Vous êtes sur la page 1sur 9

Seminars in Fetal & Neonatal Medicine 14 (2009) 209–217

Contents lists available at ScienceDirect

Seminars in Fetal & Neonatal Medicine


journal homepage: www.elsevier.com/locate/siny

Varicella in the fetus and newborn


Candice K. Smith*, Ann M. Arvin
Stanford University School of Medicine, 300 Pasteur Drive, G322, Stanford, CA 94305, USA

s u m m a r y

Keywords: Varicella (chickenpox) in pregnancy is unusual because most women of childbearing age are immune. It
Congenital varicella syndrome can, however, cause significant morbidity for the pregnant woman and in rare cases cause congenital
Diagnosis varicella syndrome. The incidence of congenital varicella syndrome after maternal varicella during the
Fetal infection
first two trimesters is <1% across multiple cohort studies. Maternal infection in the third trimester is not
Herpes zoster
associated with congenital varicella syndrome, but the infant may develop herpes zoster during the first
Pregnancy
Vaccination one or two years. Maternal infection just before or after delivery presents a high risk for disseminated
Varicella varicella in the infant. Serious infection can be prevented with passive antibody prophylaxis and antiviral
therapy. Maternal herpes zoster does not result in adverse fetal or neonatal outcomes.
Ó 2008 Elsevier Ltd. All rights reserved.

1. Introduction the widespread annual varicella epidemics, w90% of adults born in


the USA and Europe are immune to VZV. The introduction of
Varicella zoster virus (VZV) is a human alphaherpesvirus found universal varicella vaccination in the USA has modified the pattern
throughout the world. As is characteristic of the alphaherpesvi- of VZV transmission since 1995, but before then, the annual inci-
ruses, VZV establishes latency in the cells of the sensory nerve dence of varicella was roughly equal to the birth cohort. Morbidity
ganglia after primary infection. It causes varicella (chickenpox) as and mortality rates are higher in adults than in children, and
the primary infection and herpes zoster when it reactivates. The pregnant women in particular are at risk, specifically for varicella
clinical disease, which is usually self-limited, is a febrile illness with pneumonia.4
a pruritic vesicular rash. However, primary VZV infection can have Although VZV infections occur in a worldwide distribution,
severe consequences when it is acquired in pregnancy or the there are notable variations in the incidence by latitude and
neonatal period. climate. In the absence of varicella vaccination programs, varicella
VZV is highly infectious.1 Most adults in the USA are immune to outbreaks follow a seasonal pattern of increasing numbers of cases
varicella secondary to the high prevalence of transmission during in winter and spring in temperate climates. Through these
childhood years.2 In addition, the varicella vaccine was licensed in epidemics, >90% of children become infected by age 15 years. By
1995 and has significantly reduced the incidence of primary VZV contrast, in tropical climates, VZV is acquired at less frequency and
infection in the population as a whole.3 For these reasons, varicella at older ages. These geographical differences in epidemic patterns
affecting women during pregnancy or neonates is rare. Pregnant are not understood, but may be secondary to viral strain
women who contract varicella, particularly in the third trimester, are differences, environmental conditions that limit survival of this
at higher risk for more severe disease, including pneumonia. The fetal heat-labile virus in the tropics, or host genetic factors.
outcomes include normal healthy infants in almost all cases, possible Just before introduction of varicella vaccine in the USA, varicella
zoster in infancy, and, rarely, fetal death or congenital varicella had shifted from a peak incidence in children aged 5–9 years to 1–4
syndrome (CVS). Perinatally acquired VZV can also cause significant years. This shift was attributed primarily to earlier exposure
morbidity and mortality in the newborn. Maternal herpes zoster is through daycare attendance.5 VZV vaccine was licensed in 1995 and
benign for both the pregnant woman and the fetus or newborn. decreased the incidence of disease by 85–90% in the decade
following licensure. In the USA, the national varicella vaccination
2. VZV epidemiology program follows VZV with active surveillance, documenting
a vaccine coverage rate of 80% within surveillance areas.6 Varicella
VZV is a readily transmissible virus, with w90% of susceptible incidence decreased most significantly in children aged 1–4 years,
household contacts becoming infected after exposure. Because of but decreases were shown in all age groups, including adults.
Among adults, incidence of varicella declined 74% during
* Corresponding author. Tel.: þ1 650 723 5682; fax: þ1 650 725 8040. 1995–2005, despite vaccination rates of only 3%. Herd immunity is
E-mail address: candices@stanford.edu (C.K. Smith). the likely explanation for this decrease among adults. 7

1744-165X/$ – see front matter Ó 2008 Elsevier Ltd. All rights reserved.
doi:10.1016/j.siny.2008.11.008
210 C.K. Smith, A.M. Arvin / Seminars in Fetal & Neonatal Medicine 14 (2009) 209–217

