159. Andrews WW, Kimberlin DF, Whitley R, et al: Valacyclovir therapy
1991. to reduce recurrent genital herpes in pregnant women, Am J Obstet
136. Troendle-Atkins J, Demmler GJ, Buffone GJ: Rapid diagnosis of Gynecol 194:774-781, 2006.
herpes simplex virus encephalitis by using the polymerase chain 160. Pinninti SG, Angara R, Feja KN, et al: Neonatal herpes disease
reaction, J Pediatr 123:376-380, 1993. following maternal antenatal antiviral suppressive therapy: a multi-
137. Malm G, Forsgren M: Neonatal herpes simplex virus infections: HSV center case series, J Pediatr 161:134-138.e1-e3, 2012.
DNA in cerebrospinal fluid and serum, Arch Dis Child Fetal Neonatal 161. Tita AT, Grobman WA, Rouse DJ: Antenatal herpes serologic screening:
Ed 81:F24-F29, 1999. an appraisal of the evidence, Obstet Gynecol 108:1247-1253, 2006.
28 Human Parvovirus
STUART P. ADLER and WILLIAM C. KOCH

CHAPTER OUTLINE
ERRNVPHGLFRVRUJ
Microbiology Overview
General Aspects of Pathogenesis Fetal Death
Epidemiology and Transmission Asymptomatic Fetal Infection
Overview Birth Defects
Global Distribution Other Fetal Manifestations
Seasonality and Periodicity Fetal Hydrops
Seroprevalence by Age Fetal Outcome in Relation to Maternal
Seroprevalence by Gender Manifestations
Seroprevalence by Race Long-term Outcomes
Incidence Pathogenesis of Infection in the Fetus
Risk Factors for Acquisition Fetal Immune Responses to Parvovirus B19
Hospital Transmission Pathogenesis of Parvovirus B19 Hydrops
Routes of Viral Spread Pathology in the Fetus
Risk of Parvovirus B19 Acquisition for Anatomic and Histologic Features
Women of Childbearing Age Placenta
Clinical Manifestations (Other Than Intra- Heart
uterine Infection) Other Organs
Erythema Infectiosum Diagnostic Evaluation and Manage-
Transient Aplastic Crisis ment of the Woman and Fetus Exposed
Arthropathy to or Infected by Parvovirus B19 During
Infection in Immunocompromised Hosts Pregnancy
Other Dermatologic Syndromes Overview
Central Nervous System Infection and Prevalence of Erythema Infectiosum
­Neurologic Disorders History of Exposure
Renal Disease Clinical Features Suggesting Signs and
Myocardial Disease Symptoms of Parvovirus B19 Infection
in the Pregnant Woman
General Aspects of Diagnosis
Laboratory Diagnosis in the
Laboratory Diagnostic Methods Pregnant Woman
Epidemiology of Parvovirus B19 Fetal Monitoring
­Infections and Risk of Acquisition
in Pregnant Women Fetal Therapy
Prevalence and Incidence in the Differential Diagnosis
United States Prognosis
Prevalence and Incidence in Other Countries Prevention
Clinical Manifestations of Parvovirus B19 General Measures
Infections in Pregnant Women Vaccine Development
Intrauterine Transmission Rates, Clinical
Manifestations, and Fetal Outcomes

The parvoviruses are a family of single-stranded DNA to as parvovirus B19 or simply B19. B19 is the prototype
viruses that have a wide cellular tropism and broad host of the genus Erythrovirus in the Parvoviridae family, and
range, causing infection in invertebrate species and verte- a new genus and name have been proposed for this virus,
brates, from insects to mammals. Although many parvovi- Erythrovirus B19,3 based on its cellular tropism for ery-
ruses are important veterinary pathogens, there are only throid lineage cells and to distinguish it from the other
two human pathogens in the family: human parvovirus mammalian parvoviruses. Compared with most other com-
B19 and the more recently described human bocavirus.1,2 mon human viruses, B19 is a relatively new pathogen, but
Human bocavirus seems to be primarily a respiratory since its initial description, B19 has come to be associated
pathogen of young children and is not discussed further with a variety of seemingly diverse clinical syndromes in
here. Human parvovirus B19 is most commonly referred many different patient populations (Table 28-1). Although
866

Você também pode gostar