Thus, the majority of adults who have lived since childhood in series of 347 pregnant women with varicella, 18 (5.2%) had pneu-
temperate climates are immune, as evidenced by the detection of monia, all were treated with acyclovir, and none died.4 The
VZV IgG antibodies in nearly 95% of adults in the USA.8,9 Therefore, reduction in mortality compared with historical rates of 20–45%
only w5% of women of childbearing age remain susceptible to reflects both the availability of antiviral therapy and better respi-
VZV.10 The incidence of primary VZV infection during pregnancy is ratory management.16
correspondingly low. The rate was seven per 1000 in an early study When primary VZV infection occurs in pregnancy, the virus can
of 30 000 pregnancies followed prospectively during the period be transferred across the placenta to the developing fetus as
1958–1964.11 Introduction of universal childhood immunization a consequence of viremia. While transplacental transmission is
against varicella has the potential to reduce the occurrence of assumed to be the most likely route, infection of the genital tract
varicella in pregnant women through herd immunity. Conversely, mucosa leading to an ascending infection with transfer across the
varicella vaccination programs must be comprehensive because of amniotic membranes cannot be excluded. While viral transfer
the risk that women will reach childbearing age without having occurs and is evidenced by detection of VZV DNA in amniotic fluid,
either natural or vaccine-induced immunity. the rates of transplacental transmission do not correlate directly
with fetal infection. The amniotic fluid may be positive for VZV by
3. Pathogenesis and clinical manifestations of primary VZV polymerase chain reaction (PCR) testing for viral DNA without
infection causing fetal disease.17 Spontaneous abortion, fetal demise, and
premature delivery have been reported after varicella in pregnancy,
Primary infection with VZV begins with mucosal inoculation of but the risk of these complications is low.18
the virus through respiratory exposure or direct contact with fluid
from varicella or herpes zoster skin lesions. The incubation period 4. Intrauterine VZV infection
usually is 14–16 days but can vary from as few as 10 or as many as
21 days after contact. Based upon a poxvirus model, VZV had been Primary VZV infection in the pregnant woman most often
presumed to replicate first in the regional lymphoid tissues, fol- results in the birth of a normal newborn either because the virus
lowed by primary viremia and viral replication in the liver and was not transmitted to the fetus or fetal infection was controlled
spleen; a secondary viremia was proposed to occur just before the without consequences. Some of these infants may develop herpes
appearance of skin or mucosal lesions in which infected mono- zoster in infancy. However, the majority of infants with intrauterine
nuclear cells transported the virus to these sites.12 Recently, infection evidenced by persistence of IgG at one to two years of age
however, studies in the SCID-hu mouse model of VZV pathogenesis escape without signs or symptoms.14 Rarely, the outcome is
have shown that infected T-lymphocytes transport the virus to the congenital varicella syndrome (CVS) and/or fetal death. VZV may be
skin xenografts within 24 h. A robust epidermal innate antiviral transmitted across the placenta and cause intrauterine infection at
response with interferon-a contributes to the delay in eruption of any time during gestation. However, the spectrum of clinical
varicella skin lesions.13 From these studies, one may postulate that disease that occurs correlates with the gestational age of the fetus.
the 10–21 day incubation period is the interval required for VZV to The highest risk for spontaneous abortion or CVS is in the first 20
overcome this innate immune response and create visible lesions at weeks of gestation, but the overall risk of CVS in the first 20 weeks
skin or mucosal surfaces. During the incubation period, the virus is <1%. There are a few case reports of infants with CVS whose
may spread as uninfected T-lymphocytes traffic into early stage mothers had varicella at 21–28 weeks’ gestation.16,19 If VZV trans-
lesions and transport progeny virus to new mucocutaneous sites; mission occurs as a consequence of maternal infection during the
replication in reticuloendothelial tissue may also amplify the cell- perinatal period, from approximately two days before delivery to
associated viremia. By late incubation (24–48 h before the five days after, the infant is at risk of severe life-threatening vari-
appearance of the rash), infectious VZV may be present in respi- cella disease.
ratory secretions, since respiratory transmission to susceptible
contacts has been observed after exposures during this period. 4.1. Congenital varicella syndrome
Approximately half of individuals with primary VZV infection,
more commonly older children and adults, experience a prodrome CVS was first described in 1947, and >100 cases have been
of fever, malaise, headache, and abdominal pain. The rash is initially reported subsequently.16,20 A large prospective study of 1373
manifested on the scalp, face, or trunk and consists of pruritic pregnancies complicated by maternal varicella performed in
erythematous macules that evolve to small fluid-filled vesicles. The Germany and the UK confirmed several earlier studies with fewer
vesicular fluid of skin lesions contains high concentrations of such pregnancies that had suggested a low incidence of CVS. Nine
infectious virions, and the virus may be present in respiratory cases of CVS were documented. All cases occurred during the first
droplets or secretions. The period of contagiousness is one to two 20 weeks of gestation. The highest risk (2.0%) was observed
days before the onset of rash until all the lesions are crusted, usually between 13 and 20 weeks of gestation, with seven affected infants
five to seven days after the first lesions appear. Subclinical disease is identified among 351 pregnancies. Only two cases of CVS were
rare – a difference between VZV and other herpes viruses that are identified among 472 pregnancies in which maternal varicella
typically asymptomatic during primary infection. Varicella is occurred before 13 weeks (0.4%).21 Overall the incidence of CVS in
generally short in duration and mild in healthy hosts. The initial nine reported cohort studies was 0.55% in the first trimester, 1.4% in
phase of illness may follow this apparently uncomplicated course the second trimester, and 0% in the third trimester.16
in pregnant women and neonates. However, instead of resolving, it The defects of CVS involve the skin, the limbs, the eyes, and the
sometimes progresses rapidly to life-threatening disseminated central and autonomic nervous systems (Table 1).22,23 The charac-
infection involving the lungs, liver and central nervous system. teristic cicatricial skin lesions may be depressed and pigmented in
The clinical course of varicella in adults is more likely to be a dermatomal distribution or hypopigmented (Fig. 1). Ocular
complicated, and this higher incidence of serious varicella in adults defects include cataracts, chorioretinitis, Horner syndrome, ptosis,
is magnified in pregnant women. Varicella pneumonia, which may microphthalmia and nystagmus. The typical limb and joint abnor-
progress to respiratory failure, constitutes the source of most malities are hypoplasia of the bones and muscle and/or absent or
morbidity and mortality in pregnant women, as it does in non- malformed digits (Figs. 2 and 3). Central nervous system abnor-
pregnant adults.14,15 The onset of varicella pneumonia is usually malities in CVS patients include cortical atrophy, seizures, and
three to five days after the appearance of the rash. In a recent case mental retardation. Many affected infants have microcephaly.
C.K. Smith, A.M. Arvin / Seminars in Fetal & Neonatal Medicine 14 (2009) 209–217 211

Table 1 supports the hypothesis of early viremia and with dissemination


Clinical manifestations of congenital varicella syndrome. caused by a poor host immune response.24,25 Microcephaly can be
Skin attributed to VZV encephalitis and irreversible damage to growth of
Cicatricial lesions the developing brain. Of interest, the virus does not appear to cause
Cutaneous defects intrauterine damage to the lungs or liver in infants with CVS, as it
Hypopigmentation
Nervous system
can in perinatal varicella or in other immunocompromised hosts.
Intrauterine encephalitis Fulminant infection involving these organs may result in fetal
Cortical atrophy/porencephaly demise, rather than birth of an infant with CVS. VZV is also
Seizures a neurotropic virus and many of the defects have been postulated to
Mental retardation
be a direct result of spinal cord and ganglia infection, causing
Autonomic instability
Eye destruction of the plexi during embryogenesis and leading to
Chorioretinitis denervation of the limb bud and subsequent hypoplasia. Failure of
Cataracts muscle development has consequences for limb bone formation.
Micropthalmia The cutaneous defects are also likely to reflect VZV infection of
Anisocoria
Musculoskeletal
sensory nerves. VZV infection of cells in developing optic tracts also
Limb hypoplasia explains the optic atrophy and chorioretinitis.26 In addition, VZV
Muscle hypoplasia appears to destroy neural cells that comprise the autonomic
Systemic nervous system in the affected fetus. Some skin lesions, which have
Intrauterine growth retardation
the dermatomal distribution associated with herpes zoster, may be
Developmental delay
Cardiovascular defects caused by VZV reactivation in utero. The short latency phase, if
Gastrointestinal tract latency is established at all, can be explained by an inadequate cell-
Gastrointestinal reflux mediated immune response in the fetus.27
Urinary tract Overall, the prognosis of infants born with CVS is poor, with
Hydroureter
Hydronephrosis
death in infancy resulting from intractable gastroesophageal reflux,
severe recurrent aspiration pneumonia and respiratory failure.
However, there are some reports of more favorable outcomes.28–30
Cohort studies show a higher rate of females with CVS, 66–85%.16
Dysfunction of the autonomic nervous system is evidenced by The cause for these higher rates is unknown, but one study
neurogenic bladder, hydroureter, esophageal dilation and reflux hypothesized that it is secondary to higher rates of male fetal
leading to aspiration pneumonia.24 death.31
The pathogenesis of CVS may reflect disseminated infection in
utero or consequences of failure of virus–host interaction to result
in establishment of latency, as normally occurs in postnatal VZV 4.2. Herpes zoster in infancy
infection. Since VZV is a lymphotropic virus, it has the potential to
spread to all fetal organs by the hematogenous route. A few reports Maternal varicella during pregnancy can manifest as infant zoster
that have examined fetuses infected after maternal varicella have in the first or second year.32–34 (Fig. 4). In one report, out of ten children
shown that VZV is distributed throughout fetal tissue which who were asymptomatic at birth, but later presented with zoster, eight

Fig. 1. Classic cicatricial scarring is visualized in an infant with congenital varicella syndrome.
212 C.K. Smith, A.M. Arvin / Seminars in Fetal & Neonatal Medicine 14 (2009) 209–217

Fig. 2. Infant with congenital varicella syndrome. The right lower leg has significant bone and muscle hypoplasia. The classic cicatricial scarring is also visualized.

did so in the first year of life. The gestational age at the time of maternal zoster infection in infancy was 3.8% in cases when maternal amniotic
varicella ranged from 14 to 33 weeks, with a median age of 25 weeks. fluid had been positive for VZV DNA by PCR.17
The observed risk of zoster when maternal infection occurs before 24
weeks was 0.8%, compared to a 1.7% observed risk if maternal varicella
was after 25 weeks.21 In another report, the risk of postnatal herpes 5. Diagnosis of varicella and fetal complications in pregnancy

The diagnosis of varicella in pregnant women can often be made


from the classic clinical presentation of a vesicular pruritic rash. In
addition, there are often preceding or other secondary cases of
varicella within the household. A case series of pregnant women
with varicella identified 90% of index cases as one of the woman’s
children.17 However, as the incidence of varicella decreases in
countries with high vaccination rates, younger physicians will have
less opportunity to see the characteristic skin lesions and may feel
less confident with the clinical diagnosis. If the diagnosis is in
question, laboratory testing can be performed. The base of a vesic-
ular skin lesion can be scraped for culture, immunohistochemical
staining, and/or PCR. VZV culture is highly specific, but takes seven
to 10 days and is insensitive. Direct fluorescent antigen staining
using monoclonal antibodies to detect VZV glycoproteins in cells
from a skin lesion scraping can be performed within hours and has
high sensitivity and specificity. If a qualified laboratory is available,
rapid testing can also be done by VZV PCR. Serologic tests are of
little or no value for the diagnosis of varicella as VZV IgG antibodies
often only become detectable a few days after the onset of the
illness, VZV IgG enzyme-linked immunosorbent assay (ELISA)
methods have substantial false negative rates (15–20%), and
seroconversion is a retrospective approach to diagnosis. Further, no
validated serologic tests for VZV IgM are available to clinicians and
VZV IgM antibodies can be detected in individuals who have had
varicella at some time in the past.35,36 When standard ELISA
methods are used to detect VZV IgM antibodies, the rate of false
positives is high.
Fig. 3. X-ray images of the lower extremities of the above infant, noting the bone Evaluating the fetus for intrauterine infection after maternal
hypoplasia. varicella presents several challenges and developing validated
C.K. Smith, A.M. Arvin / Seminars in Fetal & Neonatal Medicine 14 (2009) 209–217 213

Fig. 4. Infant aged 14 months with herpes zoster in the classic dermatomal pattern. There was a maternal history of varicella at 28 weeks of pregnancy.

approaches is difficult because the incidence of both varicella in amniotic fluid with regard to risk of VZV transmission to fetus or
pregnancy and any associated fetal damage is fortunately quite low. risk for fetal defects.
Several studies have sought correlations between the detection of Serial evaluations of fetal development by prenatal ultrasound
VZV IgM in fetal blood or the PCR detection of VZV DNA in fetal may be useful to identify severe manifestations of intrauterine VZV
blood or amniotic fluid postnatal evidence of fetal infection. VZV infection.37 Ultrasound is most definitive when the fetus has
IgM was positive in only 25% of infants born with clinical mani- defects such as limb atrophy or evidence of microcephaly, but
festations of disease.21 A study of 107 women who had varicella subtle findings such as liver calcifications on prenatal ultrasound
before the 24th week of pregnancy showed that negative PCR were the only sign of damage in an infant who was severely
results in amniotic fluid correlated with favorable fetal outcome – affected.38 The role of prenatal magnetic resonance imaging for
good health, with normal psychomotor development.17 However, it assessment of the fetus after maternal varicella is only beginning to
is necessary to note that this sample size was not powered to be delineated, but it may provide improved specificity, particularly
evaluate for risk of fetal pathology when the risk of such sequelae is for central nervous system damage.39
<1%. Additionally, VZV PCR has a poor positive predictive value for
fetal disease or disease severity. One limitation of testing amniotic 6. Antiviral treatment of varicella in pregnancy and
fluid for VZV by PCR is timing; an amniocentesis can only be per- intrauterine infection
formed after 16–18 weeks of gestation, so in some clinical situa-
tions the PCR could be obtained well after initial presentation of Acyclovir is an analogue of guanosine that inhibits viral repli-
varicella in the mother. The risk of fetal demise from amniocentesis, cation and is highly specific for cells infected with herpes simplex
which is low, must also be weighed against the low risk of VZV- virus and VZV. Acyclovir is activated by the virus-encoded thymi-
related fetal damage. Importantly, VZV DNA in amniotic fluid does dine kinase. Acyclovir crosses the placenta and is excreted by the
not indicate whether the fetus has been infected or, if infected, fetal kidney. Although it is concentrated in amniotic fluid, it does
whether fetal damage will occur. In the study of 107 pregnant not appear to accumulate in the fetus.40 Valacyclovir is an orally
women with varicella, nine amniotic fluid specimens were VZV PCR administered prodrug of acyclovir that has improved oral
positive (8.4%); of these, the pregnancy outcomes were five full- bioavailability and improved pharmacokinetic properties. Acyclovir
term normal infants, one infant with bilateral microphthalmia, two and valacyclovir are pregnancy category B and are not approved for
therapeutic abortions, of which one fetus showed limb hypoplasia, any indication in pregnant women. However, a registry of neonates
and one spontaneous abortion. Thus only two pregnancies were exposed to acyclovir during the first trimester showed that no
associated with signs of intrauterine varicella. Since varicella is teratogenic effects could be attributed to acyclovir.41 Therefore, in
a systemic infection, it should be assumed that the pregnant instances of serious disease or complication such as pneumonia in
woman with varicella may have viral genomic DNA in many bodily the mother, the Committee on Infectious Diseases of the American
fluids, including amniotic fluid, and the duration of persistence of Academy of Pediatrics concluded that the benefit of treating with
VZV genomes, and whether it represents the presence of infectious acyclovir in these cases outweighs risk.42 Valacylovir achieves
virus is not known. The rate of detection of VZV DNA by PCR might higher serum levels than oral acyclovir; there are no data regarding
be higher when the mother’s infection is more recent, but would the risk to the fetus, but the risk can be considered low, based on
not imply a higher risk of fetal sequelae. At this point, no conclu- outcomes when pregnant women have been given intravenous
sions can be drawn about the significance of a positive VZV PCR of acyclovir.
214 C.K. Smith, A.M. Arvin / Seminars in Fetal & Neonatal Medicine 14 (2009) 209–217

Uncomplicated maternal varicella can be treated orally with temperate climates. Because of the difference in epidemiology,
acyclovir or valacyclovir and treatment should be considered, concern about VZV susceptibility should be highest if the pregnant
particularly later in gestation when risks of maternal complication woman who has no varicella history is from a geographical area
from disease are increased and risks to the developing fetus can be where epidemic varicella is less common. Cost-benefit analysis
considered minimal. This approach addresses the problem that VZV supports testing for serum VZV IgG titers if the mother with no
dissemination can occur very rapidly in a patient whose clinical history is from a high prevalence area because prophylaxis is likely
course seems benign. The decision to treat must be based on clinical to be necessary in <5% of cases. In practice, this approach requires
judgment despite the lack of an approved indication because the access to a laboratory that can report the results on an urgent basis
low frequency of maternal varicella means that sufficient data to so that prophylaxis can be given within the 72–96 h, if required.
establish risks and benefits cannot be obtained. Intravenous anti- Since VZV IgG assays have a significant false-negative rate, some
viral therapy is indicated for the pregnant woman who develops women who are identified as susceptible will be immune to VZV.
any signs of pneumonia including cough, dyspnea, hypoxia, or an However, adverse reactions to prophylaxis are unusual in immune
abnormal chest X-ray.43 There is no information about whether or susceptible individuals.
giving acyclovir or valacyclovir to pregnant women with varicella High titer varicella zoster immune globulin (VZIG) prepared
reduces the already low risk for CVS.4 from individuals with recent zoster or from donors screened for
Because the manifestations of CVS result from intrauterine high VZV IgG titers (VariZIG) is preferred because of the intra-
damage to the developing fetus, antiviral therapy in the newborn muscular route of administration; intravenous immune globulin
period cannot be expected to have substantial impact on the (IVIG) can be substituted if necessary at a dose of 400 mg/kg. The
sequelae. However, a case-by-case evaluation of infants with CVS is VariZIG can be obtained 24 h a day from the sole authorized US
necessary in making decisions about when or whether to treat with distributor (FFF Enterprises, Temecula, CA, USA; tel. 1-800-843-
acyclovir. There is a paucity of evidence regarding the role of 7477 or online at http://www.fffenterprises.com), and the recom-
antiviral therapy in controlling any potential damage that might mended dose is 125 units/10 kg of body weight, up to a maximum
arise from continued VZV replication postnatally. Nevertheless, if of 625 units. The minimum dose is 125 units. VZIG prevents vari-
the infant has or develops clinical signs of active infection, it is cella or reduces the severity of infection in most cases; however,
prudent to treat. The acyclovir dose should be that established as patients should be monitored for breakthrough infection.46
safe for treating neonatal herpes simplex infections, which is Passive antibody prophylaxis is intended to protect the
15 mg/kg per dose given intravenously every 8 h. The duration of susceptible pregnant woman from severe varicella. Whether the
therapy must be determined from evidence of control of active VZV fetus is protected from transplacental VZV transmission is not
replication, either by cessation of lesion formation or by using known, but it is reasonable to assume that preventing maternal
laboratory tests, e.g. VZV PCR or viral culture. viremia will reduce risk. In one prospective study, maternal
Herpes zoster in infants who acquired VZV in utero has been breakthrough infection occurred despite VZIG prophylaxis in 97
self-limited or responsive to treatment with acyclovir. If the zoster cases. None of the infants had CVS, but this sample size is not
is extensive and painful, it is usually prudent to treat with intra- sufficient to conclude that the fetus was protected, as the incidence
venous acyclovir at least initially, followed by oral acyclovir. No of CVS is <1%.21 Acyclovir prophylaxis is not recommended because
antiviral regimen has been evaluated but treatment for seven to 10 of the lack of information about its safety or efficacy compared to
days can be expected to be sufficient because these infants are not VZV immune globulin. Varicella vaccine can be given for post-
otherwise immunocompromised. Serious complications have been exposure prophylaxis in non-pregnant individuals but the live
reported in only one case of a four-month-old infant of a mother attenuated vaccine is contraindicated in pregnancy.
who had had varicella at 17 weeks’ gestation. The infant was born As more women of childbearing age have vaccine-induced
full term without complications, developed herpes zoster at three immunity to VZV, the assessment of immune status will shift to
months, then had generalized seizures, with VZV DNA in cerebro- documenting that the exposed pregnant woman has received two
spinal fluid by PCR and findings consistent with central nervous doses of live attenuated varicella vaccine. If this history is docu-
system infection.44 This infant received intravenous acyclovir for 10 mented, serologic testing is not recommended. Commercially
days, followed by oral acyclovir for three weeks; VZV PCR of the CSF available serologic assays for VZV IgG antibodies have even higher
became negative but whether resolution of the infection was false negative rates in vaccine recipients than in naturally immune
accelerated or the outcome was improved cannot be judged from individuals. The correlation with protection against exposure is
a single case report. with the number of vaccine doses received. As the vaccine era
progresses, it will be important to confirm immune status because
7. Management of pregnant women exposed to VZV some women may have not been vaccinated and are less likely to
have natural infection as varicella epidemics are controlled.
Passive antibody prophylaxis with immune globulin prepara-
tions that contain VZV IgG antibodies is indicated if a pregnant 8. Perinatal varicella
woman is susceptible to VZV, the exposure was a close contact and
prophylaxis can be administered within 72–96 h after the expo- Varicella of the newborn is a life-threatening illness that may
sure. Pregnant women who have a history of varicella are not occur when maternal disease occurs within an approximate seven-
considered at risk. Symptomatic re-infection with VZV is quite rare day window that spans from five days before delivery until two
and pre-existing maternal immunity would be expected to protect days after delivery. These infants should be given VZIG if available
the fetus. A clinical history of varicella is considered sufficient or IVIG at birth or as soon as the maternal symptoms appear in the
evidence of immunity. A recent study assessing the validity of two days after delivery.47 IVIG contains high titers of VZV-specific
a self-reported history of varicella showed that it is a strong IgG, and VZV antibody titers have been shown to be equivalent after
predictor that the pregnant woman will have VZV IgG antibodies treatment with either VZIG or IVIG.48 Before the use of VZIG, an
(>98%).45 However, since 95% of adult women have VZV IgG estimated 17–30% of the newborns contracted severe varicella
antibodies, this correlation is not unexpected. Only a minority of because of the lack of maternal VZV IgG antibody transfer to protect
women who had a negative or uncertain varicella disease history the neonate and the relative immaturity of the neonatal immune
were actually seronegative, which is also consistent with the system.49 Before VZV immunoglobulin was available, the risk of
epidemiology of VZV infection in women of childbearing age from death among neonates born to mothers with the onset of rash up to
C.K. Smith, A.M. Arvin / Seminars in Fetal & Neonatal Medicine 14 (2009) 209–217 215

four days before delivery was 31%.50 The rate decreased to 7% when Infants who are born more than a week after the first appear-
the use of VZIG was introduced and neonatal intensive care ance of the maternal rash are not at high risk of severe disease. They
improved.51 will likely have lesions at birth or within the first several days of life,
Infants born to mothers with varicella within this high risk but they are protected from severe disease secondary to maternal
period are usually initially well-appearing. VZV presents with the VZV antibodies having time to traverse the placenta. Again, despite
classical skin lesions, but can disseminate with pneumonia, hepa- the lower risk, if there are signs and symptoms of active VZV
titis, encephalitis and severe coagulopathy resulting from liver disease, treating with an antiviral is recommended. When the
failure and thrombocytopenia (Fig. 5).52 Despite the administration history of maternal varicella is in the few weeks preceding delivery,
of VZIG, infants exposed to maternal varicella in this high risk the infants may be asymptomatic or may have cutaneous lesions at
period should be monitored very closely and intravenous acyclovir birth or developing shortly thereafter, but are at low risk of
should be given if there are any signs of illness. Acyclovir prophy- dissemination of varicella disease or complications; nevertheless
laxis is not recommended because IVIG is readily available even if treatment is prudent.
VZIG is not. A small uncontrolled trial of acyclovir and immuno-
globulin in combination compared to immunoglobulin alone 9. Varicella vaccination
showed no clinical varicella in any of 10 infants given treatment
with the combination versus two of four infants given immuno- The early childhood varicella immunization program in the USA
globulin alone.53 The dosing of acyclovir in this trial was 5 mg/kg was recommended based on an analysis of the annual morbidity
intravenously every 8 h for five days. The sample size is too small to and mortality associated with varicella epidemics, most of which
determine whether the combination improved outcomes, and was associated with secondary Streptococcus pyogenes and Staph-
a standard prophylaxis dosage or duration has not been deter- ylococcal aureus infections or neurologic complications, including
mined. Careful monitoring and early treatment of breakthrough encephalitis and cerebellar ataxia in otherwise healthy individuals.
varicella can be expected to be highly effective. Treatment of infants Universal vaccination was implemented because of the almost
who develop breakthrough varicella with intravenous acyclovir universal prevalence of varicella disease, causing an estimated four
should be continued for 48 h after the last new lesions have million cases each year, and because those at risk for complications
appeared; a duration of seven days is usually sufficient. There is no could not be predicted. The program of universal vaccination
indication for additional therapy with oral acyclovir, but the infant against varicella has decreased the varicella death rate in the USA
should be monitored for new lesions and re-treated with intrave- across all age groups because of the impact on the annual
nous acyclovir if necessary. epidemics.54

Fig. 5. Newborn with varicella acquired from the mother in the perinatal period.
216 C.K. Smith, A.M. Arvin / Seminars in Fetal & Neonatal Medicine 14 (2009) 209–217

When possible, maternal VZV immune status should be deter- 10. Future research directions
mined before pregnancy by reviewing the varicella history or
documenting vaccination with two doses of varicella vaccine. If Research to evaluate the persistence of VZV immunity in vaccine
both are negative, then VZV IgG antibody testing should be done recipients and to document the changing epidemiology of varicella
because the vaccine can be offered if the woman is not immune. will be important now that the universal varicella immunization
Women of childbearing age without a history of chickenpox or program has been implemented. Such research has already led to
vaccination and who are confirmed seronegative should be coun- the revision of the early childhood vaccine schedule to provide two
seled about the risks of varicella during pregnancy and receive two doses. Further modification of the current varicella vaccine sched-
doses of vaccine, given four to eight weeks apart. If more than eight ules could occur depending on observations about the rates of
weeks elapse after the first dose, the second dose can still be given breakthrough varicella. Basic research about VZV remains a high
without restarting the schedule.55 Vaccination can be given in the priority in order to better understand the molecular mechanisms of
immediate postpartum period, and breastfeeding is not a contra- pathogenesis and host control of this ubiquitous human pathogen.
indication for immunization.56 The US Advisory Committee on It is now possible to manipulate the VZV genome to reduce its
Immunization Practices recommends that postpartum women capacity to infect human T-cells and sensory ganglia.13 This
without evidence of immunity be given the first dose of vaccine research is likely to reveal strategies for creating second generation
before discharge from the hospital, and the second dose of vaccine genetically engineered live attenuated varicella vaccines that would
at the follow-up postpartum visit (six to eight weeks after be safe in immunocompromised patients and for making higher
delivery).55 As the varicella vaccine was licensed in 1995, the cohort potency vaccines that could reduce the number of doses required,
of infants immunized at 12–15 months is approaching childbearing without increasing adverse events and risk of spread of the vaccine
age. It should be emphasized that each of these women should be virus from the site of inoculation. A better understanding of the
screened for a history of two doses of vaccine to reduce the risk of VZV proteins and the host response to these components of the
varicella during pregnancy. While waning immunity following virus may also lead to the design of protein/adjuvant vaccines that
vaccine has been suggested from some epidemiological data, would be valuable for high risk patients.
infection in vaccinated children may be due to failure to induce
immunity associated with a single-dose vaccine regimen; a two-
dose regimen has been introduced in response to these epidemi- References
ologic observations.57 The active surveillance sites for varicella are
designed to identify changes in epidemiology that could indicate 1. Ross AH. Modification of chicken pox in family contacts by administration of
gamma globulin. N Engl J Med 1962;267:369–76.
the need for booster doses of the vaccine. Changes in varicella
2. Kelley PW, Petruccelli BP, Stehr-Green P, Erickson RL, Mason CJ. The suscep-
epidemiology may occur because the continued circulation of VZV tibility of young adult Americans to vaccine-preventable infections. A national
in the community may have provided a natural boosting of vaccine- serosurvey of US Army recruits. J Am Med Assoc 1991;266:2724–9.
induced immunity. As vaccine coverage rates improve and varicella *3. Seward JF, Marin M, Vazquez M. Varicella vaccine effectiveness in the US
vaccination program: a review. J Infect Dis 2008;197(Suppl. 2):S82–9.
outbreaks become increasingly rare, it may be necessary to give an 4. Harger JH, Ernest JM, Thurnau GR, et al. Risk factors and outcome of varicella-
adolescent/young adult dose of the vaccine as follow-up to the zoster virus pneumonia in pregnant women. J Infect Dis 2002;185:422–7.
universal childhood vaccination program. *5. Seward JF, Jumaan A. VZV: persistence in the population. In: Arvin AM, editor.
Human herpesviruses: biology, therapy, and immunoprophylaxis. Cambridge:
Because the effect on fetal development is not known, the Cambridge University Press; 2007. p. 713–721.
varicella vaccine is contraindicated in pregnancy. The Advisory 6. Guris D, Jumaan AO, Mascola L, et al. Changing varicella epidemiology in active
Committee on Immunization Practices recommends that preg- surveillance sites – United States, 1995–2005. J Infect Dis 2008;197(Suppl. 2):
S71–5.
nancy be avoided for one month after vaccination. However, there 7. Marin M, Watson TL, Chaves SS, et al. Varicella among adults: data from an
are cases of inadvertent vaccination of pregnant women, or vacci- active surveillance project, 1995–2005. J Infect Dis 2008;197(Suppl. 2):
nation within the few months prior to pregnancy. This is a low risk S94–100.
8. Kilgore PE, Kruszon-Moran D, Seward JF, et al. Varicella in Americans from
exposure as the vaccine strain of virus is attenuated and would be NHANES III: implications for control through routine immunization. J Med
less virulent than wild type varicella. A registry has followed Virol 2003;70(Suppl. 1):S111–8.
a cohort of 944 women and their infants since 1995 who received 9. Gershon AA, Raker R, Steinberg S, Topf-Olstein B, Drusin LM. Antibody to
Varicella-Zoster virus in parturient women and their offspring during the first
vaccine anytime during the pregnancy or three months before
year of life. Pediatrics 1976;58:692–6.
conception, and there were no infants with signs consistent with 10. Plans P, Costa J, Espunes J, Plasencia A, Salleras L. Prevalence of varicella-zoster
congenital varicella syndrome or birth defects suggesting varicella antibodies in pregnant women in Catalonia (Spain). Rationale for varicella
infection.58 vaccination of women of childbearing age. Br J Obstet Gynaecol 2007;114:
1122–7.
11. Sever J, White LR. Intrauterine viral infections. Annu Rev Med 1968;19:471–86.
9.1. Herpes zoster in pregnancy 12. Grose C. Variation on a theme by Fenner: the pathogenesis of chickenpox.
Pediatrics 1981;68:735–7.
*13. Ku CC, Besser J, Abendroth A, Grose C, Arvin AM. Varicella-Zoster virus
Herpes zoster is not unusual in pregnancy, especially in later pathogenesis and immunobiology: new concepts emerging from investiga-
gestation, probably because of some suppression of maternal tions with the SCIDhu mouse model. J Virol 2005;79:2651–8.
immune responses associated with pregnancy. However, among 14. Paryani SG, Arvin AM. Intrauterine infection with varicella-zoster virus after
maternal varicella. N Engl J Med 1986;314:1542–6.
366 cases of herpes zoster in pregnancy in which outcomes were *15. Schutte TJ, Rogers LC, Copas PR. Varicella pneumonia complicating pregnancy:
followed prospectively, no defects consistent with CVS and no cases a report of seven cases. Infect Dis Obstet Gynecol 1996;4:338–46.
of herpes zoster were reported in the infants.21 There are a few case *16. Tan MP, Koren G. Chickenpox in pregnancy: revisited. Reprod Toxicol
2006;21:410–20.
reports of infants with abnormalities after maternal herpes zoster
*17. Mouly F, Mirlesse V, Meritet JF, et al. Prenatal diagnosis of fetal varicella-zoster
but the findings were non-specific and none had signs of CVS. virus infection with polymerase chain reaction of amniotic fluid in 107 cases.
Infants whose mothers develop herpes zoster in the peripartum Am J Obstet Gynecol 1997;177:894–8.
18. Gibson CS, Goldwater PN, MacLennan AH, et al. Fetal exposure to herpesvi-
period are not at risk for serious illness from perinatal varicella.
ruses may be associated with pregnancy-induced hypertensive disorders and
However, a dermatomal vesicular rash in the lumbosacral area preterm birth in a Caucasian population. Br J Obstet Gynaecol 2008;115:
raises the possibility of herpes simplex virus (HSV) infection, as this 492–500.
(usually HSV-2) causes as many as 15% of the ‘zoster’ cases in this 19. Michie CA, Acolet D, Charlton R, et al. Varicella-zoster contracted in the second
trimester of pregnancy. Pediatr Infect Dis J 1992;11:1050–3.
distribution.52 The diagnosis should be established by laboratory 20. LaForet EG, LJC. Multiple congenital defects following maternal varicella. N
testing of the lesions for VZV and HSV. Engl J Med 1947;236:534–7.
C.K. Smith, A.M. Arvin / Seminars in Fetal & Neonatal Medicine 14 (2009) 209–217 217

*21. Enders G, Miller E, Cradock-Watson J, Bolley I, Ridehalgh M. Consequences of 40. Frenkel LM, Brown ZA, Bryson YJ, et al. Pharmacokinetics of acyclovir in
varicella and herpes zoster in pregnancy: prospective study of 1739 cases. the term human pregnancy and neonate. Am J Obstet Gynecol
Lancet 1994;343(8912):1548–51. 1991;164:569–76.
22. Gershon AA. Chickenox, measles, and mumps. In: Remington J, Klein JO, 41. Stone KM, Reiff-Eldridge R, White AD, et al. Pregnancy outcomes following
Wilson CB, Baker CJ, editors. Infectious diseases of the fetus and newborn infant. systemic prenatal acyclovir exposure: conclusions from the international
6th ed. Philadelphia: Elsevier Saunders; 2006. p. 693. acyclovir pregnancy registry, 1984–1999. Birth Defects Res A Clin Mol Teratol
23. Birthistle K, Carrington D. Fetal varicella syndrome – a reappraisal of the 2004;70:201–7.
literature. A review prepared for the UK Advisory Group on Chickenpox 42. American Academy of Pediatrics Committee on Infectious Diseases. The use of
on behalf of the British Society for the Study of Infection. J Infect oral acyclovir in otherwise healthy children with varicella. Pediatrics
1998;36(Suppl. 1):25–9. 1993;91:674–6.
24. Ussery XT, Annunziato P, Gershon AA, et al. Congenital varicella-zoster virus 43. Gardella C, Brown ZA. Managing varicella zoster infection in pregnancy. Cleve
infection and Barrett’s esophagus. J Infect Dis 1998;178:539–43. Clin J Med 2007;74:290–6.
25. Nikkels AF, Delbecque K, Pierard GE, et al. Distribution of varicella-zoster virus 44. Sauerbrei A, Pawlak J, Luger C, Wutzler P. Intracerebral varicella-zoster virus
DNA and gene products in tissues of a first-trimester varicella-infected fetus. reactivation in congenital varicella syndrome. Dev Med Child Neurol
J Infect Dis 2005;191:540–5. 2003;45:837–40.
26. Grose C. Varicella zoster virus infections: chickenpox, shingles, and varicella 45. Watson B, Civen R, Reynolds M, et al. Validity of self-reported varicella disease
vaccine. In: Glaser R, Jones JF, editors. Herpesvirus infections. New York: M. history in pregnant women attending prenatal clinics. Public Health Rep
Dekker; 1994. p. 142–152. 2007;122:499–506.
27. Higa K, Dan K, Manabe H. Varicella-zoster virus infections during pregnancy: 46. Brunell PA, Ross A, Miller LH, Kuo B. Prevention of varicella by zoster immune
hypothesis concerning the mechanisms of congenital malformations. Obstet globulin. N Engl J Med 1969;280:1191–4.
Gynecol 1987;69:214–22. 47. Chapman SJ. Varicella in pregnancy. Semin Perinatol 1998;22:339–46.
28. Kotchmar Jr GS, Grose C, Brunell PA. Complete spectrum of the varicella congenital 48. Paryani SG, Arvin AM, Koropchak CM, et al. Comparison of varicella zoster
defects syndrome in 5-year-old child. Pediatr Infect Dis 1984;3:142–5. antibody titers in patients given intravenous immune serum globulin or
29. Schulze A, Dietzsch HJ. The natural history of varicella embryopathy: a 25-year varicella zoster immune globulin. J Pediatr 1984;105:200–5.
follow-up. J Pediatr 2000;137:871–4. 49. Prober CG, Gershon AA, Grose C, McCracken Jr GH, Nelson JD. Consensus:
30. Sauerbrei A, Wutzler P. Herpes simplex and varicella-zoster virus infections varicella-zoster infections in pregnancy and the perinatal period. Pediatr Infect
during pregnancy: current concepts of prevention, diagnosis and therapy. Dis J 1990;9:865–9.
Part 2: Varicella-zoster virus infections. Med Microbiol Immunol 2007;196: 50. Meyers JD. Congenital varicella in term infants: risk reconsidered. J Infect Dis
95–102. 1974;129:215–7.
31. Alkalay AL, Pomerance JJ, Rimoin DL. Fetal varicella syndrome. J Pediatr 51. Miller E, Cradock-Watson JE, Ridehalgh MK. Outcome in newborn babies given
1987;111:320–3. anti-varicella-zoster immunoglobulin after perinatal maternal infection with
32. Dworsky M, Whitley R, Alford C. Herpes zoster in early infancy. Am J Dis Child varicella-zoster virus. Lancet 1989;2(8659):371–3.
1980;134:618–9. 52. Prober CG, Arvin AM. Perinatal viral infections. Eur J Clin Microbiol 1987;6:245–61.
33. Brunell PA, Kotchmar Jr GS. Zoster in infancy: failure to maintain virus latency 53. Huang YC, Lin TY, Lin YJ, Lien RI, Chou YH. Prophylaxis of intravenous
following intrauterine infection. J Pediatr 1981;98:71–3. immunoglobulin and acyclovir in perinatal varicella. Eur J Pediatr
34. Hope-Simpson RE. The nature of herpes zoster: a long-term study and a new 2001;160:91–4.
hypothesis. Proc R Soc Med 1965;58:9–20. *54. Nguyen HQ, Jumaan AO, Seward JF. Decline in mortality due to varicella after
35. Arvin AM, Koropchak CM, Wittek AE. Immunologic evidence of reinfection implementation of varicella vaccination in the United States. N Engl J Med
with varicella-zoster virus. J Infect Dis 1983;148:200–5. 2005;352:450–8.
36. Gershon AA, Steinberg SP, Borkowsky W, Lennette D, Lennette E. IgM to *55. Marin M, Guris D, Chaves SS, Schmid S, Seward JF. Prevention of varicella:
varicella-zoster virus: demonstration in patients with and without clinical recommendations of the Advisory Committee on Immunization Practices
zoster. Pediatr Infect Dis 1982;1:164–7. (ACIP). MMWR Recomm Rep 2007;56(RR-4):1–40.
*37. Meyberg-Solomayer GC, Fehm T, Muller-Hansen I, et al. Prenatal ultrasound 56. Bohlke K, Galil K, Jackson LA, et al. Postpartum varicella vaccination: is the
diagnosis, follow-up, and outcome of congenital varicella syndrome. Fetal vaccine virus excreted in breast milk? Obstet Gynecol 2003;102(5 Pt 1):970–7.
Diagn Ther 2006;21:296–301. 57. Gershon AA, Katz SL. Perspective on live varicella vaccine. J Infect Dis
38. Boumahni B, Kauffmann E, Laffitte A, Randrianaivo H, Fourmaintraux A. 2008;197(Suppl. 2):S242–5.
Congenital varicella: limits of prenatal diagnosis. Arch Pediatr 2005;12:1361–3. 58. Wilson E, Goss MA, Marin M, et al. Varicella vaccine exposure during
*39. Verstraelen H, Vanzieleghem B, Defoort P, Vanhaesebrouck P, pregnancy: data from 10 years of the pregnancy registry. J Infect Dis
Temmerman M. Prenatal ultrasound and magnetic resonance imaging in 2008;197(Suppl. 2):S178–84.
fetal varicella syndrome: correlation with pathology findings. Prenat Diagn
2003;23:705–9. * The most important references are indicated with an asterisks.

Vous aimerez peut-être aussi